Neuroendocrine tumours: cracking the epigenetic code

in Endocrine-Related Cancer
Authors:
A Karpathakis University College London Cancer Institute, Neuroendocrine Tumour Unit, 72 Huntley Street, London WC1E 6BT, UK
University College London Cancer Institute, Neuroendocrine Tumour Unit, 72 Huntley Street, London WC1E 6BT, UK

Search for other papers by A Karpathakis in
Current site
Google Scholar
PubMed
Close
,
H Dibra University College London Cancer Institute, Neuroendocrine Tumour Unit, 72 Huntley Street, London WC1E 6BT, UK

Search for other papers by H Dibra in
Current site
Google Scholar
PubMed
Close
, and
C Thirlwell University College London Cancer Institute, Neuroendocrine Tumour Unit, 72 Huntley Street, London WC1E 6BT, UK
University College London Cancer Institute, Neuroendocrine Tumour Unit, 72 Huntley Street, London WC1E 6BT, UK

Search for other papers by C Thirlwell in
Current site
Google Scholar
PubMed
Close

Free access

Sign up for journal news

The field of epigenetics has evolved rapidly over recent years providing insight into the tumorigenesis of many solid and haematological malignancies. Determination of epigenetic modifications in neuroendocrine tumour (NET) development is imperative if we are to improve our understanding of the biology of this heterogenous group of tumours. Epigenetic marks such as DNA methylation at RASSF1A are frequent findings in NETs of all origins and may be associated with worse prognosis. MicroRNA signatures and histone modifications have been identified which can differentiate subtypes of NET and distinguish NET from adenocarcinoma in cases of diagnostic uncertainty. Historically, candidate gene-driven approaches have yielded limited insight into the epigenetics of NET. Recent progress has been facilitated by development of high-throughput tools including second-generation sequencing and arrays for analysis of the ‘epigenome’ of tumour and normal tissue, permitting unbiased approaches such as exome sequencing that identified mutations of chromatin-remodelling genes ATRX/DAXX in 44% of pancreatic NETs. Epigenetic changes are reversible and therefore represent an attractive therapeutic target; to date, clinical outcomes of epigenetic therapies in solid tumours have been disappointing; however, in vitro studies on NETs are promising and further clinical trials are required to determine utility of this class of novel agents. In this review, we perform a comprehensive evaluation of epigenetic changes found in NETs to date, including rare NETs such as phaeochromocytoma and adrenocortical tumours. We suggest priorities for future research and discuss potential clinical applications and novel therapies.

Abstract

The field of epigenetics has evolved rapidly over recent years providing insight into the tumorigenesis of many solid and haematological malignancies. Determination of epigenetic modifications in neuroendocrine tumour (NET) development is imperative if we are to improve our understanding of the biology of this heterogenous group of tumours. Epigenetic marks such as DNA methylation at RASSF1A are frequent findings in NETs of all origins and may be associated with worse prognosis. MicroRNA signatures and histone modifications have been identified which can differentiate subtypes of NET and distinguish NET from adenocarcinoma in cases of diagnostic uncertainty. Historically, candidate gene-driven approaches have yielded limited insight into the epigenetics of NET. Recent progress has been facilitated by development of high-throughput tools including second-generation sequencing and arrays for analysis of the ‘epigenome’ of tumour and normal tissue, permitting unbiased approaches such as exome sequencing that identified mutations of chromatin-remodelling genes ATRX/DAXX in 44% of pancreatic NETs. Epigenetic changes are reversible and therefore represent an attractive therapeutic target; to date, clinical outcomes of epigenetic therapies in solid tumours have been disappointing; however, in vitro studies on NETs are promising and further clinical trials are required to determine utility of this class of novel agents. In this review, we perform a comprehensive evaluation of epigenetic changes found in NETs to date, including rare NETs such as phaeochromocytoma and adrenocortical tumours. We suggest priorities for future research and discuss potential clinical applications and novel therapies.

Introduction

Epigenetics can be defined as the study of heritable changes in gene expression without alteration of the underlying DNA sequence. In recent years, the understanding of epigenetic drivers of tumorigenesis has developed rapidly in part due to improvements in sequencing technology. While the study of genetics – mutations, deletions and alterations to DNA – has led to many significant breakthroughs in medicine, it is increasingly apparent that for many human diseases epigenetic drivers are key to pathogenesis and may have significant impact on clinical practice and patient management. Integration of genetics, epigenetics and gene expression can generate a holistic understanding of the molecular basis of malignancy. The recent discovery of mutations in the novel tumour suppressor genes and chromatin remodellers ATRX/DAXX through exome sequencing represent significant progress in our understanding of neuroendocrine tumour (NET) development.

NETs are a heterogenous group of tumours, which until recently have remained largely intractable to genetic characterisation, with many research studies being performed with a small number of samples leading to the identification of low-frequency mutations, which do not appear to have significant prognostic or therapeutic impact. There is significance to the many negative findings in the research of NET genetics: classical tumour suppressor and oncogenes implicated in the development of many solid tumours do not appear to play a significant role in NET pathogenesis (for example P53 (TP53), RB (RB1) and KRAS are only very infrequently mutated or deleted in NET), suggesting the presence of alternative pathogenic drivers (Yoshimoto et al. 1992, Yashiro et al. 1993, Chung et al. 1997a ).

Here, we provide a review into the current knowledge of epigenetic changes in NET to facilitate further research and identification of potential epigenetic drivers of NET tumorigenesis and identify novel biomarkers.

Epigenetic modifications

DNA methylation is the best-understood and most stable epigenetic mark; it occurs primarily at the 5′-position of the cytosine ring within CpG dinucleotides. CpG sites are clustered in ‘islands’ in the promoter region of up to 70% of genes, and methylation is facilitated by DNA methyltransferase enzymes (DNMTs). Three families of DNMT have been identified: DNMT1, which maintains methylation, DNMT2, whose specific role is unknown, and DNMT3, which functions as a de novo methyltransferase (Bestor 1992, Okano et al. 1998a , b , 1999). Recent identification of conversion of 5-methylcytosine to 5-hydroxymethylcytosine by the TET group of enzymes provides further insight into the complexities of regulating gene expression, but this has not yet been investigated in NET (Tahiliani et al. 2009). Patterns of DNA methylation are tissue specific; therefore, comparisons of tumour and normal tissue methylation must be carefully matched to reduce artefactual results.

Histones are the primary component of chromatin and function to package DNA into nucleosomes. Post-translational covalent histone modifications regulate accessibility of genes to transcription factors by formation of binary states either ‘permissive/open’ or ‘repressive/closed’, and the establishment and maintenance of chromatin structure help to maintain cellular identity through repression of non-essential genes (Chi & Bernstein 2009). Histone modifications include acetylation, methylation and phosphorylation; these are catalysed by several enzymes, for example histone acetyltransferases and deacetylases (HDACs).

MicroRNAs (miRNAs) are small single-stranded RNA molecules of ∼22 nucleotides in length, which post-transcriptionally regulate gene expression. Over the past decade, there has been a growing interest in miRNA and their involvement in the contribution of the development of cancer, and it has been recently shown that miRNA profiling can classify human cancers (Lu et al. 2005). Figure 1 illustrates the primary mechanisms of epigenetic alteration. These processes work synergistically to modify gene expression and determine cellular phenotype, in particular the maintenance of the differentiated cellular state. Influence from environmental factors such as diet and smoking is known to affect epigenetic status and is likely to play a significant role in tumorigenesis (Feil & Fraga 2011).

Figure 1
Figure 1

Mechanisms of histone modification and DNA methylation in human tissues.

Citation: Endocrine-Related Cancer 20, 3; 10.1530/ERC-12-0338

Key epigenetic findings in cancer

Epigenetic changes are known to play a fundamental role in cancer development (Jones & Baylin 2002, Jones & Martienssen 2005). Early research identifying global hypomethylation of tumours relative to normal tissue demonstrated one of the first documented epigenetic alterations in human cancer cell lines in 1983 (Feinberg & Vogelstein 1983). Global hypomethylation increases during tumour development (Fraga et al. 2004) and is believed to contribute to chromosomal instability (Eden et al. 2003). Hypermethylation of CpG islands in the promoter regions of tumour suppressor genes has been recognised as an early event in many cancers including retinoblastoma (Greger et al. 1989, Sakai et al. 1991). Promoter hypermethylation has subsequently been identified at sites such as VHL, BRCA, RASSF1A and p16INK4a (CDKN2A) where associated loss of expression is seen (Herman et al. 1994, Merlo et al. 1995, Catteau et al. 1999, Dammann et al. 2000). Next-generation sequencing permits definition of full tumour ‘methylomes’ at single-nucleotide resolution, and there are currently large-scale international collaborative studies underway that will provide freely available epigenetic data for normal and tumour tissues; although due to their relative rarity, NETs are not included in most of these projects (The American Association for Cancer Research Human Epigenome Task Force & European Union 2008, Chadwick 2012).

Histone modifications also contribute to tumorigenesis, for example global reduction of acetylation at Lys16 (H4K16Ac) and of trimethylation at Lys20 of histone H4 (H4K20me3) is observed in several cancers (Fraga et al. 2005). Dimethylation of histone H3 at lysine 4 (H3K4diMe) has been shown to be associated with an activated transcriptional state and is predictive of outcome in prostate and lung cancer (Seligson et al. 2005, Barlesi et al. 2007).

Epigenetic aberrations often preceded ‘classical’ genetic changes found in cancer such as tumour suppressor gene mutation, proto-oncogene activation and genomic instability and could therefore be utilised as early biomarkers. Hypermethylation of glutathione S transferase (GSTP1) is a highly specific diagnostic biomarker in prostate cancer detectable in blood and urine (Cairns et al. 2001) and hypermethylation of the DNA repair gene MGMT is a predictive biomarker of response to temozolomide (Hegi et al. 2005). There are few studies that have specifically investigated miRNA expression in NETs; although in other tumour types, miRNA signatures show utility as diagnostic and predictive biomarkers (Liu et al. 2012, Wang et al. 2012).

A key element driving therapeutic approaches to epigenetic modifications is the reversibility of DNA methylation and histone marks – this has been exploited successfully through histone deacetylase inhibitors (HDACi) and DNA demethylators. Two agents, 5-azacytidine and 5-aza-2′-deoxycytidine (decitabine), have been approved for treatment of myelodysplastic syndrome and leukaemia respectively (Muller et al. 2006, Oki et al. 2007). In ovarian cancer, demethylating agents have been shown to cause resensitisation to platinum in heavily pretreated populations resulting in response rates of up to 35% and progression-free survival of 5.6–10.2 months (Fu et al. 2011, Matei et al. 2012). However, other early phase trials in solid tumours have been largely disappointing, and to date, there have been no clinical trials of these two agents in NET patients. Figure 2 provides a timeline of key developments in the field of epigenetics, alongside key NET research findings.

Pancreatic NETs

Pancreatic NETs (pNETs) comprise functioning tumours (40–55%) including gastrinoma, insulinoma, VIPoma and glucagonoma as well as non-functional NETs. Overall 5-year survival for pNET is 80% for all stages and 25% for metastatic disease; median survival is only 10 months for high-grade/poorly differentiated tumours. Improvements in prognostication and personalisation of treatment are required in order to improve patient outcomes. PNETs are perhaps the best-characterised group of NET, but despite this, reports of frequency of epigenetic marks vary significantly between studies and will require stringent validation before clinical applications can be developed. Insulinomas display more indolent clinical behaviour than other pNETs, and this is reflected in the emergence of distinct epigenetic patterns in this class of tumours.

DNA methylation in pNET

RASSF1A promoter region is frequently hypermethylated in pNET

The Ras-association domain gene family 1 (RASSF1) tumour suppressor gene acts to induce cell cycle arrest and is inactivated in a wide variety of human cancers, usually as a result of epigenetic promoter methylation, somatic mutations of the gene bring extremely rare (Shivakumar et al. 2002, Dammann et al. 2003b ). Different isoforms of RASSF1 exist and are associated with distinct promoter regions and alternative splicing. Within this review, we cite RASSF1 as described in the original research articles.

The largest study of DNA methylation in NETs included 48 largely non-functional (94%) pNETs, which were analysed for methylation of the promoter region of 11 candidate tumour suppressor genes. The most commonly hypermethylated promoter region was that of RASSF1A (75%), which was unmethylated in the adjacent normal tissue. Fourteen cases had synchronous hepatic metastases, the majority of which demonstrated identical methylation status as the primary tumour (House et al. 2003). Separate studies have identified RASSF1 promoter methylation in 80–83% of pNETs making it the most frequently altered gene in sporadic pNET identified to date. In some cases, methylation is also identified in matched normal pancreas, however at a lower level of expression (Dammann et al. 2003a , Malpeli et al. 2011). RASSF1 methylation has been noted to be more frequent in metastatic tumours (100%) than non-metastatic (71%) (Malpeli et al. 2011). Methylation of the RASSF1 promoter occurs less frequently in pancreatic ductal adenocarcinoma (Dammann et al. 2003a ).

In contrast to RASSF1A, the RASSF1C isoform expression is 11.4 times higher in pNET than in normal tissue (P=0.001) (Malpeli et al. 2011). RASSF1C has been shown to have a role inhibiting β-catenin degradation and given the above findings may also have a pathogenic role in pNET development (Estrabaud et al. 2007). RASSF1B is ubiquitously expressed in both tumour and normal tissue (Toyota et al. 1999).

CDKN2A promoter methylation distinguishes subtypes of pNET and may be prognostic

Cyclin-dependent kinase inhibitor 2a/P16INK4a is a tumour suppressor gene that plays a vital role in cell cycle regulation. Loss of heterozygosity on chromosome 9p21 (where P16/CDKN2A is located) is one of the most frequent genetic alterations in human cancer, and promoter methylation at this site is a common feature of many malignancies including breast (33%), prostate (60%) and colorectal cancer (92%) (Herman et al. 1995).

Methylation at the CDKN2A/P16 locus has been demonstrated in QGP1 (a pNET cell line) (Lubomierski et al. 2001) and in 40% of pNETs (associated with early tumour recurrence and reduced overall survival) (House et al. 2003). Unlike RASSF1, methylation of CDKN2A/p16INK4a is less frequent in pNET than in adenocarcinoma (Dammann et al. 2003a ). In two studies of gastrinomas, hypermethylation of the CDKN2A/p16INK4a promoter has been found in 52–62.5% of cases independent of disease stage, location of primary or prognosis, suggesting that methylation of the CDKN2A/p16INK4a promoter may be an early occurrence in gastrinoma tumorigenesis (Serrano et al. 2000, de Wilde et al. 2012). By contrast, insulinomas have been shown to have a low frequency (17%) of CDKN2A/p16INK4a alterations (Bartsch et al. 2000).

Methylation of other loci may be relevant to pNET development

TIMP3 promoter hypermethylation has been demonstrated in 44% of pNETs and correlated with the presence of metastases. Interestingly, none of the five insulinomas in this cohort showed methylation at this site (Wild et al. 2003). The MGMT promoter is methylated in 40% of pNET and may be a useful predictive biomarker of response to temozolomide chemotherapy (Dammann et al. 2003b ). Only 6% of pNETs show hypermethylation of VHL (compared with 18% showing deletion of the gene), suggesting that VHL promoter methylation is not a significant mechanism of tumorigenesis (Schmitt et al. 2009).

CpG island methylator phenotype positivity is a poor prognostic marker in pNET

CpG island methylator phenotype (CIMP) positivity (a term denoting a high degree of methylation across multiple CpG sites) in 20% of a cohort of 71 mixed pancreatic and gastrointestinal NETs corresponded with higher grade tumours (Ki67>10%), while CIMP negativity was associated with improved overall survival (Arnold et al. 2007). This is in keeping with methylation at three or more loci of selected candidate genes being associated with early recurrence and worse prognosis in a separate study (House et al. 2003).

Insulinomas are distinguished from other pNET subtypes through differences in DNA methylation

A large study with a mixed cohort of 62 NET cases were analysed for DNA methylation at three regions associated with the IGF2 gene previously identified as differentially methylated in mouse models (Fontaniere et al. 2006, Dejeux et al. 2009). The gastrinomas, non-functional pNETs and small intestinal NETs in the cohort exhibited hypomethylation of varying degrees at these sites; by contrast, the 11 insulinomas were hypermethylated at these regions. This correlated with insulinoma-specific overexpression of IGF2 RNA and protein thought to be caused by loss of imprinting and reactivation of the normally silent maternal allele (Dejeux et al. 2009).

Promoter methylation of MLH1 has been identified in 31% of insulinomas and correlated with poor prognosis (Mei et al. 2009), and unlike most pNETs, TIMP3 and CDKN2A/p16INK4a methylation are not common features of insulinomas (Bartsch et al. 2000, Wild et al. 2003). In Table 1, we summarise genes known to be methylated in pNET as well as other anatomical origins.

Table 1

Summary of loci identified with aberrant methylation in neuroendocrine tumours

Association
Locus Pancreatic NET GI NET Bronchial NET Reference
CDKN2a/P16INK4a HPM 62.5% gastrinomas Muscarella et al. (1998)
HPM 50% non-functional Muscarella et al. (1998)
HPM 52% gastrinomas Serrano et al. (2000)
HPM 11.8% insulinomas Bartsch et al. (2000)
HPM in QGP1 but not BON Lubomierski et al. (2001)
HPM 17% Dammann et al. (2003a)
HPM 0% insulinomas Dammann et al. (2003a)
HPM 0% appendiceal HPM NCI-H727 and HTB-119 Zhang et al. (2006)
HPM 84.8% metastatic Zhang et al. (2006)
HPM 60.6% non-metastatic Zhang et al. (2006)
RASSF1 HPM 83% Dammann et al. (2003a)
HPM 75% House et al. (2003)
HPM 63% HPM 69% HPM 40% Liu et al. (2005)
HPM 32% (foregut GEP) Pizzi et al. (2005)
HPM 71.3% (foregut GEP) Arnold et al. (2007)
HPM 71% fore-midgut GEP, 38% colorectal Arnold et al. (2008)
HPM promoter 1 Pelosi et al. (2010)
HPM 80% Malpeli et al. (2011)
HPM 20–40% colorectal La Rosa et al. (2012)
TIMP3 HPM 0% House et al. (2003)
HPM 44% Wild et al. (2003)
HPM 0% insulinomas Wild et al. (2003)
HPM 0% (foregut GEP) Arnold et al. (2007)
HPM 0% fore-midgut GEP, 9% colorectal Arnold et al. (2008)
HPM 20–40% colorectal La Rosa et al. (2012)
MGMT HPM 25% Chan et al. (2003)
HPM 40% House et al. (2003)
HPM 13% HPM 0% HPM 27% Liu et al. (2005)
HPM 16.1% (foregut GEP) Arnold et al. (2007)
HPM 21% fore-midgut GEP, 38% colorectal Arnold et al. (2008)
hMLH1 HPM 23% House et al. (2003)
HPM 0% (foregut GEP) Arnold et al. (2007)
HPM 0% fore-midgut GEP, 12% colorectal Arnold et al. (2008)
HPM 31% insulinomas Mei et al. (2009)
P16 HPM 19% HPM 25% HPM 33% Liu et al. (2005)
HPM 40% House et al. (2003)
HPM 0% (foregut GEP) Arnold et al. (2007)
HPM 0% fore-midgut GEP, 24% colorectal Arnold et al. (2008)
APC HPM 21% House et al. (2003)
HPM 54.1% (foregut GEP) Arnold et al. (2007)
HPM 63% fore-midgut GEP, 44% colorectal Arnold et al. (2008)
HPM 20–40% colorectal La Rosa et al. (2012)
CTNNB1 HPM 0% appendiceal Zhang et al. (2006)
HPM 57.6% metastatic Zhang et al. (2006)
HPM 27.3% non-metastatic Zhang et al. (2006)
HIC1 83.1% (foregut GEP) Arnold et al. (2007)
HPM 55% fore-midgut GEP, 47% colorectal Arnold et al. (2008)
HPM 20–40% colorectal La Rosa et al. (2012)
E-cadherin HPM 23% House et al. (2003)
HPM 1.3% (foregut GEP) Arnold et al. (2007)
RAR β HPM 25% Chan et al. (2003)
HPM 20–40% colorectal La Rosa et al. (2012)
MEN1 HPM 14.3% (foregut GEP) Arnold et al. (2007)
HPM 18% fore-midgut GEP, 44% colorectal Arnold et al. (2008)
PTEN HPM 1.5% (foregut GEP) Arnold et al. (2007)
HPM 20–40% colorectal La Rosa et al. (2012)
P14 HPM 0% House et al. (2003)
HPM 44% HPM 69% HPM 33% Liu et al. (2005)
GATA5 HPM >80% colorectal La Rosa et al. (2012)
ESR1 HPM 80% colorectal La Rosa et al. (2012)
GST HPM 0% House et al. (2003)
RUNX3 HPM 6.6% (foregut GEP) Arnold et al. (2007)
P14 HPM 44% Chan et al. (2003)
THBS1 HPM 44% Chan et al. (2003)
RAR (RARA) HPM 25% House et al. (2003)
P73 (TP73) HPM 17% House et al. (2003)
VHL HPM 6% Schmitt et al. (2009)
WT1 HPM 60–80% colorectal La Rosa et al. (2012)
CDH13 HPM 40–60% colorectal La Rosa et al. (2012)
CIMP status CIMP+ve 29% fore-midgut GEP, 59% colorectal Arnold et al. (2008)

HPM, hypermethylated; NET, neuroendocrine tumour; GEP, gastroenteropancreatic; MSP, methylation-specific PCR; GI, gastrointestinal; DMR, differentially methylated region; DNMT, DNA methyltransferase; CIMP, CpG island methylator phenotype; MANEC, mixed adenoneuroendocrine carcinoma.

Chromatin remodellers in pNET

In a recent study (Jiao et al. 2011), exome sequencing was performed in ten sporadic pNETs followed by Sanger sequencing validation in a cohort of 58 pNETs identifying commonly mutated genes implicated in chromatin remodelling. Forty-four per cent of tumours had inactivating mutations of MEN1 and 43% had mutations in either DAXX or ATRX (25 and 18% respectively) (death domain-associated protein or alpha thalassemia/mental retardation syndrome X-linked) (Jiao et al. 2011).

MEN1 encodes the transcription factor menin, which recruits the H3K4me3 histone methyltransferase mixed lineage leukaemia complex that plays an essential role in chromatin remodelling (Agarwal et al. 1999). ATRX and DAXX had not previously been associated with NETs and are novel tumour suppressor genes, although ATRX has previously been implicated in glioma. The proteins encoded by ATRX and DAXX have multiple functions including chromatin remodelling during heterochromatin assembly at repetitive guanine-rich regions (such as telomeres) where they are required for incorporation of the histone variant H3.3 (Heaphy et al. 2011a ). Mutations in ATRX and DAXX were mutually exclusive but overlapped with MEN1 mutations in 16 cases (23.5%). The presence of mutations in ATRX/DAXX was associated with improved prognosis.

NETs harbouring ATRX/DAXX mutations have been shown to demonstrate alternative lengthening of telomeres (ALT), indicating that they do not increase activity of telomerase in the face of extensive cell proliferation but maintain telomeres through alternative mechanisms (Heaphy et al. 2011a ). All 19 tumours with ATRX/DAXX mutations from the Jiao study were found to have ALT positivity – a 100% correlation (P<0.008 for each gene), while in human cancers in general, the ALT phenotype is present in only 3% (Heaphy et al. 2011b ). The association of ATRX/DAXX with ALT has subsequently been confirmed in other tumour types including glioblastoma and osteosarcoma substantiating the role of ATRX/DAXX in telomere maintenance (Schwartzentruber et al. 2012). ALT phenotype has been found to be a favourable prognostic factor, possibly due to the reduction of chromosomal instability observed in these tumours (Ulaner et al. 2003).

The role of ATRX/DAXX was subsequently investigated in 28 multiple endocrine neoplasia 1 (MEN1) syndrome-related pNETs, loss of ATRX/DAXX expression occurred in only 6% of these tumours, all of which were >3 cm in size and demonstrated ALT phenotype, suggesting that ATRX/DAXX mutation is a late event in MEN1-associated pNET development (de Wilde et al. 2012). Forty-seven microadenomas (<0.5 cm) were included in the study, none of which showed ATRX/DAXX mutation or ALT positivity.

There has been little research investigating histone modifications in pNETs; however, given the frequency and prognostic impact of modifications related to chromatin remodelling, it is recommended that histone modifications should be studied more extensively in pNETs. One study was identified assessing mutation or aberrant expression of HDAC11 (selected for study due to its position at locus 3p25 which is associated with a high rate of loss of heterozygosity in pNETs) in a small cohort of sporadic malignant pNETs detected no tumour specific events suggesting that HDAC11 is unlikely to play a role in these tumours (Chung et al. 1997b , Barghorn et al. 2001).

miRNA expression in pNET

miRNAs 103/107/155 can distinguish pNET from normal pancreatic tissue

Roldo et al. (2006) investigated the global expression of miRNAs in pNETs. Analysis of global miRNA expression of 44 pancreatic primary tumours determined a common pattern of miRNA expression distinguishing pNETs from normal pancreas: expression of miR-103 and miR-107 associated with lack of expression of miR-155. Overexpression of miR-204 was specific to insulinomas and correlated with immunohistochemical expression of insulin. MiR-21 was strongly associated with a high Ki67 proliferation index and liver metastases and may be a useful prognostic marker.

Gastrointestinal NETs

Approximately two thirds of gastroenteropancreatic NET arise from the intestine, these comprise foregut, the classical ‘midgut’ NETs, and the typically higher grade hindgut and rectal NETs, which are associated with poorer prognosis. Interest in the methylation status of GI NET has been encouraged by understanding of the role of methylation and CIMP in colorectal adenocarcinoma. Methylation of certain loci in GI NET appears to be related to metastatic progression, and there may be a distinct subgroup of colorectal cancer patients with CIMP+/MSI+ who experience an improved overall prognosis. The limited studies of histone modifications in these tumours have been less informative.

DNA methylation in gastrointestinal NET

Gastrointestinal and pNETs have different methylation patterns

One of the earliest studies of DNA methylation in NET compared with CpG island methylation in 16 GI NETs and 11 pNETs showed differing profiles of methylation across a panel of 14 candidate genes commonly implicated in gastrointestinal malignancy, reflecting differing molecular pathogenesis. Specifically, GI NETs were found to be significantly more frequently methylated at the promoter regions of the following genes compared to pNETs: MGMT (25 vs 0% P=0.03), THSB1 (44 vs 9% P=0.04), P14 (44 vs 9% P=0.04), P16 (31 vs 9%) and RARβ (25 vs 0% P=0.03). PNETs were more frequently methylated at ER (64%), and of note, MEN1 was not methylated in any of the samples (Chan et al. 2003).

Global hypomethylation is a feature of gastrointestinal NET

Methylation of repeat elements such as long interspersed nucleotide elements (LINE1 (L1RE1)) and Alu is a surrogate for global methylation levels. A study of 35 NETs (15 ileal, 11 pancreatic and nine others) demonstrated that all tumour samples were hypomethylated relative to normal tissue at these sites (LINE1 P=0.04, Alu P=0.001). Global hypomethylation was more prevalent in GI NET than pancreatic and was correlated with poor prognosis, lymph node metastasis and loss of chromosome 18 (Choi et al. 2007).

However, in a more recent study of 43 GI NETs and 15 pNETs, LINE1 hypomethylation was identified in 100% pNETs, 50% gastric NETs, 82% midgut NETs and 100% colorectal NETs while NETs metastatic to lymph nodes were less frequently hypomethylated than non-metastatic NET (Stricker et al. 2012). We can conclude that global hypomethylation is a feature of pNETs, and while it is generally considered a poor prognostic marker in most solid tumours, the prognostic implication in pNET remains to be determined.

RASSF1A and CTNNB1 promoters are frequently methylated in gastrointestinal NETs

Two interesting studies of RASSF1A methylation in GI NET have been performed. In the first study, RASSF1A methylation was identified in 32% (20/62) GI NETs and was restricted to foregut tumours (Pizzi et al. 2005). The second study determined that frequency of RASSF1A methylation increased in metastatic lesions compared with primary tumours in a study of 33 midgut NET cases (61% methylated in primary tumours vs 85% in matched metastatic lesions). CTNNB1 promoter methylation is also more frequent in metastatic than in primary GI NET tumours (57.6 vs 27.3% P=0.004) and is not seen in normal tissue (Zhang et al. 2006). None of the six appendiceal NETs included in the study (all non-metastatic as most appendiceal NETs) displayed RASSF1A or CTNNB1 methylation (Zhang et al. 2006). These results suggest that RASSF1A may be involved in the development of endocrine tumours of the foregut and midgut but not hindgut and that RASSF1A/CTNNB1 may be associated with progression to metastasis.

CIMP may be a prognostic biomarker for colorectal NET

Recent studies have demonstrated CIMP positivity in 37–59% of poorly differentiated colorectal NETs (Arnold et al. 2008, La Rosa et al. 2012) compared with 29% of well-differentiated fore/midgut NETs. In one study, survival was not influenced by CIMP status; however, La Rosa et al. demonstrated a 100% correlation of CIMP positivity with the presence of microsatellite instability, and this was associated with better prognosis (MSI/CIMP+ve vs others P=0.04).

Expression of DNMT1, -3A and -3B has been demonstrated to be significantly higher in stage IV GEP NET than in stage I or II. DNMT3A (a de novo methyltransferase) was significantly higher in poorly differentiated carcinomas relative to well-differentiated tumours (P<0.01), and DNMT3A and -3B both showed significantly lower expression in midgut NET relative to fore- or hindgut tumours. We can conclude that overexpression of DNMT1, -3A and -3B is a common feature of GEP NET, and these enzymes may play a role in the development of NET and are therefore a potential novel therapeutic target (Rahman et al. 2010). The relationship between DNMT activity and CIMP is not fully characterised and further large studies are required to resolve the relationship between the activity of these enzymes, methylation status and prognosis in GI NET.

Methylation of other loci may be relevant in colorectal NET

In a study including 34 colorectal and 38 fore-midgut NETs, promoter methylation of CDKN2A/p16INK4a, hMLH1 and TIMP3 was only detected in NET of colorectal origin. CDKN2A/p16INK4a methylation was only present in poorly differentiated colorectal NET (P=0.01) and was a more significant prognostic marker of poor outcome than Ki67 (P=0.0004) (Arnold et al. 2008). Altered expression of MLH1 by both genetic and epigenetic mechanisms is an area of exciting research in colorectal adenocarcinoma and may be significant in the development of both endocrine and exocrine tumours of this site. Methylation of the GATA4/5 promoter (a gene known to be frequently methylated in colorectal adenocarcinoma) is present in >90% of colorectal NET and MANEC (La Rosa et al. 2012). Table 1 provides a summary of loci differentially methylated in GI NET.

Histone modifications in gastrointestinal NET

Research of histone modifications in NETs is limited to studies on small numbers of tumours. H3K4diMe and the associated methylating (Ash2 complex) and demethylating (LSD1) enzymes were studied in gastrointestinal carcinomas including a group of 16 mostly low-grade primary intestinal NETs (Magerl et al. 2010). Up to 100% of neuroendocrine carcinomas exhibited strong immunostaining for Ash2 and LSD1, while H3K4diMe was strongly expressed in 93% of neuroendocrine carcinomas, other carcinomas (such as HCC and pancreatic adenocarcinomas) showing lower expression. In matched normal tissue, all three markers showed weak expression. This suggests a potential role for modifications at the histone H3K4 locus in intestinal NETs but requires further validation.

Histone H1x has been demonstrated to be highly expressed in NETs but also in the corresponding non-neoplastic neuroendocrine cells of the pancreas and small intestine and therefore may reflect cell of origin rather than tumorigenic feature (Warneboldt et al. 2008).

miRNA expression in gastrointestinal NET

MiR-133a is down-regulated in metastatic progression of ileal NET

Two miRNA expression profile analyses provide evidence that miRNAs are dysregulated specifically in ileal NET progression: down-regulation of miR-133a, -145, -146, -222 and -10b and up-regulation of miR-183, -488 and -19a+b in metastatic tumours compared to primary tumours (Ruebel et al. 2010) and up-regulation of miR-96, -182, -183, -196a and -200a and down-regulation of miR-31, -129-5p, -133a and -215 (Li et al. 2012). These results suggest that miRNAs may play a role in GI NET progression, in particular miR-133a that was identified in both studies and may be a useful focus for further investigation.

miRNAs can distinguish colorectal NET from adenocarcinoma

A case of colorectal NET was included in comparison of miRNA expression in colorectal adenocarcinoma and normal mucosa, which identified 38 up-regulated miRNAs in colorectal NET compared with the normal mucosa. Of these, only six were found to be up-regulated in colorectal adenocarcinoma, suggesting distinct miRNA expression pattern in colorectal NET compared with adenocarcinomas (Hamfjord et al. 2012). Four miRNAs identified as up-regulated in colorectal NET were also noted in the analyses of ileal NET described earlier: miR-96, -182, -196a and -488; these would be good candidates for further investigation; however, it must be noted that this study only analysed one NET case and analysis of a larger sample size and validation of the data is required.

Bronchial NETs

Bronchial NETs can be classified by cellular morphology into small-cell (SCLC), large-cell (LCNEC), ‘atypical’ carcinoid (AC) and ‘typical’ carcinoid (TC) tumours. In this review, we focus on the typical and atypical carcinoid subtypes of bronchial NET.

DNA methylation in bronchial NET

The RASSF1 promoter is hypermethylated in bronchial NET

Analysis of methylation of the RASSF1 promoter regions in a cohort of 58 bronchial NETs, matched normal tissue and 20 control non-small-cell lung cancers (NSCLC) identified that promoter 1 was hypermethylated in NET but not normal tissue or non-endocrine tumours. The degree of hypermethylation correlated with tumour grade and corresponding global loss of RASSF1A/E mRNA expression (Pelosi et al. 2010), and RASSF1 promoter 1 methylation may represent a useful biomarker in lung NET to differentiate high- from low-grade NET. Up-regulation of RASSF1C was found to be an independent adverse prognostic factor in high-grade bronchial NETs (Pelosi et al. 2010).

P15.5 promoter methylation may be implicated in low-grade bronchial NET

Similar to the tumour suppressor gene CDKN2A/P16INK4a, P15INK4b (CDKN2B) also encodes a cyclin-dependent kinase inhibitor, which functions in cell cycle regulation, but its role in tumorigenesis is less well characterised. A series of five low-grade and 15 high-grade bronchial NETs showed aberrant methylation at the 5′-region of the p15INK4b gene in 15% of tumours but not in control/normal lung tissue, and there was a highly variable expression of isoform p15.5 in the low-grade bronchial NETs, suggesting potential involvement of this gene in the ‘carcinoid’-type bronchial tumours. P15 status was shown to be independent of P16 and P14 (Chaussade et al. 2001).

Histone modifications in bronchial NET

Histone 4 modifications can differentiate low- and high-grade bronchial NETs

A case series of 32 bronchial NETs demonstrated progressive loss of two different histone marks, H4KA16 and H4KM20, from low- to high-grade tumours, and found that Ki67 was inversely correlated with both H4KA16 and H4KM20 (P<0.05) (Li et al. 2011).

EZH2 is strongly expressed in high-grade bronchial NET

Increased expression of enhancer of zeste homolog 2 (EZH2), a molecule involved in methylation of histone H3 (H3K27), has been demonstrated in many human malignancies and is associated with poor prognosis (Bachmann et al. 2006). EZH2 was demonstrated to be strongly expressed in all SCLC and LCNEC (80–90% of cells) in a cohort of 54 bronchial NETs, but only rare scattered expression was seen in typical and atypical bronchial NETs (Findeis-Hosey et al. 2011). EZH2 may play a role in the development of high-grade NETs and may be a useful diagnostic biomarker.

miRNA expression in bronchial NET

miRNA-21 and -155 distinguish high- and low-grade bronchial NETs

In a recent study evaluating expression of selected miRNAs in 63 bronchial NETs, miRNA-21 and -155 were found to be up-regulated in high-grade tumours but not in TC/AC, and miRNA-21 was found to be significantly up-regulated in metastatic low-grade NETs compared with non-metastatic tumours (Lee et al. 2012). These results require validation but suggest that miRNA profiles may be utilised as diagnostic and prognostic biomarkers.

Table 2 summarises the current state of knowledge of the role of miRNA in NET. Interestingly, miRNA-155 has been identified as a biomarker of both pancreatic (down-regulated) and bronchial NETs (up-regulated), tumours of foregut embryological origin. A definitive role for miRNA involvement in NET development and signalling pathways still needs to be determined and further validation studies were performed with larger sample sets.

Table 2

Summary of miRNA associated with neuroendocrine tumours

Association
miRNA Pancreatic NET GI NET Bronchial NET Reference
miR-204: up-regulated Associated with insulinoma Roldo et al. (2006)
miR-103, -107: up-regulated Distinguishes pNET from normal Roldo et al. (2006)
miR-155: down-regulated
miR-10b, -133a, -145, -146, -122: down-regulated Associated with metastatic ileal NET Ruebel et al. (2010)
miR-183, -488, -19a+b: up-regulated
miR-31, -129-5p, -133a, -125: down-regulated Associated with metastatic small intestinal NET Li et al. (2012)
miR-96, -182, -183, -196a, 200a: up-regulated
miR-653, -7, -489, 1224-5p: up-regulated Associated with colorectal NET (vs normal mucosa) Hamfjord et al. (2012)
miR-21, -155: up-regulated Associated with high-grade NET Lee et al. (2012)

NET, neuroendocrine tumour; miRNA, microRNA.

Rarer NETs

There is limited research into the impact of epigenetic changes on rare NET subtypes, and there are no published reports of any epigenetic research performed on thymic NETs.

Hypermethylation of RASSF1A and p14ARF promoter regions is frequent in Merkel cell carcinomas

The pathogenesis of Merkel cell carcinomas (MCCs) is poorly understood. Two studies have been performed investigating epigenetics in MCC, both candidate-driven analyses of promoter methylation, which demonstrated promoter hypermethylation of RASSF1A (51%), p14ARF (42%) and CDKN2A/p16INK4a (5–22%) (Lassacher et al. 2008, Helmbold et al. 2009). The Merkel cell polyomavirus has recently been demonstrated to encode and express an miRNA MCV-miR-M1-5p in 50% of MCCs, which may play a pathogenic role in tumour development (Lee et al. 2011). No correlation was demonstrated between polyomavirus infection and methylation status (Helmbold et al. 2009).

Promoter hypermethylation of CDKN2A/p16INK4a is prognostic in paragangliomas

Promoter hypermethylation of CDKN2A/p16INK4a is present in 100% of paragangliomas with succinate dehydrogenase complex subunit B (SDHB) mutation and is associated with metastasis and poorer prognosis. MEN2/RET-associated paragangliomas in contrast did not exhibit significant hypermethylation (Kiss et al. 2008, 2013). CIMP positivity is present in 9% of paragangliomas and is associated with malignant behaviour (Geli et al. 2008).

Global hypomethylation and IGF2 promoter hypermethylation are associated with malignant behaviour of adrenocortical tumours

Malignant adrenocortical tumours (ACTs) are globally hypomethylated relative to benign tumours (which shared most of their methylation patterns with normal tissue). Two groups have analysed hypermethylation in ACT identifying 212 hypermethylated CpG islands and 52 hypermethylated promoter loci respectively. Genome wide profiling of ACT can distinguish methylation patterns of normal and malignant tumours, specifically hypermethylation of CDKN2A and GATA4 is present in ACT but not in normal tissue, while IGF2 promoter hypermethylation is identified in malignant ACT but not in normal tissue or benign tumours (Fonseca et al. 2012, Rechache et al. 2012). CIMP positivity in ACT has been associated with poor survival (Barreau et al. 2013). Treatment of an ACT cell line (NCI-H295R) with decitabine restored expression of hypermethylated genes and has anti-proliferative effects (Suh et al. 2010, Fonseca et al. 2012).

miRNA profiling distinguishes malignant and benign ACTs

Several investigations of the role of miRNA in ACTs have been performed in recent years and have demonstrated association of malignant ACT with the following expression patterns: down-regulation of miR-100, -125b and -195 and up-regulation of miR-483-5p (Patterson et al. 2011); up-regulation of miR-335 and -675 (Schmitz et al. 2011); up-regulation of miR-195 and -483-5p (Soon et al. 2009); up-regulation of miR-184 and -503 and down-regulation of miR-511 and -214 (Tombol et al. 2009); up-regulation of miR-503, -1202 and -1275 (Ozata et al. 2011). Inhibition of miR-483-3p or -483-5p and overexpression of miR-195 or -497 reduced cell proliferation in human NCI-H295R ACT cells (Ozata et al. 2011). A comprehensive review of dysregulation of miRNA in ACTs has recently been published (Singh et al. 2012).

11p15.5 imprinted genes may be pathogenic in phaeochromocytoma

Loss of imprinting of the 11p15.5 allele (which contains IGF2) has been identified in epigenetic analysis of phaeochromocytomas (both sporadic and VHL associated) (Astuti et al. 2005, Margetts et al. 2005). Other promoter regions found to be aberrantly methylated in phaeochromocytoma include HIC1 (82%) and CASP8 (31%) (Margetts et al. 2005); HSP47 (SERPINH1) (52%), HOXA9 (17%) and OPCML (12%)(Margetts et al. 2008). Methylation patterns were found to be similar in neuroblastoma tumours (Margetts et al. 2005). Promoter regions of VHL, SDHB and SDHD are demonstrably unmethylated in sporadic phaeochromocytomas (Cascon et al. 2004).

miRNA profiling can identify malignant and recurring phaeochromocytomas

Three comparative analyses of benign and malignant phaeochromocytoma miRNA profiles have been performed in recent years. Overexpression of miR-483-5p, -183 and -101 (Patterson et al. 2012); overexpression of miR-483-5p and underexpression of miR-15a and -16 (Meyer-Rochow et al. 2010); and overexpression of miR-1225-3p (Tombol et al. 2010) have been identified as markers of malignant or recurring tumours. MiR-483-5p has been identified in investigation of both phaeochromocytomas and ACTs and is an interesting candidate for further investigation given its location at 11p15.5 within the second intron of IGF2 and implication in other solid malignant tumours (Meyer-Rochow et al. 2010).

Therapeutic approaches using epigenetic targets

The use of novel targeted epigenetic therapies is an attractive concept due to the prevalence of modifications found in malignant tumours, and the inherent reversibility of DNA methylation and chromatin modifications. Despite this, epigenetic therapies have so far shown limited efficacy in solid tumours; however, preclinical and early clinical trials in NET are promising.

DNMT inhibitors suppress NET cell growth

The DNMT inhibitor azacytidine has been shown to cause a dose-dependent reduction in tumour cell proliferation in three NET cell lines (midgut: CDNT2.5, bronchial: NCI-H727 and pancreatic lymph node metastasis: BON1). Growth inhibition was associated by significant reduction in chromogranin A and neuron-specific enolase (Alexander et al. 2010). Two bronchial NET cell lines (NCI-H727 bronchial ‘carcinoid’ and HTB-119 SCLC) showed re-expression of RASSF1A mRNA after treatment with decitabine (Zhang et al. 2006), while treatment of QGP1 pNET cell lines with decitabine restored CDKN2A/p16INK4a and caused significant growth inhibition (Habbe et al. 2007). The anti-proliferative effect is likely to be due to restoration of multiple genes silenced by pathogenic de novo methylation. Differential expression of 48 genes has been identified in QGP1 cells after treatment with decitabine, including 23 in which expression decreased after treatment. Many of these genes are known to be involved in cellular proliferation, apoptosis and development of metastasis and may play a role in pNET development (Habbe et al. 2007).

HDACis suppress NET cell growth

Functional analysis of three different HDAC inhibitors – trichostatin A, sodium butyrate and entinostat – has been carried out on neuroendocrine cell lines (insulinoma: CM and pancreatic lymph node metastasis: BON1) showing dose-dependent inhibition of proliferation, cell cycle arrest and induction of apoptosis. The addition of somatostatin or octreotide did not affect the outcome (Baradari et al. 2006).

Valproic acid (VPA) (a class I and IIa HDAC inhibitor) in combination with lithium chloride has been shown to suppress chromogranin A levels while reducing cellular growth in pancreatic (BON) and pulmonary (NCI-H727) NET cell lines (Adler et al. 2009), while VPA alone up-regulates notch1 and suppresses growth in gastrointestinal and bronchial NET cell lines (Greenblatt et al. 2007). Notch1 is a transmembrane receptor that translocates to the nucleus upon ligand binding and regulates gene transcription, acting as a tumour suppressor in some human cancers (Radtke & Raj 2003). Activation of Notch1 has been shown to suppress growth in NET cell lines and is associated with reduction of NET tumour markers (Kunnimalaiyaan et al. 2006).

Early phase clinical trials of HDACis in NET are inconclusive

A pilot phase II trial of VPA in low-grade NET was conducted in eight patients (two pNETs and six midgut NETs) receiving 500 mg oral VPA twice per day with dose adjustment to maintain therapeutic serum levels. Notch1 signalling was absent in all tumours before treatment and was up-regulated with VPA. One patient had an unconfirmed partial response and four had stable disease, tumour markers improved in five out of seven (Mohammed et al. 2011). This well-tolerated agent may have a role in the management of low-grade NETs and is currently being trialled in many different tumour types.

Two trials of HDAC inhibitors have been terminated early: in 2006, depsipeptide was associated with an unexpectedly high rate of cardiac adverse events and a phase II trial in NET patients was terminated (Shah et al. 2006); and in 2012, a phase II trial panobinostat in a mixed cohort of 15 GEP NET patients was terminated due to lack of apparent benefit. In this single-arm trial, all patients received panobinostat 20 mg once daily three times per week. The stable disease rate was 92.3% and median progression-free survival was 11.8 months; however, response rate was 0% and stabilisation rate needs to be considered in context of the typical slow growth of low-grade NET. Thrombocytopaenia and fatigue were the most frequent toxicities (Rajguru et al. 2012).

While in vitro results of epigenetic therapies on NET cell lines appear promising, clinical trials have so far not demonstrated significant benefit to patients but are limited by the rarity of NET and challenges of performing early phase trials in this patient group. Demethylating agents have not yet been trialled in the NET population and the field of miRNA therapeutics is still in its infancy. Predictive biomarkers are required to assist patient stratification for this novel class of therapeutics, and as specificity of the agents to target a particular epigenetic change improves so, toxicity is likely to reduce and patient benefit increase.

Summary

Epigenetic changes are likely to play a key role in NET development and progress is being made in identifying potential diagnostic and prognostic epigenetic biomarkers for these tumours. The apparent disparity in epigenetic states of NETs of different cellular origins is intriguing and is likely to reflect diverse tumorigenic processes. Increased promoter methylation of CDKN2A/P16INK4a is a typical feature of gastrinomas, while methylation of IGF2 and MLH1 appears to be characteristic of insulinoma. This has implications for targeted therapeutics and personalisation of patient management and needs to be further explored in large-scale studies. Methylation of RASSF1A is a frequent finding in NETs of all origins and has prognostic impact, suggesting a potential ‘driver’ role in development of these tumours and a potentially effective drug target. CIMP positivity is associated with worse prognosis in pNETs but improved prognosis in colorectal NET. EZH2 expression distinguishes between high- and low-grade bronchial NETs, as do histone modifications H4KA16 and K4KM20. Characteristic miRNA signatures of NET of all subtypes are being identified that distinguish tumour from normal tissue, as well as some with prognostic significance.

A key development in our understanding of pNET has been the identification of ATRX/DAXX mutations in 43% of tumours and the associated ALT phenotype (Jiao et al. 2011). These are associated with improved prognosis in pNET and are suitable for clinical application as a prognostic biomarker. The identification of these mutations and the subsequent chromatin remodelling illustrate the synergistic nature of genetic and epigenetic regulation.

The field of epigenetic-targeted agents is still in development, and despite promising in vitro results in many solid tumours the only real clinical applications with FDA approval so far have come in haematological malignancies. However, there is enormous interest and investment, and with many novel agents entering clinical trials, and increasing personalisation/targeting of therapy, we anticipate that positive results in solid tumours will soon follow.

We have presented a comprehensive overview of current understanding of epigenetic changes in NETs. In many cases, the data presented is identified through small candidate-driven studies of mixed cohorts of NET cases; in consequence, some outcomes appear conflicting and drawing decisive conclusions is difficult. There are multiple mechanisms regulating gene transcription and cellular proliferation in NET and identifying the most powerful tumorigenic drivers within the group is challenging. Commonly mutated oncogenes in other solid tumours play little or no pathogenic role in NET and therefore epigenetic changes are good candidates for pathogenic drivers. The findings presented in this review give insight into the pathobiology of NET and provide rationale for the application of novel therapeutics and further avenues of research. There is clear requirement for further collaborative large-scale genome wide integrated (epi)genetic studies, the results of which will clarify what is currently a complex field of research.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the review.

Funding

This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector. All authors are in receipt of research grants from Cancer Research UK and The Raymond and Beverly Sackler Foundation.

Author contribution statement

All authors made significant contributions to writing the manuscript.

References

  • Adler JT , Hottinger DG , Kunnimalaiyaan M & Chen H 2009 Combination therapy with histone deacetylase inhibitors and lithium chloride: a novel treatment for carcinoid tumors. Annals of Surgical Oncology 16 481486. (doi:10.1245/s10434-008-0194-6).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Agarwal SK , Guru SC , Heppner C , Erdos MR , Collins RM , Park SY , Saggar S , Chandrasekharappa SC , Collins FS & Spiegel AM et al. 1999 Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell 96 143152. (doi:10.1016/S0092-8674(00)80967-8).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Alexander VM , Roy M , Steffens KA , Kunnimalaiyaan M & Chen H 2010 Azacytidine induces cell cycle arrest and suppression of neuroendocrine markers in carcinoids. International Journal of Clinical and Experimental Medicine 3 95102.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Allfrey VG , Faulkner R & Mirsky AE 1964 Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. PNAS 51 786794. (doi:10.1073/pnas.51.5.786).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arnold CN , Sosnowski A , Schmitt-Graff A , Arnold R & Blum HE 2007 Analysis of molecular pathways in sporadic neuroendocrine tumors of the gastro-entero-pancreatic system. International Journal of Cancer 120 21572164. (doi:10.1002/ijc.22569).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arnold CN , Nagasaka T , Goel A , Scharf I , Grabowski P , Sosnowski A , Schmitt-Graff A , Boland CR , Arnold R & Blum HE 2008 Molecular characteristics and predictors of survival in patients with malignant neuroendocrine tumors. International Journal of Cancer 123 15561564. (doi:10.1002/ijc.23690).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Astuti D , Latif F , Wagner K , Gentle D , Cooper WN , Catchpoole D , Grundy R , Ferguson-Smith AC & Maher ER 2005 Epigenetic alteration at the DLK1-GTL2 imprinted domain in human neoplasia: analysis of neuroblastoma, phaeochromocytoma and Wilms' tumour. British Journal of Cancer 92 15741580. (doi:10.1038/sj.bjc.6602478).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bachmann IM , Halvorsen OJ , Collett K , Stefansson IM , Straume O , Haukaas SA , Salvesen HB , Otte AP & Akslen LA 2006 EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. Journal of Clinical Oncology 24 268273. (doi:10.1200/JCO.2005.01.5180).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baradari V , Huether A , Hopfner M , Schuppan D & Scherubl H 2006 Antiproliferative and proapoptotic effects of histone deacetylase inhibitors on gastrointestinal neuroendocrine tumor cells. Endocrine-Related Cancer 13 12371250. (doi:10.1677/erc.1.01249).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barghorn A , Komminoth P , Bachmann D , Rutimann K , Saremaslani P , Muletta-Feurer S , Perren A , Roth J , Heitz PU & Speel EJ 2001 Deletion at 3p25.3-p23 is frequently encountered in endocrine pancreatic tumours and is associated with metastatic progression. Journal of Pathology 194 451458. (doi:10.1002/path.886).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barlesi F , Giaccone G , Gallegos-Ruiz MI , Loundou A , Span SW , Lefesvre P , Kruyt FA & Rodriguez JA 2007 Global histone modifications predict prognosis of resected non small-cell lung cancer. Journal of Clinical Oncology 25 43584364. (doi:10.1200/JCO.2007.11.2599).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barreau O , Assie G , Wilmot-Roussel H , Ragazzon B , Baudry C , Perlemoine K , Rene-Corail F , Bertagna X , Dousset B & Hamzaoui N et al. 2013 Identification of a CpG island methylator phenotype in adrenocortical carcinomas. Journal of Clinical Endocrinology and Metabolism 98 E174E184. (doi:10.1210/jc.2012-2993).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartsch DK , Kersting M , Wild A , Ramaswamy A , Gerdes B , Schuermann M , Simon B & Rothmund M 2000 Low frequency of p16(INK4a) alterations in insulinomas. Digestion 62 171177. (doi:10.1159/000007810).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bauer W , Briner U , Doepfner W , Haller R , Huguenin R , Marbach P , Petcher TJ & Pless 1982 SMS 201-995: a very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sciences 31 11331140. (doi:10.1016/0024-3205(82)90087-X).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bernstein BE , Stamatoyannopoulos JA , Costello JF , Ren B , Milosavljevic A , Meissner A , Kellis M , Marra MA , Beaudet AL & Ecker JR et al. 2010 The NIH roadmap epigenomics mapping consortium. Nature Biotechnology 28 10451048. (doi:10.1038/nbt1010-1045).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bestor TH 1992 Activation of mammalian DNA methyltransferase by cleavage of a Zn binding regulatory domain. EMBO Journal 11 26112617.

  • Bruns C , Lewis I , Briner U , Meno-Tetang G & Weckbecker G 2002 SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. European Journal of Endocrinology/European Federation of Endocrine Societies 146 707716. (doi:10.1530/eje.0.1460707).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cairns P , Esteller M , Herman JG , Schoenberg M , Jeronimo C , Sanchez-Cespedes M , Chow NH , Grasso M , Wu L & Westra WB et al. 2001 Molecular detection of prostate cancer in urine by GSTP1 hypermethylation. Clinical Cancer Research 7 27272730.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Calin GA , Sevignani C , Dumitru CD , Hyslop T , Noch E , Yendamuri S , Shimizu M , Rattan S , Bullrich F & Negrini M et al. 2004 Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. PNAS 101 29993004. (doi:10.1073/pnas.0307323101).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cascon A , Ruiz-Llorente S , Fraga MF , Leton R , Telleria D , Sastre J , Diez JJ , Martinez Diaz-Guerra G , Diaz Perez JA & Benitez J et al. 2004 Genetic and epigenetic profile of sporadic pheochromocytomas. Journal of Medical Genetics 41 e30. (doi:10.1136/jmg.2003.012658).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Catteau A , Harris WH , Xu CF & Solomon E 1999 Methylation of the BRCA1 promoter region in sporadic breast and ovarian cancer: correlation with disease characteristics. Oncogene 18 19571965. (doi:10.1038/sj.onc.1202509).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chadwick LH 2012 The NIH Roadmap Epigenomics Program data resource. Epigenomics 4 317324. (doi:10.2217/epi.12.18).

  • Chan AO , Kim SG , Bedeir A , Issa JP , Hamilton SR & Rashid A 2003 CpG island methylation in carcinoid and pancreatic endocrine tumors. Oncogene 22 924934. (doi:10.1038/sj.onc.1206123).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chaussade L , Eymin B , Brambilla E & Gazzeri S 2001 Expression of p15 and p15.5 products in neuroendocrine lung tumours: relationship with p15(INK4b) methylation status. Oncogene 20 65876596. (doi:10.1038/sj.onc.1204798).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chi AS & Bernstein BE 2009 Developmental biology. Pluripotent chromatin state. Science 323 220221. (doi:10.1126/science.1166261).

  • Choi IS , Estecio MR , Nagano Y , Kim do H , White JA , Yao JC , Issa JP & Rashid A 2007 Hypomethylation of LINE-1 and Alu in well-differentiated neuroendocrine tumors (pancreatic endocrine tumors and carcinoid tumors). Modern Pathology 20 802810. (doi:10.1038/modpathol.3800825).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chung DC , Smith AP , Louis DN , Graeme-Cook F , Warshaw AL & Arnold A 1997a Analysis of the retinoblastoma tumour suppressor gene in pancreatic endocrine tumours. Clinical Endocrinology 47 523528. (doi:10.1046/j.1365-2265.1997.2861110.x).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chung DC , Smith AP , Louis DN , Graeme-Cook F , Warshaw AL & Arnold A 1997b A novel pancreatic endocrine tumor suppressor gene locus on chromosome 3p with clinical prognostic implications. Journal of Clinical Investigation 100 404410. (doi:10.1172/JCI119547).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clark SJ , Harrison J , Paul CL & Frommer M 1994 High sensitivity mapping of methylated cytosines. Nucleic Acids Research 22 29902997. (doi:10.1093/nar/22.15.2990).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dammann R , Li C , Yoon JH , Chin PL , Bates S & Pfeifer GP 2000 Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nature Genetics 25 315319. (doi:10.1038/77083).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dammann R , Schagdarsurengin U , Liu L , Otto N , Gimm O , Dralle H , Boehm BO , Pfeifer GP & Hoang-Vu C 2003a Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene 22 38063812. (doi:10.1038/sj.onc.1206582).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dammann R , Schagdarsurengin U , Strunnikova M , Rastetter M , Seidel C , Liu L , Tommasi S & Pfeifer GP 2003b Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histology and Histopathology 18 665677.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dejeux E , Olaso R , Dousset B , Audebourg A , Gut IG , Terris B & Tost J 2009 Hypermethylation of the IGF2 differentially methylated region 2 is a specific event in insulinomas leading to loss-of-imprinting and overexpression. Endocrine-Related Cancer 16 939952. (doi:10.1677/ERC-08-0331).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eckhardt F , Beck S , Gut IG & Berlin K 2004 Future potential of the Human Epigenome Project. Expert Review of Molecular Diagnostics 4 609618. (doi:10.1586/14737159.4.5.609).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eden A , Gaudet F , Waghmare A & Jaenisch R 2003 Chromosomal instability and tumors promoted by DNA hypomethylation. Science 300 455. (doi:10.1126/science.1083557).

  • Estrabaud E , Lassot I , Blot G , Le Rouzic E , Tanchou V , Quemeneur E , Daviet L , Margottin-Goguet F & Benarous R 2007 RASSF1C, an isoform of the tumor suppressor RASSF1A, promotes the accumulation of β-catenin by interacting with βTrCP. Cancer Research 67 10541061. (doi:10.1158/0008-5472.CAN-06-2530).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Feil R & Fraga MF 2011 Epigenetics and the environment: emerging patterns and implications. Nature Reviews. Genetics 13 97109.

  • Feinberg AP & Vogelstein B 1983 Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 301 8992. (doi:10.1038/301089a0).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Findeis-Hosey JJ , Huang J , Li F , Yang Q , McMahon LA & Xu H 2011 High-grade neuroendocrine carcinomas of the lung highly express enhancer of zeste homolog 2, but carcinoids do not. Human Pathology 42 867872. (doi:10.1016/j.humpath.2010.09.019).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fonseca AL , Kugelberg J , Starker LF , Scholl U , Choi M , Hellman P , Akerstrom G , Westin G , Lifton RP & Bjorklund P et al. 2012 Comprehensive DNA methylation analysis of benign and malignant adrenocortical tumors. Genes, Chromosomes & Cancer 51 949960. (doi:10.1002/gcc.21978).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fontaniere S , Tost J , Wierinckx A , Lachuer J , Lu J , Hussein N , Busato F , Gut I , Wang ZQ & Zhang CX 2006 Gene expression profiling in insulinomas of Men1 β-cell mutant mice reveals early genetic and epigenetic events involved in pancreatic β-cell tumorigenesis. Endocrine-Related Cancer 13 12231236. (doi:10.1677/erc.1.01294).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fraga MF , Herranz M , Espada J , Ballestar E , Paz MF , Ropero S , Erkek E , Bozdogan O , Peinado H & Niveleau A et al. 2004 A mouse skin multistage carcinogenesis model reflects the aberrant DNA methylation patterns of human tumors. Cancer Research 64 55275534. (doi:10.1158/0008-5472.CAN-03-4061).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fraga MF , Ballestar E , Villar-Garea A , Boix-Chornet M , Espada J , Schotta G , Bonaldi T , Haydon C , Ropero S & Petrie K et al. 2005 Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nature Genetics 37 391400. (doi:10.1038/ng1531).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Friedman OM , Mahapatra GN & Stevenson R 1963 The methylation of deoxyribonucleosides by diazomethane. Biochimica et Biophysica Acta 68 144146. (doi:10.1016/0926-6550(63)90422-5).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frolich JC , Bloomgarden ZT , Oates JA , McGuigan JE & Rabinowitz D 1978 The carcinoid flush. Provocation by pentagastrin and inhibition by somatostatin. New England Journal of Medicine 299 10551057. (doi:10.1056/NEJM197811092991908).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fu S , Hu W , Iyer R , Kavanagh JJ , Coleman RL , Levenback CF , Sood AK , Wolf JK , Gershenson DM & Markman M et al. 2011 Phase 1b-2a study to reverse platinum resistance through use of a hypomethylating agent, azacitidine, in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Cancer 117 16611669. (doi:10.1002/cncr.25701).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Geli J , Kiss N , Karimi M , Lee JJ , Backdahl M , Ekstrom TJ & Larsson C 2008 Global and regional CpG methylation in pheochromocytomas and abdominal paragangliomas: association to malignant behavior. Clinical Cancer Research 14 25512559. (doi:10.1158/1078-0432.CCR-07-1867).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greenblatt DY , Vaccaro AM , Jaskula-Sztul R , Ning L , Haymart M , Kunnimalaiyaan M & Chen H 2007 Valproic acid activates notch-1 signaling and regulates the neuroendocrine phenotype in carcinoid cancer cells. Oncologist 12 942951. (doi:10.1634/theoncologist.12-8-942).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greger V , Passarge E , Hopping W , Messmer E & Horsthemke B 1989 Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Human Genetics 83 155158. (doi:10.1007/BF00286709).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Habbe N , Bert T & Simon B 2007 Identification of methylation-associated gene expression in neuroendocrine pancreatic tumor cells. Pancreatology 7 352359. (doi:10.1159/000107270).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamfjord J , Stangeland AM , Hughes T , Skrede ML , Tveit KM , Ikdahl T & Kure EH 2012 Differential expression of miRNAs in colorectal cancer: comparison of paired tumor tissue and adjacent normal mucosa using high-throughput sequencing. PLoS ONE 7 e34150. (doi:10.1371/journal.pone.0034150).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heaphy CM , de Wilde RF , Jiao Y , Klein AP , Edil BH , Shi C , Bettegowda C , Rodriguez FJ , Eberhart CG & Hebbar S et al. 2011a Altered telomeres in tumors with ATRX and DAXX mutations. Science 333 425. (doi:10.1126/science.1207313).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heaphy CM , Subhawong AP , Hong SM , Goggins MG , Montgomery EA , Gabrielson E , Netto GJ , Epstein JI , Lotan TL & Westra WH et al. 2011b Prevalence of the alternative lengthening of telomeres telomere maintenance mechanism in human cancer subtypes. American Journal of Pathology 179 16081615. (doi:10.1016/j.ajpath.2011.06.018).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hegi ME , Diserens AC , Gorlia T , Hamou MF , de Tribolet N , Weller M , Kros JM , Hainfellner JA , Mason W & Mariani L et al. 2005 MGMT gene silencing and benefit from temozolomide in glioblastoma. New England Journal of Medicine 352 9971003. (doi:10.1056/NEJMoa043331).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Helmbold P , Lahtz C , Enk A , Herrmann-Trost P , Marsch W , Kutzner H & Dammann RH 2009 Frequent occurrence of RASSF1A promoter hypermethylation and Merkel cell polyomavirus in Merkel cell carcinoma. Molecular Carcinogenesis 48 903909. (doi:10.1002/mc.20540).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herman JG , Latif F , Weng Y , Lerman MI , Zbar B , Liu S , Samid D , Duan DS , Gnarra JR & Linehan WM et al. 1994 Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. PNAS 91 97009704. (doi:10.1073/pnas.91.21.9700).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herman JG , Merlo A , Mao L , Lapidus RG , Issa JP , Davidson NE , Sidransky D & Baylin SB 1995 Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Research 55 45254530.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • House MG , Herman JG , Guo MZ , Hooker CM , Schulick RD , Lillemoe KD , Cameron JL , Hruban RH , Maitra A & Yeo CJ 2003 Aberrant hypermethylation of tumor suppressor genes in pancreatic endocrine neoplasms. Annals of Surgery 238 423431.(discussion 431-422)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jenuwein T & Allis CD 2001 Translating the histone code. Science 293 10741080. (doi:10.1126/science.1063127).

  • Jiao Y , Shi C , Edil BH , de Wilde RF , Klimstra DS , Maitra A , Schulick RD , Tang LH , Wolfgang CL & Choti MA et al. 2011 DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331 11991203. (doi:10.1126/science.1200609).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jones PA & Baylin SB 2002 The fundamental role of epigenetic events in cancer. Nature Reviews. Genetics 3 415428. (doi:10.1038/nrg962).

  • Jones PA & Martienssen R 2005 A blueprint for a Human Epigenome Project: the AACR Human Epigenome Workshop. Cancer Research 65 1124111246. (doi:10.1158/0008-5472.CAN-05-3865).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaminskas E , Farrell A , Abraham S , Baird A , Hsieh LS , Lee SL , Leighton JK , Patel H , Rahman A & Sridhara R et al. 2005 Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clinical Cancer Research 11 36043608. (doi:10.1158/1078-0432.CCR-04-2135).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kiss NB , Geli J , Lundberg F , Avci C , Velazquez-Fernandez D , Hashemi J , Weber G , Hoog A , Ekstrom TJ & Backdahl M et al. 2008 Methylation of the p16INK4A promoter is associated with malignant behavior in abdominal extra-adrenal paragangliomas but not pheochromocytomas. Endocrine-Related Cancer 15 609621. (doi:10.1677/ERC-07-0285).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kiss NB , Muth A , Andreasson A , Juhlin CC , Geli J , Backdahl M , Hoog A , Wangberg B , Nilsson O & Ahlman H et al. 2013 Acquired hypermethylation of the P16INK4A promoter in abdominal paraganglioma: relation to adverse tumor phenotype and predisposing mutation. Endocrine-Related Cancer 20 6578. (doi:10.1530/ERC-12-0267).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Krenning EP , Bakker WH , Breeman WA , Koper JW , Kooij PP , Ausema L , Lameris JS , Reubi JC & Lamberts SW 1989 Localisation of endocrine-related tumours with radioiodinated analogue of somatostatin. Lancet 1 242244. (doi:10.1016/S0140-6736(89)91258-0).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Krenning EP , de Jong M , Kooij PP , Breeman WA , Bakker WH , de Herder WW , van Eijck CH , Kwekkeboom DJ , Jamar F & Pauwels S et al. 1999 Radiolabelled somatostatin analogue(s) for peptide receptor scintigraphy and radionuclide therapy. Annals of Oncology 10 (Suppl 2) S23S29. (doi:10.1093/annonc/10.suppl_2.S23).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kunnimalaiyaan M , Vaccaro AM , Ndiaye MA & Chen H 2006 Overexpression of the NOTCH1 intracellular domain inhibits cell proliferation and alters the neuroendocrine phenotype of medullary thyroid cancer cells. Journal of Biological Chemistry 281 3981939830. (doi:10.1074/jbc.M603578200).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lagos-Quintana M , Rauhut R , Lendeckel W & Tuschl T 2001 Identification of novel genes coding for small expressed RNAs. Science 294 853858. (doi:10.1126/science.1064921).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • LaRosa S , Marando A , Furlan D , Sahnane N & Capella C 2012 Colorectal poorly differentiated neuroendocrine carcinomas and mixed adenoneuroendocrine carcinomas: insights into the diagnostic immunophenotype, assessment of methylation profile, and search for prognostic markers. American Journal of Surgical Pathology 36 601611. (doi:10.1097/PAS.0b013e318242e21c).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lassacher A , Heitzer E , Kerl H & Wolf P 2008 p14ARF hypermethylation is common but INK4a-ARF locus or p53 mutations are rare in Merkel cell carcinoma. Journal of Investigative Dermatology 128 17881796. (doi:10.1038/sj.jid.5701256).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee S , Paulson KG , Murchison EP , Afanasiev OK , Alkan C , Leonard JH , Byrd DR , Hannon GJ & Nghiem P 2011 Identification and validation of a novel mature microRNA encoded by the Merkel cell polyomavirus in human Merkel cell carcinomas. Journal of Clinical Virology 52 272275. (doi:10.1016/j.jcv.2011.08.012).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee HW , Lee EH , Ha SY , Lee CH , Chang HK , Chang S , Kwon KY , Hwang IS , Roh MS & Seo JW 2012 Altered expression of microRNA miR-21, miR-155, and let-7a and their roles in pulmonary neuroendocrine tumors. Pathology International 62 583591. (doi:10.1111/j.1440-1827.2012.02845.x).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lembeck F 1954 Detection of 5-hydroxytryptamine in carcinoid metastases. Naunyn-Schmiedebergs Archiv fur experimentelle Pathologie und Pharmakologie 221 5066.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li F , Ye B , Hong L , Xu H & Fishbein MC 2011 Epigenetic modifications of histone h4 in lung neuroendocrine tumors. Applied Immunohistochemistry & Molecular Morphology 19 389394. (doi:10.1097/PAI.0b013e3182108e2e).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li SC , Martinjn C , Essaghir A , Lloyd RV , Demoulin J , Oberg K & Giandomenico V 2012 Role of microRNAs in small intestinal neuroendocrine tumours. Neuroendocrinology 96 172.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lister R , O'Malley RC , Tonti-Filippini J , Gregory BD , Berry CC , Millar AH & Ecker JR 2008 Highly integrated single-base resolution maps of the epigenome in Arabidopsis. Cell 133 523536. (doi:10.1016/j.cell.2008.03.029).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu L , Broaddus RR , Yao JC , Xie S , White JA , Wu TT , Hamilton SR & Rashid A 2005 Epigenetic alterations in neuroendocrine tumors: methylation of RAS-association domain family 1, isoform A and p16 genes are associated with metastasis. Modern Pathology 18 16321640.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu R , Liao J , Yang M , Shi Y , Peng Y , Wang Y , Pan E , Guo W , Pu Y & Yin L 2012 Circulating miR-155 expression in plasma: a potential biomarker for early diagnosis of esophageal cancer in humans. Journal of Toxicology and Environmental Health. Part A 75 11541162. (doi:10.1080/15287394.2012.699856).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu J , Getz G , Miska EA , Alvarez-Saavedra E , Lamb J , Peck D , Sweet-Cordero A , Ebert BL , Mak RH & Ferrando AA et al. 2005 MicroRNA expression profiles classify human cancers. Nature 435 834838. (doi:10.1038/nature03702).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lubomierski N , Kersting M , Bert T , Muench K , Wulbrand U , Schuermann M , Bartsch D & Simon B 2001 Tumor suppressor genes in the 9p21 gene cluster are selective targets of inactivation in neuroendocrine gastroenteropancreatic tumors. Cancer Research 61 59055910.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Magerl C , Ellinger J , Braunschweig T , Kremmer E , Koch LK , Holler T , Buttner R , Luscher B & Gutgemann I 2010 H3K4 dimethylation in hepatocellular carcinoma is rare compared with other hepatobiliary and gastrointestinal carcinomas and correlates with expression of the methylase Ash2 and the demethylase LSD1. Human Pathology 41 181189. (doi:10.1016/j.humpath.2009.08.007).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Malpeli G , Amato E , Dandrea M , Fumagalli C , Debattisti V , Boninsegna L , Pelosi G , Falconi M & Scarpa A 2011 Methylation-associated down-regulation of RASSF1A and up-regulation of RASSF1C in pancreatic endocrine tumors. BMC Cancer 11 351. (doi:10.1186/1471-2407-11-351).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mann BS , Johnson JR , Cohen MH , Justice R & Pazdur R 2007 FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12 12471252. (doi:10.1634/theoncologist.12-10-1247).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Margetts CD , Astuti D , Gentle DC , Cooper WN , Cascon A , Catchpoole D , Robledo M , Neumann HP , Latif F & Maher ER 2005 Epigenetic analysis of HIC1, CASP8, FLIP, TSP1, DCR1, DCR2, DR4, DR5, KvDMR1, H19 and preferential 11p15.5 maternal-allele loss in von Hippel–Lindau and sporadic phaeochromocytomas. Endocrine-Related Cancer 12 161172. (doi:10.1677/erc.1.00865).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Margetts CD , Morris M , Astuti D , Gentle DC , Cascon A , McRonald FE , Catchpoole D , Robledo M , Neumann HP & Latif F et al. 2008 Evaluation of a functional epigenetic approach to identify promoter region methylation in phaeochromocytoma and neuroblastoma. Endocrine-Related Cancer 15 777786. (doi:10.1677/ERC-08-0072).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matei D , Fang F , Shen C , Schilder J , Arnold A , Zeng Y , Berry WA , Huang T & Nephew KP 2012 Epigenetic resensitization to platinum in ovarian cancer. Cancer Research 72 21972205. (doi:10.1158/0008-5472.CAN-11-3909).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mei M , Deng D , Liu TH , Sang XT , Lu X , Xiang HD , Zhou J , Wu H , Yang Y & Chen J et al. 2009 Clinical implications of microsatellite instability and MLH1 gene inactivation in sporadic insulinomas. Journal of Clinical Endocrinology and Metabolism 94 34483457. (doi:10.1210/jc.2009-0173).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Merlo A , Herman JG , Mao L , Lee DJ , Gabrielson E , Burger PC , Baylin SB & Sidransky D 1995 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nature Medicine 1 686692. (doi:10.1038/nm0795-686).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Meyer-Rochow GY , Jackson NE , Conaglen JV , Whittle DE , Kunnimalaiyaan M , Chen H , Westin G , Sandgren J , Stalberg P & Khanafshar E et al. 2010 MicroRNA profiling of benign and malignant pheochromocytomas identifies novel diagnostic and therapeutic targets. Endocrine-Related Cancer 17 835846. (doi:10.1677/ERC-10-0142).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Modlin IM , Shapiro MD & Kidd M 2004 Siegfried Oberndorfer: origins and perspectives of carcinoid tumors. Human Pathology 35 14401451. (doi:10.1016/j.humpath.2004.09.018).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moertel CG , Hanley JA & Johnson LA 1980 Streptozocin alone compared with streptozocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. New England Journal of Medicine 303 11891194. (doi:10.1056/NEJM198011203032101).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mohammed TA , Holen KD , Jaskula-Sztul R , Mulkerin D , Lubner SJ , Schelman WR , Eickhoff J , Chen H & Loconte NK 2011 A pilot phase II study of valproic acid for treatment of low-grade neuroendocrine carcinoma. Oncologist 16 835843. (doi:10.1634/theoncologist.2011-0031).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Muller CI , Ruter B , Koeffler HP & Lubbert M 2006 DNA hypermethylation of myeloid cells, a novel therapeutic target in MDS and AML. Current Pharmaceutical Biotechnology 7 315321. (doi:10.2174/138920106778521523).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Muscarella P , Melvin WS , Fisher WE , Foor J , Ellison EC , Herman JG , Schirmer WJ , Hitchcock CL , DeYoung BR & Weghorst CM 1998 Genetic alterations in gastrinomas and nonfunctioning pancreatic neuroendocrine tumors: an analysis of p16/MTS1 tumor suppressor gene inactivation. Cancer Research 58 237240.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Oberg K , Funa K & Alm G 1983 Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. New England Journal of Medicine 309 129133. (doi:10.1056/NEJM198307213090301).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • O'Connor DT & Deftos LJ 1986 Secretion of chromogranin A by peptide-producing endocrine neoplasms. New England Journal of Medicine 314 11451151. (doi:10.1056/NEJM198605013141803).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Okano M , Xie S & Li E 1998a Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nature Genetics 19 219220. (doi:10.1038/890).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Okano M , Xie S & Li E 1998b Dnmt2 is not required for de novo and maintenance methylation of viral DNA in embryonic stem cells. Nucleic Acids Research 26 25362540. (doi:10.1093/nar/26.11.2536).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Okano M , Bell DW , Haber DA & Li E 1999 DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99 247257. (doi:10.1016/S0092-8674(00)81656-6).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Oki Y , Aoki E & Issa JP 2007 Decitabine – bedside to bench. Critical Reviews in Oncology/Hematology 61 140152. (doi:10.1016/j.critrevonc.2006.07.010).

  • Ozata DM , Caramuta S , Velazquez-Fernandez D , Akcakaya P , Xie H , Hoog A , Zedenius J , Backdahl M , Larsson C & Lui WO 2011 The role of microRNA deregulation in the pathogenesis of adrenocortical carcinoma. Endocrine-Related Cancer 18 643655. (doi:10.1530/ERC-11-0082).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Patterson EE , Holloway AK , Weng J , Fojo T & Kebebew E 2011 MicroRNA profiling of adrenocortical tumors reveals miR-483 as a marker of malignancy. Cancer 117 16301639. (doi:10.1002/cncr.25724).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Patterson E , Webb R , Weisbrod A , Bian B , He M , Zhang L , Holloway AK , Krishna R , Nilubol N & Pacak K et al. 2012 The microRNA expression changes associated with malignancy and SDHB mutation in pheochromocytoma. Endocrine-Related Cancer 19 157166. (doi:10.1530/ERC-11-0308).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pelosi G , Fumagalli C , Trubia M , Sonzogni A , Rekhtman N , Maisonneuve P , Galetta D , Spaggiari L , Veronesi G & Scarpa A et al. 2010 Dual role of RASSF1 as a tumor suppressor and an oncogene in neuroendocrine tumors of the lung. Anticancer Research 30 42694281.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pernow B & Waldenstrom J 1954 Paroxysmal flushing and other symptoms caused by 5-hydroxytryptamine and histamine in patients with malignant tumours. Lancet 267 951. (doi:10.1016/S0140-6736(54)92559-3).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pizzi S , Azzoni C , Bottarelli L , Campanini N , D'Adda T , Pasquali C , Rossi G , Rindi G & Bordi C 2005 RASSF1A promoter methylation and 3p21.3 loss of heterozygosity are features of foregut, but not midgut and hindgut, malignant endocrine tumours. Journal of Pathology 206 409416. (doi:10.1002/path.1784).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Radtke F & Raj K 2003 The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nature Reviews. Cancer 3 756767. (doi:10.1038/nrc1186).

  • Rahman MM , Qian ZR , Wang EL , Yoshimoto K , Nakasono M , Sultana R , Yoshida T , Hayashi T , Haba R & Ishida M et al. 2010 DNA methyltransferases 1, 3a, and 3b overexpression and clinical significance in gastroenteropancreatic neuroendocrine tumors. Human Pathology 41 10691078. (doi:10.1016/j.humpath.2010.01.011).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rajguru S , Lubner SJ , Mulkerin D , Schelman WR , Winterle N , Holen KD , Leverson G , Chen H & LoConte NK 2012 A phase II study of the histone deacetylase inhibitor panobinostat (LBH589) in low-grade neuroendocrine tumors. ASCO Meeting Abstracts 30 e14554.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Raymond E , Dahan L , Raoul JL , Bang YJ , Borbath I , Lombard-Bohas C , Valle J , Metrakos P , Smith D & Vinik A et al. 2011 Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. New England Journal of Medicine 364 501513. (doi:10.1056/NEJMoa1003825).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rechache NS , Wang Y , Stevenson HS , Killian JK , Edelman DC , Merino M , Zhang L , Nilubol N , Stratakis CA & Meltzer PS et al. 2012 DNA methylation profiling identifies global methylation differences and markers of adrenocortical tumors. Journal of Clinical Endocrinology and Metabolism 97 E1004E1013. (doi:10.1210/jc.2011-3298).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rinke A , Muller HH , Schade-Brittinger C , Klose KJ , Barth P , Wied M , Mayer C , Aminossadati B , Pape UF & Blaker M et al. 2009 Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. Journal of Clinical Oncology 27 46564663. (doi:10.1200/JCO.2009.22.8510).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roldo C , Missiaglia E , Hagan JP , Falconi M , Capelli P , Bersani S , Calin GA , Volinia S , Liu CG & Scarpa A et al. 2006 MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. Journal of Clinical Oncology 24 46774684. (doi:10.1200/JCO.2005.05.5194).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ruebel K , Leontovich AA , Stilling GA , Zhang S , Righi A , Jin L & Lloyd RV 2010 MicroRNA expression in ileal carcinoid tumors: downregulation of microRNA-133a with tumor progression. Modern Pathology 23 367375. (doi:10.1038/modpathol.2009.161).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sakai T , Toguchida J , Ohtani N , Yandell DW , Rapaport JM & Dryja TP 1991 Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene. American Journal of Human Genetics 48 880888.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmitt AM , Schmid S , Rudolph T , Anlauf M , Prinz C , Kloppel G , Moch H , Heitz PU , Komminoth P & Perren A 2009 VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocrine-Related Cancer 16 12191227. (doi:10.1677/ERC-08-0297).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmitz KJ , Helwig J , Bertram S , Sheu SY , Suttorp AC , Seggewiss J , Willscher E , Walz MK , Worm K & Schmid KW 2011 Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. Journal of Clinical Pathology 64 529535. (doi:10.1136/jcp.2010.085621).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schwartzentruber J , Korshunov A , Liu XY , Jones DT , Pfaff E , Jacob K , Sturm D , Fontebasso AM , Quang DA & Tonjes M et al. 2012 Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482 226231. (doi:10.1038/nature10833).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Seligson DB , Horvath S , Shi T , Yu H , Tze S , Grunstein M & Kurdistani SK 2005 Global histone modification patterns predict risk of prostate cancer recurrence. Nature 435 12621266. (doi:10.1038/nature03672).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Serrano J , Goebel SU , Peghini PL , Lubensky IA , Gibril F & Jensen RT 2000 Alterations in the p16INK4a/CDKN2A tumor suppressor gene in gastrinomas. Journal of Clinical Endocrinology and Metabolism 85 41464156. (doi:10.1210/jc.85.11.4146).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shah MH , Binkley P , Chan K , Xiao J , Arbogast D , Collamore M , Farra Y , Young D & Grever M 2006 Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clinical Cancer Research 12 39974003. (doi:10.1158/1078-0432.CCR-05-2689).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shivakumar L , Minna J , Sakamaki T , Pestell R & White MA 2002 The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Molecular and Cellular Biology 22 43094318. (doi:10.1128/MCB.22.12.4309-4318.2002).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Singh P , Soon PS , Feige JJ , Chabre O , Zhao JT , Cherradi N , Lalli E & Sidhu SB 2012 Dysregulation of microRNAs in adrenocortical tumors. Molecular and Cellular Endocrinology 351 118128. (doi:10.1016/j.mce.2011.09.041).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Soon PS , Tacon LJ , Gill AJ , Bambach CP , Sywak MS , Campbell PR , Yeh MW , Wong SG , Clifton-Bligh RJ & Robinson BG et al. 2009 miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clinical Cancer Research 15 76847692. (doi:10.1158/1078-0432.CCR-09-1587).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stricker I , Tzivras D , Nambiar S , Wulf J , Liffers ST , Vogt M , Verdoodt B , Tannapfel A & Mirmohammadsadegh A 2012 Site- and grade-specific diversity of LINE1 methylation pattern in gastroenteropancreatic neuroendocrine tumours. Anticancer Research 32 36993706.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suh I , Weng J , Fernandez-Ranvier G , Shen WT , Duh QY , Clark OH & Kebebew E 2010 Antineoplastic effects of decitabine, an inhibitor of DNA promoter methylation, in adrenocortical carcinoma cells. Archives of Surgery 145 226232. (doi:10.1001/archsurg.2009.292).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tahiliani M , Koh KP , Shen Y , Pastor WA , Bandukwala H , Brudno Y , Agarwal S , Iyer LM , Liu DR & Aravind L et al. 2009 Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324 930935. (doi:10.1126/science.1170116).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • The American Association for Cancer Research Human Epigenome Task Force & European Union NoE, Scientific Advisory Board Moving AHEAD with an international human epigenome project Nature 454 2008 711715. (doi:10.1038/454711a).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tombol Z , Szabo PM , Molnar V , Wiener Z , Tolgyesi G , Horanyi J , Riesz P , Reismann P , Patocs A & Liko I et al. 2009 Integrative molecular bioinformatics study of human adrenocortical tumors: microRNA, tissue-specific target prediction, and pathway analysis. Endocrine-Related Cancer 16 895906. (doi:10.1677/ERC-09-0096).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tombol Z , Eder K , Kovacs A , Szabo PM , Kulka J , Liko I , Zalatnai A , Racz G , Toth M & Patocs A et al. 2010 MicroRNA expression profiling in benign (sporadic and hereditary) and recurring adrenal pheochromocytomas. Modern Pathology 23 15831595. (doi:10.1038/modpathol.2010.164).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Toyota M , Ahuja N , Ohe-Toyota M , Herman JG , Baylin SB & Issa JP 1999 CpG island methylator phenotype in colorectal cancer. PNAS 96 86818686. (doi:10.1073/pnas.96.15.8681).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ulaner GA , Huang HY , Otero J , Zhao Z , Ben-Porat L , Satagopan JM , Gorlick R , Meyers P , Healey JH & Huvos AG et al. 2003 Absence of a telomere maintenance mechanism as a favorable prognostic factor in patients with osteosarcoma. Cancer Research 63 17591763.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Waddington CH 1957 The Strategy of the Genes: a Discussion of Some Aspects of Theoretical Biology. London: Allen, Unwin

    • PubMed
    • Export Citation
  • Wang T , Lv M , Shen S , Zhou S , Wang P , Chen Y , Liu B , Yu L & Hou Y 2012 Cell-free microRNA expression profiles in malignant effusion associated with patient survival in non-small cell lung cancer. PLoS ONE 7 e43268. (doi:10.1371/journal.pone.0043268).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Warneboldt J , Haller F , Horstmann O , Danner BC , Fuzesi L , Doenecke D & Happel N 2008 Histone H1x is highly expressed in human neuroendocrine cells and tumours. BMC Cancer 8 388. (doi:10.1186/1471-2407-8-388).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wild A , Ramaswamy A , Langer P , Celik I , Fendrich V , Chaloupka B , Simon B & Bartsch DK 2003 Frequent methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene in pancreatic endocrine tumors. Journal of Clinical Endocrinology and Metabolism 88 13671373. (doi:10.1210/jc.2002-021027).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Wilde RF , Heaphy CM , Maitra A , Meeker AK , Edil BH , Wolfgang CL , Ellison TA , Schulick RD , Molenaar IQ & Valk GD et al. 2012 Loss of ATRX or DAXX expression and concomitant acquisition of the alternative lengthening of telomeres phenotype are late events in a small subset of MEN-1 syndrome pancreatic neuroendocrine tumors. Modern Pathology 25 10331039.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yao JC , Shah MH , Ito T , Bohas CL , Wolin EM , Van Cutsem E , Hobday TJ , Okusaka T , Capdevila J & de Vries EG et al. 2011 Everolimus for advanced pancreatic neuroendocrine tumors. New England Journal of Medicine 364 514523. (doi:10.1056/NEJMoa1009290).

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yashiro T , Fulton N , Hara H , Yasuda K , Montag A , Yashiro N , Straus F II , Ito K , Aiyoshi Y & Kaplan EL 1993 Comparison of mutations of ras oncogene in human pancreatic exocrine and endocrine tumors. Surgery 114 758763.(discussion 763–754)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoshimoto K , Iwahana H , Fukuda A , Sano T , Saito S & Itakura M 1992 Role of p53 mutations in endocrine tumorigenesis: mutation detection by polymerase chain reaction-single strand conformation polymorphism. Cancer Research 52 50615064.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang HY , Rumilla KM , Jin L , Nakamura N , Stilling GA , Ruebel KH , Hobday TJ , Erlichman C , Erickson LA & Lloyd RV 2006 Association of DNA methylation and epigenetic inactivation of RASSF1A and β-catenin with metastasis in small bowel carcinoid tumors. Endocrine 30 299306. (doi:10.1007/s12020-006-0008-1).

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand