TERT promoter mutations in primary liver tumors

TERT promoter mutations in primary liver tumors

+Model CLINRE-798; No. of Pages 6 ARTICLE IN PRESS Clinics and Research in Hepatology and Gastroenterology (2015) xxx, xxx—xxx Available online at ...

728KB Sizes 7 Downloads 86 Views

+Model CLINRE-798; No. of Pages 6

ARTICLE IN PRESS

Clinics and Research in Hepatology and Gastroenterology (2015) xxx, xxx—xxx

Available online at

ScienceDirect www.sciencedirect.com

MINI REVIEW

TERT promoter mutations in primary liver tumors Jean-Charles Nault a,b,c,d,e, Jessica Zucman-Rossi a,b,c,d,f,∗ a

Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, Équipe Labellisée Ligue Contre le Cancer, 75010 Paris, France b Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, 75006 Paris, France c Université Paris 13, Sorbonne Paris Cité, UFR SMBH, 93000 Bobigny, France d Université Paris Diderot, 75013 Paris, France e AP—HP, Hôpitaux Universitaires Paris — Seine-Saint-Denis, Site Jean-Verdier, Pôle d’Activité Cancérologique Spécialisée, Service d’Hépatologie, 93143 Bondy, France f Assistance publique—Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, 75015 Paris, France

Summary Next-generation sequencing has drawn the genetic landscape of hepatocellular carcinoma and several signaling pathways are altered at the DNA level in tumors: Wnt/␤catenin, cell cycle regulator, epigenetic modifier, histone methyltransferase, oxidative stress, ras/raf/map kinase and akt/mtor pathways. Hepatocarcinogenesis is a multistep process starting with the exposure to different risk factors, followed by the development of a chronic liver disease and cirrhosis precede in the vast majority of the cases the development of HCC. Several lines of evidence have underlined the pivotal role of telomere maintenance in both cirrhosis and HCC pathogenesis. TERT promoter mutations were identified as the most frequent genetic alterations in hepatocellular carcinoma with an overall frequency around 60%. Moreover, in cirrhosis, TERT promoter mutationsare observed at the early steps of hepatocarcinogenesis since they are recurrently identified in low-grade and high-grade dysplastic nodules. In contrast, acquisition of genomic diversity through mutations of classical oncogenes and tumor suppressor genes (TP53, CTNNB1, ARID1A. . .) occurred only in progressed HCC. In normal liver, a subset of HCC can derived from the malignant transformation of hepatocellular adenoma (HCA). In HCA, CTNNB1 mutations predispose to transformation of HCA in HCC and TERT promoter mutations are required in most of the cases as a second hit for a full malignant transformation. All these findings have refined our knowledge of HCC pathogenesis and have pointed telomerase as a target for tailored therapy in the future. © 2015 Elsevier Masson SAS. All rights reserved.

∗ Corresponding author at: Inserm U 1162, Génomique fonctionnelle des tumeurs solides, 27, rue Juliette-Dodu, 75010 Paris, France. Tel.: +33 1 53 72 51 66; fax: +33 1 53 72 51 92. E-mail address: [email protected] (J. Zucman-Rossi).

http://dx.doi.org/10.1016/j.clinre.2015.07.006 2210-7401/© 2015 Elsevier Masson SAS. All rights reserved.

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006

+Model CLINRE-798; No. of Pages 6

ARTICLE IN PRESS

2

J.-C. Nault, J. Zucman-Rossi

Hepatocellular carcinoma (HCC) has emerged as an important health problem worldwide. The main etiologies of HCC are hepatitis B virus (HBV), hepatitis C virus (HCV), alcohol consumption, obesity (non-alcoholic steatohepatitis [NASH]) and rare metabolic disorder like hemochromatosis [1,2]. In area of low incidence of HCC-like in western countries, HCC develop mainly on cirrhosis related to HCV, high alcohol intake and NASH [1,2]. In Asia, the high incidence of HCC follows the huge number of patients with chronic HBV infection. During HBV infection, HCC could also arise in otherwise normal liver or in liver with limited fibrosis [3]. Moreover, a small percentage of HCC developed on normal liver and without chronic viral infection in both western and eastern countries. In clinical care, a limited number of patients with HCC are eligible to curative treatment (liver transplantation, radiofrequency ablation and liver resection) [1,4]. Most of the times, patients only benefit of palliative treatment (chemoembolization or sorafenib a tyrosine kinase inhibitor) or best supportive care [2,5]. From a basic point of view, HCC have a complex pathogenesis characterized by genomic diversity [6—8]. As other cancer, accumulation of somatic molecular alterations in malignant hepatocytes draws a unique profile in each HCC [9]. In this review, we will describe how the identification of telomerase reverse transcriptase (TERT) promoter mutations as the most frequent somatic genetics alterations in HCC has underlined the major role of telomere and telomerase in liver carcinogenesis.

The genetic landscape of hepatocellular carcinoma Recent technological breakthroughs have accelerated the exploration of the tumor genome and have increased our knowledge of HCC pathogenesis [9,10]. Whole exome (exploring the whole coding region of the genome), whole genome (exploring both coding and non-coding region) or RNA sequencing (exploring the whole-transcriptome sequence) allow to draw quickly the genetic portrait of a tumor [10]. Each tumor is a unique combination of somatic mutations in genes driver of the mechanism of tumorigenesis and in passenger genes. Driver genes mutated in HCC belong to several key signaling pathway of oncogenesis [11] and the genetic landscape of HCC includes recurrent somatic mutations in CTNNB1 and AXIN1 (WNT/␤-catenin pathway), TP53, RB1, CDKN2A inactivation (cell cycle gene), ARID1A and ARID2 mutations (chromatin remodeling gene), MLL2, MLL3 and MLL4 mutations (histone methyltransferase gene), NFE2L2 and KEAP1 mutations (stress oxidative pathway), RPS6KA3, PIK3CA, TSC1 and TSC2 mutations (AKT/MTOR and RAS/RAF/MAP kinase pathways) and VEGFA and CCND1/FGF19 amplification [12,13].

Telomere and telomerase in cirrhosis pathogenesis and liver carcinogenesis Telomeres are short repeated DNA sequences (TTAGGG) situated at the extremity of each chromosome [14,15]. Due to the replication end problem, telomere shortened at each round of cell division and when a critical point is

attained DNA damage protein are activated and lead to cell senescence [15,16]. The telomerase complex function is to maintain telomere length and to avoid senescence by adding repeat sequence at the extremity of the chromosome [17]. The telomerase complex is composed of the core catalytic enzyme named telomerase reverse transcriptase (TERT) and of the RNA template named TERC. Cirrhosis is characterized by senescent hepatocytes with short telomere and absence of telomerase activity [18]. Several lines of evidence have shown that telomere deficient mice have a high incidence of cirrhosis occurrence when the liver is exposed to chronic liver injury [19]. In contrast, in the same model, telomerase reactivation is required to promote full malignant transformation on a cirrhotic background [20]. The limiting factor of the complex is TERT and reactivation of telomerase is a widely observed phenomena in cancer [21,22]. It avoids senescence induced by telomere shortening during the uncontrolled proliferation of cancer cells [23—26]. In the same line, several studies have reported a frequent reactivation of TERT in HCC compared to normal liver or cirrhosis [26,27]. However, the mechanism leading to telomerase reactivation was poorly understood until recently.

TERT promoter mutations in hepatocellular carcinoma Activating mutations of the TERT promoter that increase TERT expression have been recently described in several types of tumors including melanoma, glioblastoma, hepatocellular carcinoma, bladder cancer or anaplastic thyroid cancer [28—31]. These mutations are substitutions located in two hot spots situated 124 and 146 base paired before the ATG start [28,29]. They created a new consensus binding sequence (CCGGAA or CCGGAT) that could bind ETS/TCF transcription factor and lead to an increase promoter activity. In HCC, we identified TERT promoter mutations in 59% of the tumors (Table 1) [32]. Other teams confirmed these results and the frequency of TERT promoter mutations is more frequent in western countries (54 to 60%) whereas 29 to 31% of HCC are mutated in eastern countries [12,13,30,33—36]. Almost all TERT promoter mutations in HCC (95%) occurred at the first hot spot -124G>A [32]. These mutations were not identified in the seminal whole exome studies of the HCC because this region of the genome was not included in the whole exome data. Consequently, TERT promoter mutations are the most frequent genetic alterations observed in HCC and these mutations are associated with an increase TERT expression. Interestingly, TERT promoter mutations were more frequent in old patients and were also significantly associated with activating mutations of ␤catenin suggesting cooperation between these two pathways [13,32]. Moreover, insertion of HBV in the TERT gene, most frequently in the promoter, has been described as an alternative mechanism leading to telomerase expression [37,38]. TERT amplifications, is another mechanism of telomerase reactivation, they have been also described by two different teams in 5 to 6% of HCC [12,13]. Strikingly, TERT promoter mutations, TERT amplification and HBV insertion in the TERT promoter were exclusive together. All these data have underlined the pivotal role of telomere maintenance in liver carcinogenesis.

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006

% mut.

Hepatocellular carcinoma

305

59

94

Hepatocellular carcinoma Hepatocellular carcinoma

61 195

44 29

Hepatocellular carcinoma Hepatocellular carcinoma

78 469

Hepatocellular carcinoma Hepatocellular carcinoma Liver high-grade dysplastic nodule Liver low-grade dysplastic nodule Hepatocellular adenoma Hepatocellular adenoma Borderline HCC/HCA and HCC on HCA Hepatoblastoma Hepatocholangiocarcinoma Intrahepatic cholangiocarcinoma Gallbladder carcinoma Gallbladder carcinoma

44 35 16 32 223 15 27 15 30 52 10 154

124 hot spot (%)

146 hot spot (%)

Association

Article

6

Old age, CTNNB1 mutation, low tumor size, low AFP and low frequency in HBV infection

Nault et al., 2013 [32]

95

5

47 54

100 93

5

Old age, hepatitis C and low frequency in HBV infection High-grade of differentiation CTNNB1 mutation, low frequency in HBV infection

34 31 19 6 0 0 44 0 15 0 0 9

66

34

87 87

13 13

Hepatitis C

43

57

ARTICLE IN PRESS

Number of cases

+Model

Type of tumor

CLINRE-798; No. of Pages 6

TERT promoter mutations in liver tumors.

TERT promoter and liver tumors

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006

Table 1

Killela et al., 2013 [30] Chen et al., 2014 [33] Quaas et al., 2014 [34] Totoki et al., 2015 [13] Cevik et al., 2015 [35] Huang et al., 2015 [36] Nault et al., 2014 [43] Nault et al., 2014 [43] Pilati et al., 2014 [52] Quaas et al., 2014 [34] Pilati et al., 2014 [52] Eichenmuller et al., 2014 [53] Fujimoto et al., 2015 [55] Quaas et al., 2014 [34] Killela et al., 2013 [30] Qu et al., 2014 [54]

TERT: telomerase reverse transcriptase; % mut.: percentage of mutations; HCC: hepatocellular carcinoma; HCA: hepatocellular adenoma; HBV: hepatitis B virus.

3

+Model CLINRE-798; No. of Pages 6

ARTICLE IN PRESS

4

J.-C. Nault, J. Zucman-Rossi

TERT promoter mutations in premalignant lesions on cirrhosis On cirrhosis, liver carcinogenesis is a multistep process with the sequential development of low-grade dysplastic nodules, high-grade dysplastic macronodules, early HCC and small and progress HCC [39,40]. Usually, TERT is not expressed in mature hepatocytes and in cirrhotic liver however it is re-expressed in some cirrhotic premalignant lesions (as high-grade dysplastic nodules) in addition of the wellknown re-expression of TERT in HCC [27,41,42]. Recently, we have reported somatic recurrent mutations in the TERT promoter in 6% of low-grade dysplastic nodules and 19% of high-grade dysplastic nodules (Table 1) [32,43]. Other mutations in cancer genes like TP53, CTNNB1, ARID1A, ARID2, RPS6KA3 occurred only in small and progressed HCC [43]. It is the first report of recurrent genetic alterations in cirrhotic premalignant lesions and TERT promoter mutations could be considered as a gatekeeper gene like APC in the colorectal adenoma-carcinoma sequence [44]. These data underlined that reactivation of telomerase trough mutation of TERT promoter is required at the earlier step in order to bypass replicative senescence of cirrhotic hepatocytes. In contrast, acquisition of genomic diversity appears to be a late event in liver carcinogenesis on cirrhosis [9].

TERT promoter mutations in hepatocellular adenomas A subset of HCC developed on normal liver derived from a malignant transformation of HCA. HCA are rare liver tumors composed of benign proliferation of hepatocytes [45]. The description of the genetic landscape of HCA is a paradigm of genotype/phenotype classification in liver tumors. Four groups of HCA have been identified [46,47]. The first one is composed of HCA with bi-allelic inactivating mutations of HNF1A and is characterized by steatosis at pathological reviewing [48]. The second group is HCA with activating mutations of CTNNB1 (coding for ␤-catenin). Male are overrepresented in this subgroup and cholestasis and pseudoglandular formation are frequently observed. CTNNB1 mutations were associated with an increased risk of malignant transformation [47]. The third group is composed of inflammatory HCA characterized by inflammatory infiltrate, sinusoidal dilatation and dystrophic arteries. In this subgroups, recurrent somatic activating mutations of IL6ST (coding for gp130), STAT3, FRK, JAK1 and GNAS lead to the JAK/STAT pathway activation [49—51]. Interestingly, IL6ST and CTNNB1 mutations could be associated whereas HNF1A are exclusive from CTNNB1 and the inflammatory mutations activating the JAK/STAT pathway. Finally, the last group is composed of the remaining unclassified HCA. In a series of 250 ‘‘classical’’ HCA without any signs of malignant transformation, we did not identify any TERT promoter mutations and these results were confirmed by an independent group [32,34,52]. Recently, in the subset of HCA with malignant transformation we identified TERT promoter mutations in 44% (Table 1) [32,52]. Moreover, TERT promoter mutations were associated with mutations of CTNNB1, suggesting again a cooperation between telomerase maintenance and

Wnt/␤-catenin pathway [32,52]. To summarize, CTNNB1 mutations with or without IL6ST mutations lead to HCA initiation and TERT promoter mutations is required at the last step of malignant transformation into HCC. As HCA arise on normal liver with hepatocytes having their full renewal ability, reactivation of telomerase is required later in the process to bypass oncogene-induced senescence and increase the proliferative ability of tumor hepatocytes [9].

TERT promoter mutations in other primary liver cancers As in other pediatric cancers, TERT promoter mutations were not identified in hepatoblastoma (Table 1) [53]. Quaas et al. have analyzed 52 intrahepatic cholangiocarcinoma and found no mutations of the TERT promoter [34]. TERT promoter mutations were observed in 0 to 9% of gallbladder cancer [30,54]. In contrast, among 30 hepatocholangiocarcinoma, 15% of the tumors were mutated in the TERT promoter [55]. Consequently, TERT promoter mutations were not recurrently involved in the pathogenesis of cholangiocarcinoma and of hepatoblastoma and were observed only in a subset of hepatocholangiocarcinoma. It suggests that acquisition of TERT promoter mutations during carcinogenesis is dependent of the cell of origins of the tumor in the liver.

Conclusion Reactivation of telomerase is a major event of malignant transformation of hepatocytes in either normal or cirrhotic liver. TERT promoter mutations, the leading somatic genetic defect in HCC, are one of the main mechanisms leading to telomerase re-expression. TERT amplification and insertion of HBV virus in the TERT promoter are alternative mechanism explaining telomerase reactivation. However, 5% of HCC lack telomerase expression; in these tumors an alternative mechanisms of telomere lengthening could exist, its mechanism remains to be elucidate [56]. Moreover, telomerase is a potential therapeutic target and telomerase inhibitors (including small molecule inhibitor, G-quadruplex stabilizer and antisense oligonucleotide) or telomerase vaccine are currently in development to treat cancer harboring high telomerase expression [57,58]. These molecules are promising drugs that should be tested in HCC in the next future.

Disclosure of interest The authors declare that they have no conflicts of interest concerning this article.

Acknowledgements The team is supported by grants from Ligue Nationale Contre le Cancer (équipe Labllisée), Inserm and INCa (PRTK14).

References [1] Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet 2012;379(9822):1245—55 [Epub 2012/02/23].

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006

+Model CLINRE-798; No. of Pages 6

ARTICLE IN PRESS

TERT promoter and liver tumors [2] El-Serag HB. Hepatocellular carcinoma. N Engl J Med 2011;365(12):1118—27 [Epub 2011/10/14]. [3] Nault JC. Pathogenesis of hepatocellular carcinoma according to aetiology. Best Pract Res Clin Gastroenterol 2014;28(5):937—47 [Epub 2014/09/28]. [4] Beaugrand M, N’Kontchou G, Seror O, Ganne N, Trinchet JC. Local/regional and systemic treatments of hepatocellular carcinoma. Semin Liver Dis 2005;25(2):201—11 [Epub 2005/05/27]. [5] Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359(4):378—90 [Epub 2008/07/25]. [6] Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer 2006;6(9):674—87 [Epub 2006/08/25]. [7] Nault JC, Zucman-Rossi J. Genetics of hepatobiliary carcinogenesis. Semin Liver Dis 2011;31(2):173—87 [Epub 2011/05/04]. [8] Pinyol R, Nault JC, Quetglas IM, Zucman-Rossi J, Llovet JM. Molecular profiling of liver tumors: classification and clinical translation for decision making. Semin Liver Dis 2014;34(4):363—75 [Epub 2014/11/05]. [9] Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. The genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology 2015 [In press]. [10] Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz Jr LA, Kinzler KW. Cancer genome landscapes. Science 2013;339(6127):1546—58 [Epub 2013/03/30]. [11] Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet 2012;44(6):694—8 [Epub 2012/05/09]. [12] Schulze K, Imbeaud S, Letouze E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet 2015;47(5): 505—11. [13] Totoki Y, Tatsuno K, Covington KR, Ueda H, Creighton CJ, Kato M, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet 2014;46(12):1267—73 [Epub 2014/11/05]. [14] Gunes C, Rudolph KL. The role of telomeres in stem cells and cancer. Cell 2013;152(3):390—3 [Epub 2013/02/05]. [15] Calado RT, Young NS. Telomere diseases. N Engl J Med 2009;361(24):2353—65 [Epub 2009/12/17]. [16] Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: the senescence connection. Nat Rev Cancer 2008;8(6):450—8. [17] Rudolph KL, Hartmann D, Opitz OG. Telomere dysfunction and DNA damage checkpoints in diseases and cancer of the gastrointestinal tract. Gastroenterology 2009;137(3):754—62 [Epub 2009/07/22]. [18] Satyanarayana A, Manns MP, Rudolph KL. Telomeres and telomerase: a dual role in hepatocarcinogenesis. Hepatology 2004;40(2):276—83 [Epub 2004/09/16]. [19] Rudolph KL, Chang S, Millard M, Schreiber-Agus N, DePinho RA. Inhibition of experimental liver cirrhosis in mice by telomerase gene delivery. Science 2000;287(5456):1253—8 [Epub 2000/02/26]. [20] Farazi PA, Glickman J, Jiang S, Yu A, Rudolph KL, DePinho RA. Differential impact of telomere dysfunction on initiation and progression of hepatocellular carcinoma. Cancer Res 2003;63(16):5021—7 [Epub 2003/08/28]. [21] Kolquist KA, Ellisen LW, Counter CM, Meyerson M, Tan LK, Weinberg RA, et al. Expression of TERT in early premalignant lesions and a subset of cells in normal tissues. Nat Genet 1998;19(2):182—6 [Epub 1998/06/10].

5 [22] Meyerson M, Counter CM, Eaton EN, Ellisen LW, Steiner P, Caddle SD, et al. hEST2, the putative human telomerase catalytic subunit gene, is up-regulated in tumor cells and during immortalization. Cell 1997;90(4):785—95 [Epub 1997/08/22]. [23] Artandi SE, DePinho RA. Telomeres and telomerase in cancer. Carcinogenesis 2010;31(1):9—18 [Epub 2009/11/06]. [24] Hahn WC, Stewart SA, Brooks MW, York SG, Eaton E, Kurachi A, et al. Inhibition of telomerase limits the growth of human cancer cells. Nat Med 1999;5(10):1164—70 [Epub 1999/09/30]. [25] Paradis V, Youssef N, Dargere D, Ba N, Bonvoust F, Deschatrette J, et al. Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum Pathol 2001;32(3):327—32. [26] Nakayama J, Tahara H, Tahara E, Saito M, Ito K, Nakamura H, et al. Telomerase activation by hTRT in human normal fibroblasts and hepatocellular carcinomas. Nat Genet 1998;18(1):65—8 [Epub 1998/01/13]. [27] Kotoula V, Hytiroglou P, Pyrpasopoulou A, Saxena R, Thung SN, Papadimitriou CS. Expression of human telomerase reverse transcriptase in regenerative and precancerous lesions of cirrhotic livers. Liver 2002;22(1):57—69 [Epub 2002/03/22]. [28] 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(6122):959—61 [Epub 2013/01/26]. [29] Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science 2013;339(6122):957—9 [Epub 2013/01/26]. [30] Killela PJ, Reitman ZJ, Jiao Y, Bettegowda C, Agrawal N, Diaz Jr LA, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of selfrenewal. Proc Natl Acad Sci U S A 2013;110(15):6021—6 [Epub 2013/03/27]. [31] 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 [Epub 2013/07/28]. [32] Nault JC, Mallet M, Pilati C, Calderaro J, Bioulac-Sage P, Laurent C, et al. High frequency of telomerase reverse transcriptase promoter somatic mutations in hepatocellular carcinoma and preneoplastic lesions. Nat Commun 2013;4:2218. [33] Chen YL, Jeng YM, Chang CN, Lee HJ, Hsu HC, Lai PL, et al. TERT promoter mutation in resectable hepatocellular carcinomas: a strong association with hepatitis C infection and absence of hepatitis B infection. Int J Surg 2014;12(7):659—65. [34] Quaas A, Oldopp T, Tharun L, Klingenfeld C, Krech T, Sauter G, et al. Frequency of TERT promoter mutations in primary tumors of the liver. Virchows Arch 2014;465(6):673—7. [35] Cevik D, Yildiz G, Ozturk M. Common telomerase reverse transcriptase promoter mutations in hepatocellular carcinomas from different geographical locations. World J Gastroenterol 2015;21(1):311—7. [36] Huang DS, Wang Z, He XJ, Diplas BH, Yang R, Killela PJ, et al. Recurrent TERT promoter mutations identified in a large-scale study of multiple tumour types are associated with increased TERT expression and telomerase activation. Eur J Cancer 2015;51(8):969—76. [37] Paterlini-Brechot P, Saigo K, Murakami Y, Chami M, Gozuacik D, Mugnier C, et al. Hepatitis B virus-related insertional mutagenesis occurs frequently in human liver cancers and recurrently targets human telomerase gene. Oncogene 2003;22(25):3911—6 [Epub 2003/06/19]. [38] Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet 2012;44(7):765—9 [Epub 2012/05/29]. [39] Libbrecht L, Desmet V, Roskams T. Preneoplastic lesions in human hepatocarcinogenesis. Liver Int 2005;25(1):16—27 [Epub 2005/02/09].

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006

+Model CLINRE-798; No. of Pages 6

ARTICLE IN PRESS

6

J.-C. Nault, J. Zucman-Rossi

[40] Di Tommaso L, Sangiovanni A, Borzio M, Park YN, Farinati F, Roncalli M. Advanced precancerous lesions in the liver. Best Pract Res Clin Gastroenterol 2013;27(2):269—84 [Epub 2013/07/03]. [41] Tahara H, Nakanishi T, Kitamoto M, Nakashio R, Shay JW, Tahara E, et al. Telomerase activity in human liver tissues: comparison between chronic liver disease and hepatocellular carcinomas. Cancer Res 1995;55(13):2734—6 [Epub 1995/07/01]. [42] Oh BK, Kim YJ, Park YN, Choi J, Kim KS, Park C. Quantitative assessment of hTERT mRNA expression in dysplastic nodules of HBV-related hepatocarcinogenesis. Am J Gastroenterol 2006;101(4):831—8 [Epub 2006/02/24]. [43] Nault JC, Calderaro J, Di Tommaso L, Balabaud C, Zafrani ES, Bioulac-Sage P, et al. Telomerase reverse transcriptase promoter mutation is an early somatic genetic alteration in the transformation of premalignant nodules in hepatocellular carcinoma on cirrhosis. Hepatology 2014;60(6):1983—92 [Epub 2014/08/16]. [44] Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, et al. APC mutations occur early during colorectal tumorigenesis. Nature 1992;359(6392):235—7 [Epub 1992/09/17]. [45] Nault JC, Bioulac-Sage P, Zucman-Rossi J. Hepatocellular benign tumors-from molecular classification to personalized clinical care. Gastroenterology 2013;144(5):888—902 [Epub 2013/03/15]. [46] Bioulac-Sage P, Rebouissou S, Thomas C, Blanc JF, Saric J, Sa Cunha A, et al. Hepatocellular adenoma subtype classification using molecular markers and immunohistochemistry. Hepatology 2007;46(3):740—8 [Epub 2007/08/01]. [47] Zucman-Rossi J, Jeannot E, Nhieu JT, Scoazec JY, Guettier C, Rebouissou S, et al. Genotype-phenotype correlation in hepatocellular adenoma: new classification and relationship with HCC. Hepatology 2006;43(3):515—24 [Epub 2006/02/24]. [48] Bluteau O, Jeannot E, Bioulac-Sage P, Marques JM, Blanc JF, Bui H, et al. Bi-allelic inactivation of TCF1 in hepatic adenomas. Nat Genet 2002;32(2):312—5 [Epub 2002/10/02].

[49] Rebouissou S, Amessou M, Couchy G, Poussin K, Imbeaud S, Pilati C, et al. Frequent in-frame somatic deletions activate gp130 in inflammatory hepatocellular tumours. Nature 2009;457(7226):200—4 [Epub 2008/11/21]. [50] Nault JC, Fabre M, Couchy G, Pilati C, Jeannot E, Tran Van Nhieu J, et al. GNAS-activating mutations define a rare subgroup of inflammatory liver tumors characterized by STAT3 activation. J Hepatol 2012;56(1):184—91 [Epub 2011/08/13]. [51] Pilati C, Amessou M, Bihl MP, Balabaud C, Nhieu JT, Paradis V, et al. Somatic mutations activating STAT3 in human inflammatory hepatocellular adenomas. J Exp Med 2011;208(7):1359—66 [Epub 2011/06/22]. [52] Pilati C, Letouze E, Nault JC, Imbeaud S, Boulai A, Calderaro J, et al. Genomic profiling of hepatocellular adenomas reveals recurrent FRK-activating mutations and the mechanisms of malignant transformation. Cancer Cell 2014;25(4):428—41 [Epub 2014/04/17]. [53] Eichenmuller M, Trippel F, Kreuder M, Beck A, Schwarzmayr T, Haberle B, et al. The genomic landscape of hepatoblastoma and their progenies with HCC-like features. J Hepatol 2014;61(6):1312—20 [Epub 2014/08/20]. [54] Qu Y, Shi L, Wang D, Zhang B, Yang Q, Ji M, et al. Low frequency of TERT promoter mutations in a large cohort of gallbladder and gastric cancers. Int J Cancer 2014;134(12):2993—4. [55] Fujimoto A, Furuta M, Shiraishi Y, Gotoh K, Kawakami Y, Arihiro K, et al. Whole genome mutational landscape of liver cancers displaying biliary phenotype reveals hepatitis impact and molecular diversity. Nat Commun 2015;6:6120. [56] Cesare AJ, Reddel RR. Alternative lengthening of telomeres: models, mechanisms and implications. Nat Rev Genet 2010;11(5):319—30. [57] Harley CB. Telomerase and cancer therapeutics. Nat Rev Cancer 2008;8(3):167—79. [58] Greten TF, Forner A, Korangy F, N’Kontchou G, Barget N, Ayuso C, et al. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer 2010;10:209.

Please cite this article in press as: Nault J-C, Zucman-Rossi J. TERT promoter mutations in primary liver tumors. Clin Res Hepatol Gastroenterol (2015), http://dx.doi.org/10.1016/j.clinre.2015.07.006