Next Article in Journal
Mammogram Analysis with YOLO Models on an Affordable Embedded System
Previous Article in Journal
An Above-Average Lymph Node Yield Removed During Curative Neck Dissection in Advanced Head and Neck Squamous Cell Carcinomas Improves Survival
Previous Article in Special Issue
Decoding the Epigenome: Comparative Analysis of Uterine Leiomyosarcoma and Leiomyoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetics and the Tumor Microenvironment in Neuroendocrine Tumors

Cancer Cell Signalling Lab, Department of Medicine, University of Udine, 33100 Udine, Italy
*
Author to whom correspondence should be addressed.
Cancers 2026, 18(1), 69; https://doi.org/10.3390/cancers18010069
Submission received: 21 November 2025 / Revised: 19 December 2025 / Accepted: 23 December 2025 / Published: 25 December 2025

Simple Summary

The incidence of neuroendocrine neoplasms is increasing over the years, and therapy effectiveness is still relatively limited by the development of resistance mechanisms. Given the low rate mutation of these neoplasia, there is evidence of an emerging role of epigenetic modifications in neuroendocrine tumor pathogenesis and therapy resistance, leading to a need to investigate more deeply how these mechanisms are altered in these tumor types, how they can affect therapeutic efficacy, and if they can mediate the crosstalk between tumor cells, tumor microenvironment components, and tumor-stroma crosstalk that support tumoral development and progression. In this review we aim to describe all the recent evidence involving epigenetic mechanisms and the tumor microenvironment components that support tumor onset, development and resistance to treatment in the context of neuroendocrine neoplasm. The goal is to highlight potential new path to improve treatment response and outcomes in patients affected by neuroendocrine neoplasms.

Abstract

Background: Neuroendocrine neoplasms (NENs) represent a heterogeneous group of malignancies arising from neuroendocrine cells and are most commonly localized in the gastroenteropancreatic and pulmonary systems. Overall, most NENs are characterized by a low mutational burden. Consequently, increasing research attention has focused on epigenetic mechanisms and the tumor microenvironment, which may actively contribute to tumor pathogenesis. Moreover, epigenetic modifications represent plausible mediators of communication in the crosstalk between neuroendocrine cancer cells and components of the tumor microenvironment, as observed in other non-neuroendocrine malignancies. Objectives: The aim of this review is to summarize current knowledge on the role of epigenetic regulation and the tumor microenvironment in the pathogenesis of neuroendocrine tumors and to discuss their potential as targets for more effective therapeutic strategies. Methods: We reviewed research articles published over the past decades that investigated the involvement of epigenetic mechanisms and the tumor microenvironment in NENs. Results: Several studies highlight the pivotal role of epigenetic alterations and tumor microenvironment interactions in NEN biology, influencing tumor development, progression, and therapeutic response. Conclusions: Further studies are warranted to deepen understanding in this field, as this may lead to the development of more effective therapies and improved clinical outcomes for patients with neuroendocrine neoplasms.

1. Neuroendocrine Neoplasms

Neuroendocrine neoplasms (NENs) represent a group of rare epithelial malignancies, with a heterogeneous spectrum that ranges from well-differentiated neuroendocrine tumors (NETs) (80–90%) to very aggressive poorly differentiated neuroendocrine carcinomas (NECs) (10–20%) Table 1 [1]. These neoplasms arise from neuroendocrine cells diffused throughout the body, although the most frequent locations are the gastroenteropancreatic (GEP) and bronchopulmonary tracts [2], and are associated with symptoms secondary to the release of vasoactive amines and hormones [3]. NENs are mostly sporadic, but in 10–30%, they can arise within the context of inherited genetic syndromes, such as MEN1 (multiple endocrine neoplasia type 1), NF1 (neurofibromatosis type 1), TSC1/2 (tuberous sclerosis), and VHL (Von Hippel–Lindau disease) [4].

2. Epigenetics of NETs

Epigenetics represent the heritable and stable changes in gene expression determined by chemical modifications of DNA and its associated proteins, rather than alterations to the DNA sequence [12]. The most common epigenetic mechanisms include DNA methylation status, histone post-translational modifications, and regulation by noncoding RNAs (ncRNAs) [13] Figure 1.
Neuroendocrine tumors present a low mutation rate compared to other tumors, while the role of epigenetic mechanisms regulating gene expression seems to be relevant in their pathogenesis and development; this supports the rational to further investigate the epigenetic mechanisms at the basis of NEN pathogenesis. Furthermore, epigenetic modifications are reversible and targetable, representing an attractive target for treatment [14], assuming prior clarification of their molecular role in NET pathogenesis.
At present, the gold standard for NET treatment is surgical removal of the tumoral mass, while new methods are being studied, such as preoperative contrast-enhanced computed tomography (CE-CT), to implement tumor characterization [15]. Whenever, in more aggressive NETs, surgery is not feasible, somatostatin analogs (SSAs), chemotherapy, targeted therapies, and specialized radiation, such as peptide receptor radionuclide therapy (PRRT), could be used, despite often leading to resistance development.
This approach may also allow, through combination with established medical therapies—including classical somatostatin analogs (SSAs), targeted agents, and cytotoxic chemotherapy—the overcoming of resistance mechanisms, thereby achieving more effective treatment strategies for neuroendocrine tumors [16,17]. In more aggressive NETs, for which surgical intervention is not feasible, SSAs are currently used; however, their efficacy is often limited by the development of therapeutic resistance. The proposed causative mechanisms for development of resistance upon SSA administration include epigenetic changes within tumor cells, as histone methylation and/or acetylation, that reduce SSTR expression. Similarly, since high SSTR2 levels are also essential for PRRT to ensure efficient delivery of radiation, epigenetic modifications that reduce SSTR expression can lead to resistance development. This hypothesis is supported by the observation that using HDAC inhibitors (HDACi) and DNMT inhibitors (DNMTi), NET cells presented an overexpression of SSTR2 [13]. Consequently, the combination of HDACi and/or DNMTi with SSAs or PRRT might overcome this epigenetically derived resistance, resulting in a more efficient antitumoral treatment [18]. This combination is also attractive for targeted therapies, including mTOR or VEGFR inhibitors, such as Everolimus and Sunitinib, respectively. In fact, the potential involvement of epigenetic mechanisms in Everolimus resistance development is supported by the observed dysregulated expression of the downstream mTOR components [19]. Similarly, Sunitinib is limited in its efficacy by the development of resistance mechanisms that are in part epigenetically derived, such as the activation of HIF-1α that in turn is responsible for the activation of alternative pathways associated with the expression of pro-angiogenic factors [20].
Given that neuroendocrine tumors present a low mutational burden, it is fundamental to identify new epigenetic markers rather than genetic ones, for both diagnosis and prognosis, which would also allow a more precise distinction between different subtypes, which can be treated with more personalized and efficient therapies. Recently, different epigenetic markers are under investigation in neuroendocrine tumors. For instance, alterations in methylated levels of ZNF536, encoding for a transcriptional repressor, have been proposed to have both diagnostic and prognostic implications in NETs [21]. Furthermore, the expression of mitochondrial genes related to oxidative stress (MTGs-OS) that is epigenetically regulated can be considered a prognostic marker that also provides clinical guidance for PNET treatment [22]. There are even some biomarkers reflecting the interaction between tumor cells and the tumor microenvironment, which are relevant to tumor progression and metastasis, such as the sEV-miR-183-5p prognostic marker, a noncoding RNA embedded in small extracellular vesicles promoting the polarization of tumor-associated macrophages into the immunosuppressive M2 phenotype and upregulating SPP1, which represents a good candidate for a therapeutic target [23].

2.1. DNA Methylation

DNA methylation is catalyzed by DNA methyltransferases (DNMTs) [24] and frequently occurs on cytosine residues that precede guanine nucleotides or CpG sites, with the exception of CpG islands [25]. Perturbation of methylation patterns could give rise to gene expression alterations responsible for malignant cell transformation [26]. In particular, hypermethylation of promoter regions in tumor suppressor genes often results in their silencing, contributing to tumor growth and progression [27].
In lung NETs, the hypermethylation of many tumor suppressors has been reported, such as the cell-cycle inhibitor CDKN2A [7,28], RASSF1A, a scaffold protein involved in cell signaling pathways such as Erk, Hippo, apoptotic, and p53 [29,30], SOX17, a transcription factor inhibiting proliferation of lung cancer cells and downregulating the Wnt/β-catenin pathway [31], and TAC1, which encodes for tachykinins, involved in neurogenic inflammatory processes and in endotoxin-induced airway inflammation (Figure 2A) [32,33].
Similarly to pulmonary NETs, the majority of pancreatic NETs present hypermethylated RASSF1A and CDKN2A genes [34]. Other hypermethylated tumor suppressors in PanNETs include PHLDA3, which downregulates the PI3K/AKT oncogenic signaling pathway [35], SSTR2, a G-protein coupled receptor that inhibits the release of hormones [36], APC, an antagonist of the Wnt/β-catenin pathway, and MEN1 and MGMT, encoding for DNA repair proteins involved in cellular defense (Figure 2B) [37,38]. Moreover, many PanNETs are characterized by significant hypomethylation of LINE-1 and Alu sequences [39], resulting in transcriptional activation and consequently in genetical instability [40].
In gastrointestinal NETs, promoters of RASSF1A, SEMA3F, encoding for semaphorin 3 that induces apoptosis, and CTNNB1, representing the catenin β1 gene that constitutes adherence junctions to regulate cell growth and inter-cellular adhesions, are hypermethylated [41]. Moreover, MAPK4, which encodes for a kinase regulating proliferation, migration, and apoptosis via the AKT/mTOR and/or PDK1 signaling pathways [42,43], and WIF1, which is a Wnt antagonist, are silenced through hypermethylation (Figure 2C) [44]. Similarly to what is reported for PanNETs, in many GI-NETs, LINE1 and Alu repetitive elements are also hypomethylated, causing genetical instability [42,45].
MTC, which represents the most common thyroid NET subtype, presents some genes regulated in their expression by diverse methylation levels, such as MGMT, which has been shown to be hypermethylated at the level of the CG_16698623 dinucleotide [46], RASSF1A, which is frequently hypermethylated in thyroid NETs as a promoter of endocrine-related genes, including SLC5A5 and THST, and as CASP8 and ATM genes (Figure 2D). It is interesting to note that nondifferentiated medullary tumors exhibit more hypomethylated than hypermethylated genes. Some examples of hypomethylated genes in MTC are INSL4 and DPPA2, which represent two genes reported to promote tumorigenesis in different cancer types, acting as oncogenes [47].

2.2. Histone Modifications

Histone modifications, such as methylation, acetylation, and phosphorylation of amino acidic residues on histone protein tails, affect chromatin structure and accessibility, regulating gene expression [48]. They represent an important epigenetic mechanism to regulate the chromatin state and consequently gene expression. If altered, some of them can contribute to the pathogenesis of tumors, including neuroendocrine neoplasms [7]. Acetylation levels are regulated by the opposite activity of HATs (histone acetyltransferases) and HDACs (histone deacetylases), which have been shown to be altered in different cancer types [26]. HDACs have been under investigation as potential drivers of cancer progression since their inhibition has been shown to reverse the malignant phenotype in various tumor entities [49], including NETs [17] (Figure 1).
In pulmonary NETs, a significant correlation has been reported between the acetylation level of histone H3 surrounding the promoter region of Notch1 gene and its expression. In fact, histone deacetylation results in Notch1 silencing in small-cell lung cancer (SCLC) (Figure 2A) [50]. Expression of Notch1 in SCLC is associated with a reduction in its proliferation while promoting cell apoptosis, a reduction in cell motility, invasion, and metastasis, and enhancement of cell–cell adhesion by EMT inhibition [51]. Compared to histone H3, H4 acetylation has a wider distribution along transcribed genes, and there is evidence that histone H4 acetylation at lysine 16 (H4KA16) and trimethylation at lysine 20 (H4KM20) gradually decrease from low- to high-grade in lung NETs [52]. KMT2 proteins regulate methylation of lysine 4 and 27 of histone 3 tails (H3K4 and H3K27). In SCLC, mutations in KMT2 genes are associated with low protein levels, resulting in a reduction in mono-methylation of histone H3 lysine 4 (H3K4me1) [53] and in histone and DNA hypomethylation, finally affecting DNMT3A expression levels [54].
In PanNETs, loss of menin is responsible for H3K4me3 loss, because menin represents a subunit of an MLL1/MLL2-containing histone methyltransferase complex that tri-methylates histone H3 at lysine 4, enhancing transcriptional activity [55]. Thus, MEN1 loss alters the epigenetic landscape of its target genes, such as IGF2, CDKN2C, and CDKN1B (Figure 2B) [56]. Furthermore, in some PanNETs, downregulation of the WIF1, DKK-1, and DKK-3 genes has been observed, which is caused by repressive histone modifications resulting in an increased H3K9me2 presence at the promoter level [38].
In primary intestinal NETs, a high expression of di-methylation on lysine 4 of histone 3 (H3K4) has been observed [53]. Unfortunately, despite evidence of DNA methylation levels regulating SST2 expression, there are still some gaps related to an effective regulatory role of histone modifications, maybe due to the limited sample size. However, the effects of HDACi on SST2 expression led researchers to think about a possible role of histone acetylation in SST2 expression regulation in pancreatic NETs (Figure 2C) [57,58].
Concerning thyroid NETs, in more aggressive MTCs, a significant increase in the EZH2 and SMYD3 genes has been observed. An EZH2 increase leads to the silencing of genes that promote differentiation and restrain proliferation and to altered regulation of genes involved in the Wnt/β-catenin, Erk, and Akt signaling pathways, which are mainly involved in MTC tumorigenesis (Figure 2D). SMYD3 is a histone methyltransferase that regulates several target genes, including cell cycle mediators and oncogenes [59].

2.3. Noncoding RNAs

MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) regulate gene expression at the post-transcriptional level [60,61]. In more detail, miRNAs are involved in RNA interference machinery suppressing the protein translation or decay of mRNA by binding to the untranslated regions (UTRs) of mRNA. Through this mechanism, miRNAs act as epigenetic modulators by targeting enzymes involved in epigenetic reactions and, in turn, miRNA expression is regulated by epigenetic machinery, forming the miRNA–epigenetic feedback loop [62,63]. Long noncoding RNAs mediate DNA and histone modifications and chromatin organization by recruiting DNA- and histone-modifying enzymes and chromatin organization-associated proteins at specific genomic sites to regulate pre-transcriptional gene expression [64].
Noncoding RNAs are involved in the regulation of proliferation, differentiation, metabolism, and cell death, and their dysregulation is implicated in oncogenesis, as observed in NETs [65].
Consistently, lung NETs present an altered ncRNA pattern, including upregulation of miR-21, which directs cell growth by inhibiting apoptosis, and miR-34a, which acts as a tumor suppressor (Figure 2A) [66]. Many other ncRNAs have been shown to be altered in their expression, supporting tumor pathogenesis, such as miR-1 downregulation, which is associated with SCLC growth and metastasis through the regulation of the CXCR4/FOXM1/RRM2 axis [67], and lncRNA PROX1-AS1 upregulation, which acts as a tumor promoter [68].
Additionally, in pancreatic NETs, the upregulation of miR-21 has been observed, similarly to what has been observed for miR-196a, which regulates Akt signaling, and miR-670-3p, which regulates claudins CLDN1 and CLDN2, whose dysregulated expression is associated with tumor initiation, progression, and metastasis (Figure 2B) [69,70,71]. In addition, miRNAs could be used by tumors as a mechanism of adaptation to hypoxia-related stress, as demonstrated by upregulation of miR-210 induced by HIF-1α [70]. PanNETs also present alterations in lncRNA expression patterns, which can contribute to tumor pathogenesis, as observed for the overexpression of lncRNA XLOC_221242 compared to normal tissue, which is positively related with DNER mRNA, a factor involved in the Notch signaling pathway [72]. Another example involves MEN1, which, in physiological conditions, activates lncRNA MEG3, which downregulates c-Met proto-oncogene expression, acting as a tumor suppressor. In MEN1-mutated PanNETs, since MEG3 is not activated, it does not present a c-Met reduction [73].
Many dysregulated ncRNAs were also identified in GI-NETs, whose altered expression is linked to different mechanisms that support tumor development. For instance, in rectal NEN upregulation of miRNAs, including miR-885-5p, miR-135a, miR-198, miR-204, miR-216a/b, miR-452, miR-486-3p, miR-499, and miR-146b-3p, has been associated with the invasion of lymphatic vessels. Furthermore, miR-186 downregulation in colorectal NETs is related to PTTG1 overexpression, resulting in the promotion of cell proliferation, invasion, and metastasis [74,75]. In addition, dysregulated lncRNAs were also identified in GI-NETs, such as HOTAIR, which acts as a chromatin state regulator, which could represent a potential therapeutic target against cancer (Figure 2C) [76].
In both familial and sporadic MTC forms, dysregulated expression of ncRNAs has been observed, such as overexpressed miR-375, which targets a growth inhibitor gene, YAP1, which is a transcription factor involved in the Hippo signaling pathway, which presents a role in development, growth, repair, and homeostasis (Figure 2D) [77].

2.4. Emerging Epigenetic Mechanisms

As well as the previously described more “classical” epigenetic processes, others have recently been discovered to contribute to neuroendocrine tumor biology, such as RNA methylation and chromatin-remodeling complexes (Figure 1).
The RNA methylation pattern is a critical post-transcriptional regulator mechanism that governs gene expression. As confirmed by many tumor types, aberrant N6-methyladenosine (m6A) modification of ncRNAs may affect cancer progression [78]. Consistently, in sparsely granulated subtypes of pituitary neuroendocrine tumors, the fat mass and obesity-associated protein (FTO), an m6A demethylase, was observed to be upregulated and, in turn, modulate the m6A levels of the mRNA encoding for DSP, a critical desmosomal component. This results in a reduction in desmosome organization, leading to enhanced tumor invasiveness and metastasis [79].
Another recently highlighted epigenetic mechanism that plays an important role in NET pathogenesis involves the Switch/Sucrose Nonfermentable (SWI/SNF) complex, a chromatin remodeling complex that regulates nucleosome structures. In particular, in NETs, the aberrant expression of SWI/SNF complex subunits affects downstream gene expression, including NE markers, highlighting its emerging role in NET pathogenesis [80].

3. Tumor Microenvironments in NETs

The tumor microenvironment (TME) represents a complex and dynamic surrounding that actively interacts with tumor cells, affecting processes such as cancer development, progression, metastasis, and immune evasion. Although its specific composition varies between different tumor types, the TME includes immune cells of innate and adaptive immunity, stromal cells, extracellular matrix (ECM), and blood vessels [81].
Immune cells, including B and T cells, NK cells, mast cells, dendritic cells, and macrophages, are known to infiltrate NETs, creating an immunosuppressive microenvironment that enables tumor progression [82]. In recent years, there has been evidence of the role of TME and tumor-infiltrating lymphocytes (TILs) in cancer prognosis and therapy response. In this regard, a study on neuroendocrine tumors of the lung demonstrated that in LCNECs, high levels of CD8+ TILs and PD-L1 expression may reflect a greater responsiveness to immune checkpoint inhibitors compared to carcinoids, characterized by a distinct TME with a desert-immune phenotype, which may show a limited benefit from immunotherapy. Consequently, an integrated approach combining molecular profiling and immune data for different neuroendocrine tumor types may define a more effective treatment [83].
ECM degradation contributes to NET development and progression by releasing chemoattractants that recruit inflammatory cells and pro-angiogenic factors and creating a mechanic stimulus that may influence tumor behavior [82].
Among stromal cells, fibroblasts present a crosstalk with NET cells, where tumor cells stimulate the proliferation and activation of fibroblasts by secreting soluble factors, including TGF-β, serotonin, and PDGF, and, in turn, activated CAFs modulate the proliferative capability of NET cells. Besides soluble factors, the means of communication between TME components, such as CAFs, and tumor cells are almost unknown, although epigenetic modifications represent plausible candidates. Supporting this concept, emerging evidence indicates that epigenetic regulation of tumor cells by the TME occurs in neuroendocrine tumors. In particular, hypoxia within the TME of gastroenteropancreatic NETs (GEP-NETs) has been shown to induce promoter hypermethylation-mediated downregulation of carbonyl reductase 4 (CBR4), thereby promoting tumor progression through an ubiquitin-dependent reduction in fatty acid synthase (FASN) expression and the activation of the mTOR signaling pathway [84].
As previously mentioned, there is still limited information reported in the literature about specific mechanisms that TME elements use to regulate epigenetic modifications in NET cells. Nevertheless, some hypotheses, which, in future, may be interesting to investigate in NET cells, can be proposed based on the non-NE tumor knowledge on this topic.
In general, cellular components of the TME, such as TAMs, MDSCs, DCs, and CAFs, secrete different cytokines, among which members of the IL-6 family, including leukemia inducible factor (LIF), IL-10, TNF-α, and TGF-β [85], that affect both tumor cells and other stromal components of the TME through different mechanisms, including epigenetic regulation. For instance, transient TGF-β stimulus can induce LIF expression in both tumor and stromal cells via DNA hypomethylation of CpG pairs and changes in histone methylation status [86].
In different tumor types, the epigenetic modifications induced by CAFs in tumor cells have been analyzed, such as in ovarian cancer cells, where it was observed that CAFs prompt EZH2 histone methyltransferase upregulation, which results in cancer cell migration and increased invasion [87]. Similarly in SI-NETs, a high differential expression of EZH2 has been reported; this catalyzes H3K27me3 marks and is associated with high proliferation rates, metastasis, and poor survival [34]. Hence, we can hypothesize that neuroendocrine tumor cells may also be influenced by CAFs present in the TME through epigenetic regulation, as suggested by the EZH2 upregulation observed in SI-NETs and thyroid NETs.
The crosstalk between CAFs and tumor cells was further confirmed by the overexpression of TNF-α observed in ovarian cancer, which is determined by promoter DNA hypomethylation and chromatin remodeling, which favors TGF-α transcription in CAFs of the TME, which in turn promotes EGFR signaling, resulting in an increased metastatic ability of cancer cells. This mechanism generates a loop of epigenetic regulation between CAFs and cancer cells [88].
Other TME components that can have a role in regulating tumor cell expression pattern and development include immune cells, where TAMs were observed to increase DNMT1 expression in gastric cancer cells, leading to the silencing of the tumor suppressor gelsolin gene via hypermethylation [89].
Moreover, another epigenetically driven mechanism of communication between TME and tumor cells is the exosome release. In fact, both cancer and non-cancer cells in the TME secrete exosomes to “talk” with each other, and in these structures, ncRNAs can regulate metabolic reprogramming, angiogenesis, epithelial–mesenchymal transition, and ECM remodeling [90].

4. Conclusions

From this review, we can conclude that epigenetic mechanisms can have an important role in neuroendocrine tumor pathogenesis, representing interesting potential therapeutic targets but also prognostic and diagnostic biomarkers. For this reason, a further understanding of these regulatory mechanisms in neuroendocrine tumor cells is required in order to both define in more detail the impact of epigenetics on NEN pathogenesis and develop more efficient therapeutic strategies.
Another important player in tumoral progression is the tumor microenvironment, which, even through epigenetic mechanisms, is able to interact with cancer cells, promoting tumor progression, invasion, and immune response regulation. Consequently, it would be interesting to achieve a deeper knowledge on the complex molecular networks regulating the communication and balance between tumor cells and the surrounding microenvironment, especially considering the relatively limited knowledge about this topic in NETs. At present, the main gap consists of the mechanisms by which TME elements are able to influence cancer cells leading to tumoral progression, where epigenetic regulation can be a strong candidate, as suggested by other tumor types. Hence, further investigation is required to identify possible therapeutic targets that enable more efficient NEN treatment.

Author Contributions

A.C. wrote the original draft, and T.G. conceived the idea and reviewed the original manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Departmental Strategic Plan (PSD) of the University of Udine-Interdepartmental Project on Healthy Ageing (2020-25). The Cancer Cell Signalling Lab is a partner of COST Action CA24162, Adaptive and acquired resistance in gastrointestinal cancers—contemporary and emerging resolutions (AdResCanCER).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
NENNeuroendocrine Neoplasm
NETNeuroendocrine Tumor
NECNeuroendocrine Carcinoma
GEPGastroenteropancreatic
MEN1Multiple Endocrine Neoplasia Type 1
NF1Neurofibromatosis Type 1
TSC1/2Tuberous Sclerosis
VHLVon Hippel–Lindau
TCTypical Carcinoid
ACAtypical Carcinoid
LCNECLarge-Cell Neuroendocrine Carcinoma
SCLCSmall-Cell Lung Carcinoma
PanNECPancreatic Neuroendocrine Carcinoma
PanNETPancreatic Neuroendocrine Tumor
SSASomatostatin Analog
HDACiHistone Deacetylase Inhibitor
DNMTiDNA Methyltransferase Inhibitor
MTCMedullary Thyroid Carcinoma
TMETumor Microenvironment
ECMExtracellular Matrix
TILTumor-Infiltrating Lymphocyte
CAFCancer-Associated Fibroblast
TAMTumor-Associated Macrophage
MDSCMyeloid-Derived Suppressor Cell
DCDendritic Cell
LIFLeukemia Inducible Factor

References

  1. Rizen, E.N.; Phan, A.T. Neuroendocrine Tumors: A Relevant Clinical Update. Curr. Oncol. Rep. 2022, 24, 703–714. [Google Scholar] [CrossRef] [PubMed]
  2. Wahba, A.; Tan, Z.; Dillon, J.S. Management of Functional Neuroendocrine Tumors. Curr. Probl. Cancer 2024, 52, 101130. [Google Scholar] [CrossRef]
  3. Alexander, E.S.; Ziv, E. Neuroendocrine Tumors: Genomics and Molecular Biomarkers with a Focus on Metastatic Disease. Cancers 2023, 15, 2249. [Google Scholar] [CrossRef]
  4. Lauricella, E.; Chaoul, N.; D’Angelo, G.; Giglio, A.; Cafiero, C.; Porta, C.; Palmirotta, R. Neuroendocrine Tumors: Germline Genetics and Hereditary Syndromes. Curr. Treat. Options Oncol. 2025, 26, 55–71. [Google Scholar] [CrossRef] [PubMed]
  5. Hendifar, A.E.; Marchevsky, A.M.; Tuli, R. Neuroendocrine Tumors of the Lung: Current Challenges and Advances in the Diagnosis and Management of Well-Differentiated Disease. J. Thorac. Oncol. 2017, 12, 425–436. [Google Scholar] [CrossRef]
  6. Vocino Trucco, G.; Righi, L.; Volante, M.; Papotti, M. Updates on Lung Neuroendocrine Neoplasm Classification. Histopathology 2024, 84, 67–85. [Google Scholar] [CrossRef]
  7. Colapietra, F.; Della Monica, P.; Di Napoli, R.; França Vieira e Silva, F.; Settembre, G.; Marino, M.M.; Ballini, A.; Cantore, S.; Di Domenico, M. Epigenetic Modifications as Novel Biomarkers for Diagnosis, Prognosis, and Therapeutic Targeting in Thyroid, Pancreas, and Lung Neuroendocrine Tumors. JCM 2025, 14, 2622. [Google Scholar] [CrossRef]
  8. Sun, J. Pancreatic Neuroendocrine Tumors. IRDR 2017, 6, 21–28. [Google Scholar] [CrossRef]
  9. Khanna, L.; Prasad, S.R.; Sunnapwar, A.; Kondapaneni, S.; Dasyam, A.; Tammisetti, V.S.; Salman, U.; Nazarullah, A.; Katabathina, V.S. Pancreatic Neuroendocrine Neoplasms: 2020 Update on Pathologic and Imaging Findings and Classification. RadioGraphics 2020, 40, 1240–1262. [Google Scholar] [CrossRef] [PubMed]
  10. Stawarski, A.; Maleika, P. Neuroendocrine Tumors of the Gastrointestinal Tract and Pancreas: Is It Also a Challenge for Pediatricians? Adv. Clin. Exp. Med. 2020, 29, 265–270. [Google Scholar] [CrossRef]
  11. Patel, N.; Barbieri, A.; Gibson, J. Neuroendocrine Tumors of the Gastrointestinal Tract and Pancreas. Surg. Pathol. Clin. 2019, 12, 1021–1044. [Google Scholar] [CrossRef] [PubMed]
  12. Farsetti, A.; Illi, B.; Gaetano, C. How Epigenetics Impacts on Human Diseases. Eur. J. Intern. Med. 2023, 114, 15–22. [Google Scholar] [CrossRef]
  13. Colao, A.; De Nigris, F.; Modica, R.; Napoli, C. Clinical Epigenetics of Neuroendocrine Tumors: The Road Ahead. Front. Endocrinol. 2020, 11, 604341. [Google Scholar] [CrossRef]
  14. Di Domenico, A.; Wiedmer, T.; Marinoni, I.; Perren, A. Genetic and Epigenetic Drivers of Neuroendocrine Tumours (NET). Endocr.-Relat. Cancer 2017, 24, R315–R334. [Google Scholar] [CrossRef] [PubMed]
  15. Ren, S.; Chen, X.; Wang, J.; Zhao, R.; Song, L.; Li, H.; Wang, Z. Differentiation of Duodenal Gastrointestinal Stromal Tumors from Hypervascular Pancreatic Neuroendocrine Tumors in the Pancreatic Head Using Contrast-Enhanced Computed Tomography. Abdom. Radiol. 2019, 44, 867–876. [Google Scholar] [CrossRef]
  16. Toffoli, L.; Ditsiou, A.; Gagliano, T. Exploring Emerging Therapeutic Targets and Opportunities in Neuroendocrine Tumors: Updates on Receptor Tyrosine Kinases. Receptors 2024, 3, 145–154. [Google Scholar] [CrossRef]
  17. Gagliano, T.; Brancolini, C. Epigenetic Mechanisms beyond Tumour–Stroma Crosstalk. Cancers 2021, 13, 914. [Google Scholar] [CrossRef]
  18. McClellan, K.; Chen, E.Y.; Kardosh, A.; Lopez, C.D.; Del Rivero, J.; Mallak, N.; Rocha, F.G.; Koethe, Y.; Pommier, R.; Mittra, E.; et al. Therapy Resistant Gastroenteropancreatic Neuroendocrine Tumors. Cancers 2022, 14, 4769. [Google Scholar] [CrossRef]
  19. Zanini, S.; Renzi, S.; Giovinazzo, F.; Bermano, G. mTOR Pathway in Gastroenteropancreatic Neuroendocrine Tumor (GEP-NETs). Front. Endocrinol. 2020, 11, 562505. [Google Scholar] [CrossRef]
  20. Pozas, J.; San Román, M.; Alonso-Gordoa, T.; Pozas, M.; Caracuel, L.; Carrato, A.; Molina-Cerrillo, J. Targeting Angiogenesis in Pancreatic Neuroendocrine Tumors: Resistance Mechanisms. IJMS 2019, 20, 4949. [Google Scholar] [CrossRef]
  21. Zeng, L.; Zhang, L.; Yin, C.; Chen, X.; Chen, X.; Sun, L.; Sun, J. Characterization of Zinc Finger Protein 536, a Neuroendocrine Regulator, Using Pan-Cancer Analysis. Eur. J. Med. Res. 2024, 29, 273. [Google Scholar] [CrossRef] [PubMed]
  22. Cui, Y.; Yuan, Q.; Chen, J.; Jiang, J.; Guan, H.; Zhu, R.; Li, N.; Liu, W.; Wang, C. Determination and Characterization of Molecular Heterogeneity and Precision Medicine Strategies of Patients with Pancreatic Cancer and Pancreatic Neuroendocrine Tumor Based on Oxidative Stress and Mitochondrial Dysfunction-Related Genes. Front. Endocrinol. 2023, 14, 1127441. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, W.; Xu, J.; Huang, D.; Qin, Y.; Lou, X.; Gao, H.; Ye, Z.; Wang, F.; Wang, Y.; Jing, D.; et al. Small Extracellular Vesicle-miR-183-5p Mediated Crosstalk between Tumor Cells and Macrophages in High-Risk Pancreatic Neuroendocrine Tumors. Oncogene 2025, 44, 2907–2923. [Google Scholar] [CrossRef] [PubMed]
  24. Moore, L.D.; Le, T.; Fan, G. DNA Methylation and Its Basic Function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [PubMed]
  25. Nishiyama, A.; Nakanishi, M. Navigating the DNA Methylation Landscape of Cancer. Trends Genet. 2021, 37, 1012–1027. [Google Scholar] [CrossRef]
  26. Malavasi, E.; Adamo, M.; Zamprogno, E.; Vella, V.; Giamas, G.; Gagliano, T. Decoding the Tumour Microenvironment: Molecular Players, Pathways, and Therapeutic Targets in Cancer Treatment. Cancers 2024, 16, 626. [Google Scholar] [CrossRef] [PubMed]
  27. Geissler, F.; Nesic, K.; Kondrashova, O.; Dobrovic, A.; Swisher, E.M.; Scott, C.L.; Wakefield, M.J. The Role of Aberrant DNA Methylation in Cancer Initiation and Clinical Impacts. Ther. Adv. Med. Oncol. 2024, 16, 17588359231220511. [Google Scholar] [CrossRef]
  28. Foulkes, W.D.; Flanders, T.Y.; Pollock, P.M.; Hayward, N.K. The CDKN2A (P16) Gene and Human Cancer. Mol. Med. 1997, 3, 5–20. [Google Scholar] [CrossRef]
  29. Raos, D.; Ulamec, M.; Katusic Bojanac, A.; Bulic-Jakus, F.; Jezek, D.; Sincic, N. Epigenetically Inactivated RASSF1A as a Tumor Biomarker. Bosn. J. Basic. Med. Sci. 2020, 21, 386. [Google Scholar] [CrossRef]
  30. García-Gutiérrez, L.; McKenna, S.; Kolch, W.; Matallanas, D. RASSF1A Tumour Suppressor: Target the Network for Effective Cancer Therapy. Cancers 2020, 12, 229. [Google Scholar] [CrossRef]
  31. Yin, D.; Jia, Y.; Yu, Y.; Brock, M.V.; Herman, J.G.; Han, C.; Su, X.; Liu, Y.; Guo, M. SOX17 Methylation Inhibits Its Antagonism of Wnt Signaling Pathway in Lung Cancer. Discov. Med. 2014, 14, 33. [Google Scholar]
  32. Helyes, Z.; Elekes, K.; Sándor, K.; Szitter, I.; Kereskai, L.; Pintér, E.; Kemény, Á.; Szolcsányi, J.; McLaughlin, L.; Vasiliou, S.; et al. Involvement of Preprotachykinin A Gene-Encoded Peptides and the Neurokinin 1 Receptor in Endotoxin-Induced Murine Airway Inflammation. Neuropeptides 2010, 44, 399–406. [Google Scholar] [CrossRef]
  33. Du, C.; Tan, L.; Xiao, X.; Xin, B.; Xiong, H.; Zhang, Y.; Ke, Z.; Yin, J. Detection of the DNA Methylation of Seven Genes Contribute to the Early Diagnosis of Lung Cancer. J. Cancer Res. Clin. Oncol. 2024, 150, 77. [Google Scholar] [CrossRef]
  34. Crabtree, J.S. Epigenetic Regulation in Gastroenteropancreatic Neuroendocrine Tumors. Front. Oncol. 2022, 12, 901435. [Google Scholar] [CrossRef]
  35. Kamel, W.A.; Krishnaraj, J.; Ohki, R. The Role of PHLDA3 in Cancer Progression and Its Potential as a Therapeutic Target. Cancers 2025, 17, 1069. [Google Scholar] [CrossRef]
  36. Evans, J.S.; Beaumont, J.; Braga, M.; Masrour, N.; Mauri, F.; Beckley, A.; Butt, S.; Karali, C.S.; Cawthorne, C.; Archibald, S.; et al. Epigenetic Potentiation of Somatostatin-2 by Guadecitabine in Neuroendocrine Neoplasias as a Novel Method to Allow Delivery of Peptide Receptor Radiotherapy. Eur. J. Cancer 2022, 176, 110–120. [Google Scholar] [CrossRef] [PubMed]
  37. Tirosh, A.; Kebebew, E. Genetic and Epigenetic Alterations in Pancreatic Neuroendocrine Tumors. J. Gastrointest. Oncol. 2020, 11, 567–577. [Google Scholar] [CrossRef]
  38. Cives, M.; Simone, V.; Rizzo, F.M.; Silvestris, F. NETs: Organ-Related Epigenetic Derangements and Potential Clinical Applications. Oncotarget 2016, 7, 57414–57429. [Google Scholar] [CrossRef]
  39. Stefanoli, M.; La Rosa, S.; Sahnane, N.; Romualdi, C.; Pastorino, R.; Marando, A.; Capella, C.; Sessa, F.; Furlan, D. Prognostic Relevance of Aberrant DNA Methylation in G1 and G2 Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2014, 100, 26–34. [Google Scholar] [CrossRef] [PubMed]
  40. Marinoni, I.; Wiederkeher, A.; Wiedmer, T.; Pantasis, S.; Di Domenico, A.; Frank, R.; Vassella, E.; Schmitt, A.; Perren, A. Hypo-Methylation Mediates Chromosomal Instability in Pancreatic NET. Endocr.-Relat. Cancer 2017, 24, 137–146. [Google Scholar] [CrossRef]
  41. Samsom, K.G.; Van Veenendaal, L.M.; Valk, G.D.; Vriens, M.R.; Tesselaar, M.E.T.; Van Den Berg, J.G. Molecular Prognostic Factors in Small-Intestinal Neuroendocrine Tumours. Endocr. Connect. 2019, 8, 906–922. [Google Scholar] [CrossRef] [PubMed]
  42. Verdugo, A.D.; Crona, J.; Starker, L.; Stålberg, P.; Åkerström, G.; Westin, G.; Hellman, P.; Björklund, P. Global DNA Methylation Patterns through an Array-Based Approach in Small Intestinal Neuroendocrine Tumors. Endocr.-Relat. Cancer 2014, 21, L5–L7. [Google Scholar] [CrossRef] [PubMed]
  43. Yan, Y.; Dai, T.; Guo, M.; Zhao, X.; Chen, C.; Zhou, Y.; Qin, M.; Xu, L.; Zhao, J. A Review of Non-Classical MAPK Family Member, MAPK4: A Pivotal Player in Cancer Development and Therapeutic Intervention. Int. J. Biol. Macromol. 2024, 271, 132686. [Google Scholar] [CrossRef]
  44. Fotouhi, O.; Adel Fahmideh, M.; Kjellman, M.; Sulaiman, L.; Höög, A.; Zedenius, J.; Hashemi, J.; Larsson, C. Global Hypomethylation and Promoter Methylation in Small Intestinal Neuroendocrine Tumors: An In Vivo and In Vitro Study. Epigenetics 2014, 9, 987–997. [Google Scholar] [CrossRef]
  45. Baba, Y.; Yasuda, N.; Bundo, M.; Nakachi, Y.; Ueda, J.; Ishimoto, T.; Iwatsuki, M.; Miyamoto, Y.; Yoshida, N.; Oshiumi, H.; et al. LINE -1 Hypomethylation, Increased Retrotransposition and Tumor-specific Insertion in Upper Gastrointestinal Cancer. Cancer Sci. 2024, 115, 247–256. [Google Scholar] [CrossRef]
  46. Della Monica, R.; Cuomo, M.; Visconti, R.; Di Mauro, A.; Buonaiuto, M.; Costabile, D.; De Riso, G.; Di Risi, T.; Guadagno, E.; Tafuto, R.; et al. Evaluation of MGMT Gene Methylation in Neuroendocrine Neoplasms. Oncol. Res. 2021, 28, 837–845. [Google Scholar] [CrossRef]
  47. Rodríguez-Rodero, S.; Fernández, A.F.; Fernández-Morera, J.L.; Castro-Santos, P.; Bayon, G.F.; Ferrero, C.; Urdinguio, R.G.; Gonzalez-Marquez, R.; Suarez, C.; Fernández-Vega, I.; et al. DNA Methylation Signatures Identify Biologically Distinct Thyroid Cancer Subtypes. J. Clin. Endocrinol. Metab. 2013, 98, 2811–2821. [Google Scholar] [CrossRef]
  48. Yao, W.; Hu, X.; Wang, X. Crossing Epigenetic Frontiers: The Intersection of Novel Histone Modifications and Diseases. Signal Transduct. Target. Ther. 2024, 9, 232. [Google Scholar] [CrossRef]
  49. Schmitz, R.L.; Weissbach, J.; Kleilein, J.; Bell, J.; Hüttelmaier, S.; Viol, F.; Clauditz, T.; Grabowski, P.; Laumen, H.; Rosendahl, J.; et al. Targeting HDACs in Pancreatic Neuroendocrine Tumor Models. Cells 2021, 10, 1408. [Google Scholar] [CrossRef]
  50. Hassan, W.A.; Takebayashi, S.; Abdalla, M.O.A.; Fujino, K.; Kudoh, S.; Motooka, Y.; Sato, Y.; Naito, Y.; Higaki, K.; Wakimoto, J.; et al. Correlation between Histone Acetylation and Expression of Notch1 in Human Lung Carcinoma and Its Possible Role in Combined Small-Cell Lung Carcinoma. Lab. Investig. 2017, 97, 913–921. [Google Scholar] [CrossRef] [PubMed]
  51. Charbat, M.A.; Abdulhalim, Y.H.; Al Rabeai, M.; Abdou Hassan, W.A.M. Role of Notch1 Signaling Pathway in Small Cell Lung Carcinoma. Iran. J. Pathol. 2024, 19, 365–375. [Google Scholar] [CrossRef]
  52. Li, F.; Ye, B.; Hong, L.; Xu, H.; Fishbein, M.C. Epigenetic Modifications of Histone H4 in Lung Neuroendocrine Tumors. Appl. Immunohistochem. Mol. Morphol. 2011, 19, 389–394. [Google Scholar] [CrossRef]
  53. Kim, D.-W.; Kim, K.-C.; Kim, K.-B.; Dunn, C.T.; Park, K.-S. Transcriptional Deregulation Underlying the Pathogenesis of Small Cell Lung Cancer. Transl. Lung Cancer Res. 2018, 7, 4–20. [Google Scholar] [CrossRef]
  54. Na, F.; Pan, X.; Chen, J.; Chen, X.; Wang, M.; Chi, P.; You, L.; Zhang, L.; Zhong, A.; Zhao, L.; et al. KMT2C Deficiency Promotes Small Cell Lung Cancer Metastasis through DNMT3A-Mediated Epigenetic Reprogramming. Nat. Cancer 2022, 3, 753–767. [Google Scholar] [CrossRef] [PubMed]
  55. Agarwal, S.K.; Jothi, R. Genome-Wide Characterization of Menin-Dependent H3K4me3 Reveals a Specific Role for Menin in the Regulation of Genes Implicated in MEN1-Like Tumors. PLoS ONE 2012, 7, e37952. [Google Scholar] [CrossRef]
  56. Lin, W.; Watanabe, H.; Peng, S.; Francis, J.M.; Kaplan, N.; Pedamallu, C.S.; Ramachandran, A.; Agoston, A.; Bass, A.J.; Meyerson, M. Dynamic Epigenetic Regulation by Menin During Pancreatic Islet Tumor Formation. Mol. Cancer Res. 2015, 13, 689–698. [Google Scholar] [CrossRef] [PubMed]
  57. Magerl, C.; Ellinger, J.; Braunschweig, T.; Kremmer, E.; Koch, L.K.; Höller, T.; Büttner, R.; Lüscher, B.; Gütgemann, I. 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. Hum. Pathol. 2010, 41, 181–189. [Google Scholar] [CrossRef]
  58. Klomp, M.J.; Refardt, J.; Van Koetsveld, P.M.; Campana, C.; Dalm, S.U.; Dogan, F.; Van Velthuysen, M.-L.F.; Feelders, R.A.; De Herder, W.W.; Hofland, J.; et al. Epigenetic Regulation of SST2 Expression in Small Intestinal Neuroendocrine Tumors. Front. Endocrinol. 2023, 14, 1184436. [Google Scholar] [CrossRef] [PubMed]
  59. Sponziello, M.; Durante, C.; Boichard, A.; Dima, M.; Puppin, C.; Verrienti, A.; Tamburrano, G.; Di Rocco, G.; Redler, A.; Lacroix, L.; et al. Epigenetic-Related Gene Expression Profile in Medullary Thyroid Cancer Revealed the Overexpression of the Histone Methyltransferases EZH2 and SMYD3 in Aggressive Tumours. Mol. Cell. Endocrinol. 2014, 392, 8–13. [Google Scholar] [CrossRef]
  60. Shenker, N.; Flanagan, J.M. Intragenic DNA Methylation: Implications of This Epigenetic Mechanism for Cancer Research. Br. J. Cancer 2012, 106, 248–253. [Google Scholar] [CrossRef]
  61. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA Expression Profiles Classify Human Cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef] [PubMed]
  62. Yao, Q.; Chen, Y.; Zhou, X. The Roles of microRNAs in Epigenetic Regulation. Curr. Opin. Chem. Biol. 2019, 51, 11–17. [Google Scholar] [CrossRef]
  63. Hussen, B.M.; Hidayat, H.J.; Salihi, A.; Sabir, D.K.; Taheri, M.; Ghafouri-Fard, S. MicroRNA: A Signature for Cancer Progression. Biomed. Pharmacother. 2021, 138, 111528. [Google Scholar] [CrossRef] [PubMed]
  64. Dai, C.; Qianjiang, H.; Fu, R.; Yang, H.; Shi, A.; Luo, H. Epigenetic and Epitranscriptomic Role of lncRNA in Carcinogenesis (Review). Int. J. Oncol. 2025, 66, 29. [Google Scholar] [CrossRef]
  65. McMurry, H.S.; Rivero, J.D.; Chen, E.Y.; Kardosh, A.; Lopez, C.D.; Pegna, G.J. Gastroenteropancreatic Neuroendocrine Tumors: Epigenetic Landscape and Clinical Implications. Curr. Probl. Cancer 2024, 52, 101131. [Google Scholar] [CrossRef]
  66. Demes, M.; Aszyk, C.; Bartsch, H.; Schirren, J.; Fisseler-Eckhoff, A. Differential miRNA-Expression as an Adjunctive Diagnostic Tool in Neuroendocrine Tumors of the Lung. Cancers 2016, 8, 38. [Google Scholar] [CrossRef] [PubMed]
  67. Khan, P.; Siddiqui, J.A.; Kshirsagar, P.G.; Venkata, R.C.; Maurya, S.K.; Mirzapoiazova, T.; Perumal, N.; Chaudhary, S.; Kanchan, R.K.; Fatima, M.; et al. MicroRNA-1 Attenuates the Growth and Metastasis of Small Cell Lung Cancer through CXCR4/FOXM1/RRM2 Axis. Mol. Cancer 2023, 22, 1. [Google Scholar] [CrossRef]
  68. Zhang, C.; Zhou, Y.; Zhang, B.; Sheng, Z.; Sun, N.; Yuan, B.; Wu, X. Identification of lncRNA, miRNA and mRNA Expression Profiles and ceRNA Networks in Small Cell Lung Cancer. BMC Genom. 2023, 24, 217. [Google Scholar] [CrossRef]
  69. Matyasovska, N.; Valkova, N.; Gala, M.; Bendikova, S.; Abdulhamed, A.; Palicka, V.; Renwick, N.; Čekan, P.; Paul, E. Deep Sequencing Reveals Distinct microRNA-mRNA Signatures That Differentiate Pancreatic Neuroendocrine Tumor from Non-Diseased Pancreas Tissue. BMC Cancer 2025, 25, 669. [Google Scholar] [CrossRef]
  70. Pipinikas, C.P.; Berner, A.M.; Sposito, T.; Thirlwell, C. The Evolving (Epi)Genetic Landscape of Pancreatic Neuroendocrine Tumours. Endocr.-Relat. Cancer 2019, 26, R519–R544. [Google Scholar] [CrossRef]
  71. Galasso, L.; Vitale, F.; Giansanti, G.; Esposto, G.; Borriello, R.; Mignini, I.; Nicoletti, A.; Zileri Dal Verme, L.; Gasbarrini, A.; Ainora, M.E.; et al. Decoding Pancreatic Neuroendocrine Tumors: Molecular Profiles, Biomarkers, and Pathways to Personalized Therapy. IJMS 2025, 26, 7814. [Google Scholar] [CrossRef]
  72. Blázquez-Encinas, R.; Moreno-Montilla, M.T.; García-Vioque, V.; Gracia-Navarro, F.; Alors-Pérez, E.; Pedraza-Arevalo, S.; Ibáñez-Costa, A.; Castaño, J.P. The Uprise of RNA Biology in Neuroendocrine Neoplasms: Altered Splicing and RNA Species Unveil Translational Opportunities. Rev. Endocr. Metab. Disord. 2023, 24, 267–282. [Google Scholar] [CrossRef]
  73. Ji, M.; Tang, L.; Ding, R.; Shi, L.; Liu, A.; Chen, D.; Shao, C. Long Noncoding RNA-mRNA Expression Profiles and Validation in Pancreatic Neuroendocrine Neoplasms. Clin. Endocrinol. 2020, 92, 312–322. [Google Scholar] [CrossRef]
  74. Malczewska, A.; Kidd, M.; Matar, S.; Kos-Kudla, B.; Modlin, I.M. A Comprehensive Assessment of the Role of miRNAs as Biomarkers in Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology 2018, 107, 73–90. [Google Scholar] [CrossRef]
  75. Zimmermann, N.; Knief, J.; Kacprowski, T.; Lazar-Karsten, P.; Keck, T.; Billmann, F.; Schmid, S.; Luley, K.; Lehnert, H.; Brabant, G.; et al. MicroRNA Analysis of Gastroenteropancreatic Neuroendocrine Tumors and Metastases. Oncotarget 2018, 9, 28379–28390. [Google Scholar] [CrossRef] [PubMed]
  76. Di Mauro, A.; Scognamiglio, G.; Aquino, G.; Cerrone, M.; Liguori, G.; Clemente, O.; Di Bonito, M.; Cantile, M.; Botti, G.; Tafuto, S.; et al. Aberrant Expression of Long Non Coding RNA HOTAIR and De-Regulation of the Paralogous 13 HOX Genes Are Strongly Associated with Aggressive Behavior of Gastro-Entero-Pancreatic Neuroendocrine Tumors. IJMS 2021, 22, 7049. [Google Scholar] [CrossRef]
  77. Hudson, J.; Duncavage, E.; Tamburrino, A.; Salerno, P.; Xi, L.; Raffeld, M.; Moley, J.; Chernock, R.D. Overexpression of miR-10a and miR-375 and Downregulation of YAP1 in Medullary Thyroid Carcinoma. Exp. Mol. Pathol. 2013, 95, 62–67. [Google Scholar] [CrossRef] [PubMed]
  78. Hashemi, M.; Daneii, P.; Zandieh, M.A.; Raesi, R.; Zahmatkesh, N.; Bayat, M.; Abuelrub, A.; Khazaei Koohpar, Z.; Aref, A.R.; Zarrabi, A.; et al. Non-Coding RNA-Mediated N6-Methyladenosine (m6A) Deposition: A Pivotal Regulator of Cancer, Impacting Key Signaling Pathways in Carcinogenesis and Therapy Response. Non-Coding RNA Res. 2024, 9, 84–104. [Google Scholar] [CrossRef] [PubMed]
  79. Zou, Y.; Bao, X.; Li, D.; Ye, Z.; Xiang, R.; Yang, Y.; Zhu, Z.; Chen, Z.; Zeng, L.; Xue, C.; et al. FTO-Mediated DSP m6A Demethylation Promotes an Aggressive Subtype of Growth Hormone-Secreting Pituitary Neuroendocrine Tumors. Mol. Cancer 2024, 23, 205. [Google Scholar] [CrossRef]
  80. Sakurai, K.; Ochiai, M.; Kojima, K.; Kato, K.; Ando, T.; Kato, T.; Ito, H. The SWI/SNF Chromatin Remodeling Complex Subunit BAF53B as an Immunohistochemical Marker for Neuroendocrine Neoplasms. Hum. Cell 2025, 38, 158. [Google Scholar] [CrossRef]
  81. De Visser, K.E.; Joyce, J.A. The Evolving Tumor Microenvironment: From Cancer Initiation to Metastatic Outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef]
  82. Cives, M.; Pelle’, E.; Quaresmini, D.; Rizzo, F.M.; Tucci, M.; Silvestris, F. The Tumor Microenvironment in Neuroendocrine Tumors: Biology and Therapeutic Implications. Neuroendocrinology 2019, 109, 83–99. [Google Scholar] [CrossRef]
  83. Centonze, G.; Maisonneuve, P.; Mathian, É.; Grillo, F.; Sabella, G.; Lagano, V.; Mangogna, A.; Pusceddu, S.; Bossi, P.; Spaggiari, P.; et al. Digital Immunophenotyping of Lung Atypical Carcinoids and Large Cell Neuroendocrine Carcinomas Identifies Three Subtypes with Specific Tumor-Immune Microenvironment Features. Endocr. Pathol. 2025, 36, 39. [Google Scholar] [CrossRef] [PubMed]
  84. Ye, M.; Xu, L.; Lu, F.; Chen, L.; Hu, C.; Chen, J.; Xue, B.; Gu, D.; Xu, R.; Xu, Y.; et al. Hypoxia Drives CBR4 Down-regulation Promotes Gastroenteropancreatic Neuroendocrine Tumors via Activation Mammalian Target of Rapamycin Mediated by Fatty Acid Synthase. J. Cell Commun. Signal 2024, 18, e12041. [Google Scholar] [CrossRef]
  85. Pradhan, R.; Kundu, A.; Kundu, C.N. The Cytokines in Tumor Microenvironment: From Cancer Initiation-Elongation-Progression to Metastatic Outgrowth. Crit. Rev. Oncol./Hematol. 2024, 196, 104311. [Google Scholar] [CrossRef]
  86. Shin, J.E.; Park, S.H.; Jang, Y.K. Epigenetic Up-Regulation of Leukemia Inhibitory Factor (LIF) Gene During the Progression to Breast Cancer. Mol. Cells 2011, 31, 181–190. [Google Scholar] [CrossRef]
  87. Wang, D.; Zhang, Y.; Li, Q.; Li, Y.; Li, W.; Zhang, A.; Xu, J.; Meng, J.; Tang, L.; Lyu, S. Epigenetics: Mechanisms, Potential Roles, and Therapeutic Strategies in Cancer Progression. Genes. Dis. 2024, 11, 101020. [Google Scholar] [CrossRef] [PubMed]
  88. Lau, T.-S.; Chan, L.K.-Y.; Wong, E.C.-H.; Hui, C.W.-C.; Sneddon, K.; Cheung, T.-H.; Yim, S.-F.; Lee, J.H.-S.; Yeung, C.S.-Y.; Chung, T.K.-H.; et al. A Loop of Cancer-Stroma-Cancer Interaction Promotes Peritoneal Metastasis of Ovarian Cancer via TNFα-TGFα-EGFR. Oncogene 2017, 36, 3576–3587. [Google Scholar] [CrossRef]
  89. Wang, H.-C.; Chen, C.-W.; Yang, C.-L.; Tsai, I.-M.; Hou, Y.-C.; Chen, C.-J.; Shan, Y.-S. Tumor-Associated Macrophages Promote Epigenetic Silencing of Gelsolin through DNA Methyltransferase 1 in Gastric Cancer Cells. Cancer Immunol. Res. 2017, 5, 885–897. [Google Scholar] [CrossRef]
  90. Chen, Q.; Li, Y.; Liu, Y.; Xu, W.; Zhu, X. Exosomal Non-Coding RNAs-Mediated Crosstalk in the Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 646864. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic workflow illustrating the interplay between epigenetic mechanisms and the tumor microenvironment (TME) in neuroendocrine neoplasms (NENs). In NEN cells, characterized by a low mutational burden, epigenetic regulation plays a pivotal role; this includes DNA methylation, histone modifications, and noncoding RNA–mediated mechanisms. These epigenetic alterations are dynamically influenced by components of the TME, such as cancer-associated fibroblasts, immune cells, extracellular matrix, hypoxia, and extracellular vesicles, through bidirectional crosstalk. TME-driven epigenetic reprogramming contributes to tumor progression, therapy resistance, and immune evasion. Targeting epigenetic mechanisms in combination with established therapies—such as somatostatin analogs, peptide receptor radionuclide therapy, targeted agents, and cytotoxic chemotherapy—may overcome resistance mechanisms and improve therapeutic efficacy in NENs (created in https://BioRender.com).
Figure 1. Schematic workflow illustrating the interplay between epigenetic mechanisms and the tumor microenvironment (TME) in neuroendocrine neoplasms (NENs). In NEN cells, characterized by a low mutational burden, epigenetic regulation plays a pivotal role; this includes DNA methylation, histone modifications, and noncoding RNA–mediated mechanisms. These epigenetic alterations are dynamically influenced by components of the TME, such as cancer-associated fibroblasts, immune cells, extracellular matrix, hypoxia, and extracellular vesicles, through bidirectional crosstalk. TME-driven epigenetic reprogramming contributes to tumor progression, therapy resistance, and immune evasion. Targeting epigenetic mechanisms in combination with established therapies—such as somatostatin analogs, peptide receptor radionuclide therapy, targeted agents, and cytotoxic chemotherapy—may overcome resistance mechanisms and improve therapeutic efficacy in NENs (created in https://BioRender.com).
Cancers 18 00069 g001
Figure 2. (A) Different types of altered epigenetic modifications resulting in favored tumor progression in lung NETs; (B) in pancreatic NETs; (C) in gastrointestinal NETs; (D) in thyroid NETs (created in https://BioRender.com).
Figure 2. (A) Different types of altered epigenetic modifications resulting in favored tumor progression in lung NETs; (B) in pancreatic NETs; (C) in gastrointestinal NETs; (D) in thyroid NETs (created in https://BioRender.com).
Cancers 18 00069 g002
Table 1. NET types, WHO subclassification, association with hereditary syndromes/genetic mutations.
Table 1. NET types, WHO subclassification, association with hereditary syndromes/genetic mutations.
Tumor Site Origin Cells Classification Mutated Genes/Hereditary Syndromes
LungsPulmonary neuroendocrine cells (neuroepithelial bodies)
  • Well-differentiated low-grade typical carcinoids (TCs)
  • Well-differentiated intermediate grade atypical carcinoids (ACs)
  • Poorly differentiated high-grade large-cell neuroendocrine carcinomas (LCNECs)
  • Poorly-differentiated high-grade small-cell lung carcinomas (SCLCs) [5]
MEN1, SWI/SNF complex, KMT2/MLL, and PSIP1 [6]
PancreasIslet cells of the pancreas
  • PanNECs
  • Well-differentiated PanNETs [7]
MEN1, VHL, NF-1, tuberous sclerosis complex, and glucagon cell adenomatosis [8,9]
GI tractEnterochromaffin cells of the gut neuroendocrine system
  • NET G1 (well-differentiated, low malignancy)
  • NET G2 (well-differentiated, medium malignancy)
  • NET G3 (well-differentiated, less aggressive than NECs)
  • NECs (poorly differentiated very aggressive) [10]
MEN1, VHL syndrome, NF1, and tuberous sclerosis [11]
ThyroidMainly C-cells of the thyroid gland
  • Follicular cell-derived neoplasms
  • Parafollicular cell-derived tumors [7]
MEN2 syndromes and RET [7]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Castenetto, A.; Gagliano, T. Epigenetics and the Tumor Microenvironment in Neuroendocrine Tumors. Cancers 2026, 18, 69. https://doi.org/10.3390/cancers18010069

AMA Style

Castenetto A, Gagliano T. Epigenetics and the Tumor Microenvironment in Neuroendocrine Tumors. Cancers. 2026; 18(1):69. https://doi.org/10.3390/cancers18010069

Chicago/Turabian Style

Castenetto, Alice, and Teresa Gagliano. 2026. "Epigenetics and the Tumor Microenvironment in Neuroendocrine Tumors" Cancers 18, no. 1: 69. https://doi.org/10.3390/cancers18010069

APA Style

Castenetto, A., & Gagliano, T. (2026). Epigenetics and the Tumor Microenvironment in Neuroendocrine Tumors. Cancers, 18(1), 69. https://doi.org/10.3390/cancers18010069

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop