Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Multiple endocrine neoplasia type 1 (MEN1), a rare tumor syndrome that is inherited in an autosomal dominant pattern, is continuing to raise great interest for endocrinology, gastroenterology, surgery, radiology, genetics, and molecular biology specialists. There have been 2 major clinical practice guidance papers published in the past 2 decades, with the most recent published 8 years ago. Since then, several new insights on the basic biology and clinical features of MEN1 have appeared in the literature, and those data are discussed in this review. The genetic and molecular interactions of the MEN1-encoded protein menin with transcription factors and chromatin-modifying proteins in cell signaling pathways mediated by transforming growth factor β/bone morphogenetic protein, a few nuclear receptors, Wnt/β-catenin, and Hedgehog, and preclinical studies in mouse models have facilitated the understanding of the pathogenesis of MEN1-associated tumors and potential pharmacological interventions. The advancements in genetic diagnosis have offered a chance to recognize MEN1-related conditions in germline MEN1 mutation-negative patients. There is rapidly accumulating knowledge about clinical presentation in children, adolescents, and pregnancy that is translatable into the management of these very fragile patients. The discoveries about the genetic and molecular signatures of sporadic neuroendocrine tumors support the development of clinical trials with novel targeted therapies, along with advancements in diagnostic tools and surgical approaches. Finally, quality of life studies in patients affected by MEN1 and related conditions represent an effort necessary to develop a pharmacoeconomic interpretation of the problem. Because advances are being made both broadly and in focused areas, this timely review presents and discusses those studies collectively.

Free full text 


Logo of edrvLink to Publisher's site
Endocr Rev. 2021 Apr; 42(2): 133–170.
Published online 2020 Nov 28. https://doi.org/10.1210/endrev/bnaa031
PMCID: PMC7958143
PMID: 33249439

Multiple Endocrine Neoplasia Type 1: Latest Insights

Associated Data

Data Availability Statement

Abstract

Multiple endocrine neoplasia type 1 (MEN1), a rare tumor syndrome that is inherited in an autosomal dominant pattern, is continuing to raise great interest for endocrinology, gastroenterology, surgery, radiology, genetics, and molecular biology specialists. There have been 2 major clinical practice guidance papers published in the past 2 decades, with the most recent published 8 years ago. Since then, several new insights on the basic biology and clinical features of MEN1 have appeared in the literature, and those data are discussed in this review. The genetic and molecular interactions of the MEN1-encoded protein menin with transcription factors and chromatin-modifying proteins in cell signaling pathways mediated by transforming growth factor β/bone morphogenetic protein, a few nuclear receptors, Wnt/β-catenin, and Hedgehog, and preclinical studies in mouse models have facilitated the understanding of the pathogenesis of MEN1-associated tumors and potential pharmacological interventions. The advancements in genetic diagnosis have offered a chance to recognize MEN1-related conditions in germline MEN1 mutation–negative patients. There is rapidly accumulating knowledge about clinical presentation in children, adolescents, and pregnancy that is translatable into the management of these very fragile patients. The discoveries about the genetic and molecular signatures of sporadic neuroendocrine tumors support the development of clinical trials with novel targeted therapies, along with advancements in diagnostic tools and surgical approaches. Finally, quality of life studies in patients affected by MEN1 and related conditions represent an effort necessary to develop a pharmacoeconomic interpretation of the problem. Because advances are being made both broadly and in focused areas, this timely review presents and discusses those studies collectively.

Keywords: MEN1, phenocopy, menin, mutation-negative, neuroendocrine tumors, epigenetics, mouse models, pharmacological therapies, surgical approaches, quality of life

ESSENTIAL POINTS

  • The discovered pathways regulated by menin are opening new opportunities for novel therapeutical interventions in multiple endocrine neoplasia type 1 (MEN1)

  • Genetic diagnosis of MEN1 is making possible a distinct management for genetically positive and negative patients

  • Sensitive areas, such as MEN1 clinical course in youngsters and during pregnancy, can be clinically managed by valuing accumulated experiences

  • MEN1-associated pancreatic neuroendocrine tumors are better understood in their pathogenesis and in their impact in the management of affected patients

  • Surgical and pharmacological therapies are opening to a brighter future in the clinical course of MEN1

Multiple endocrine neoplasia type 1 (MEN1) or Wermer syndrome (OMIM *131100) is a rare (prevalence 3-20/100 000) highly penetrant autosomal dominant disorder caused by germline mutations in the tumor suppressor gene MEN1, which encodes a 610 amino acid protein, menin (1, 2). The diagnosis of MEN1 in a patient has relevant implications for family members, as first-degree relatives have a 50% risk of developing the syndrome and can be identified by MEN1 mutational analysis (3). Even though, as an autosomal dominant disorder, a sex dimorphism is not expected in MEN1, a female prevalence has been described (4), but the implications of these findings may need further validation. The age-related penetrance of MEN1 for all clinical features surpasses 50% by age 20 years and 95% by age 40 years (3). Also, instances of geographical clustering as a consequence of founder effects have been reported (5).

MEN1 is characterized by varying combinations of more than 20 endocrine and nonendocrine tumors that show loss of heterozygosity (LOH) at 11q13, the location of the MEN1 gene, resulting in biallelic loss of MEN1 (3, 4, 6-8) (Fig. 1). Endocrine tumors become evident either by hormonal overproduction or by growth of the tumor itself. The diagnosis is clinically suspected by the combined occurrence of 2 or more of the following classical endocrinopathies: primary hyperparathyroidism (PHPT) due to parathyroid glands hyperplasia, anterior pituitary tumors, and duodenopancreatic neuroendocrine tumors (DP-NETs). Other MEN1-associated tumors include thymus and lung NETs, type 2 gastric NETs, adrenocortical tumors, pheochromocytomas, facial angiofibromas, collagenomas, hibernomas, meningiomas, ependymomas, leiomyomas, and lipomas, and an increased risk of developing breast cancer in female patients (9-12). Uncommon neoplasia associated with MEN1, such as carcinoid tumors, mammary cancer, parathyroid carcinoma (13), or adrenocortical carcinoma, are the causes of death among MEN1 patients.

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig1.jpg

Tumors associated with MEN1.

MEN1 patients have a decreased life expectancy, and the outcomes of treatment used in sporadic endocrine tumor counterparts are not as successful because of tumor multiplicity and aggressiveness (3). The prognosis is considerably improved by presymptomatic tumor detection and undertaking treatment specific for MEN1 tumors. This can happen only if clinical care for MEN1 patients and families is provided by a multidisciplinary MEN1-specialists’ team, a true permanent task force fully dedicated to this disorder, as suggested by the last guidance report published in 2012 (3). In the past 8 years several important discoveries have been published about the genetic diagnosis of MEN1, genetics of sporadic endocrine tumors, MEN1-related conditions, the biological functions of menin, potential pharmacological therapies, disease pathogenesis, surgical advances, and the clinical course and prognosis of MEN1. Therefore, a review focusing on these latest findings is certainly timely and useful both for the basic science investigators and clinicians to understand the molecular basis of MEN1 and for the management of MEN1 patients.

New Findings About the MEN1–Encoded Protein Menin and its Functional Characterization

Menin, the protein product of the MEN1 gene, consists of 610 amino acids (menin isoform 2, NCBI reference sequence: NM_130799.2). There is a very rare minor isoform of menin (menin isoform 1, NCBI reference sequence: NM_000244.3), from a potential alternative splice site 15 bp downstream of exon-2 that inserts 5 amino acid residues (after amino acid 148). In the gene and protein databases, the norm is to designate the longest transcript as the primary isoform; therefore, sometimes menin is described as a 615 amino acid protein.

All studies of menin and its mutants have been conducted with the 610 amino acid isoform because the rare 615 amino acid isoform has not been observed in any cell types. The 67 kDa menin is widely expressed, and at the C-terminus contains 2 nuclear localization signals (NLSs) (NLS1: 479–497 and NLS2: 588–608) and 1 accessory NLS (aNLS: 546-572) (9, 14). Therefore, menin is detected in the nucleus as shown from experiments using green fluorescent protein–tagged menin, immunofluorescence, and Western blot analysis of subcellular fractions (9, 14). Posttranslational modifications of menin include phosphorylation at Ser394, Thr397, Thr399, Ser487, Ser543, and Ser583, SUMOylation, and palmitoylation that may enhance or suppress menin’s action as a tumor suppressor in the nucleus or its potential association with the cell membrane (15-17). The functional contribution of these posttranslational modifications has not been studied in MEN1-associated endocrine cell types or in a clinical context.

The 3-dimensional (3D) crystal structure of human menin (Protein Data Bank No. 3U84) has been successfully deciphered after the deletion of a single internal loop region that was predicted to be unstructured (amino acid residues 460-519) (18). The 3D structure of menin resembles a “curved left hand,” with a pocket formed by the “thumb” and the “palm.” The structure consists of 4 domains: a long β-hairpin N-terminal domain, a transglutaminase-like domain (“thumb”), a helical domain that contains 3 tetratricopeptide motifs (“palm”), followed by a C-terminal domain (“fingers”) (Fig. 2). The pocket or cavity formed by the “thumb” and the “palm” has been shown to facilitate protein-protein interactions (18, 19).

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig2.jpg

Three-dimensional crystal structure of human menin alone or together with interacting partners. A, Structure of menin showing a pocket/cavity for protein-protein interaction (Protein Data Bank; PDB ID 3U84). The different domains of menin are color coded: “N-terminus” in pale green (1-101), “thumb” in green-cyan (102-230), “palm” in olive green (231-386), and “fingers” in dark green (387-end). B, Structure of menin interacting with the menin-binding motif (MBM) of JUND (amino acid 27-47, in purple) (PDB ID 3U86). C, Structure of menin interacting with the MBM of MLL1 (amino acid 6-13, in gold) (PDB ID 3U85). D, The ternary complex of menin with interacting regions in LEDGF (amino acid 347-435, in red) and MLL1 (MBM-LEGDF binding motif [LBM], amino acid 6-153, in gold) (PDB ID 3U88). To facilitate crystallization, the following regions were deleted (and not present in the structures shown in this figure): an unstructured loop (amino acid 460-519) in menin, a short fragment (amino acid 40-45) in the JUND-MBM and 2 loop regions (amino acids 16-22 and 36-102) in the MLL1-MBM-LBM. Structural images were generated by using PyMOL (Schrodinger Inc; https://pymol.org).

The tumor-suppressor role of menin in MEN1 and the tissue restricted pattern of MEN1-associated tumors has been replicated in mouse models. Germline homozygous knockout of Men1 (Men1–/–) is embryonic lethal at E11.5 to E14.5, and germline heterozygous knockout of Men1 (Men1+/–) generates viable mice that develop (at age > 12-15 months) hormone-hypersecreting tumors in the pancreatic islets (mainly insulinoma), anterior pituitary (mainly prolactinoma), and the parathyroid glands (mainly hyperplasia) (20-24). Consistent with the tumor suppressor role of menin in human MEN1 tumors, a second hit to the nontargeted copy of Men1 resulting in LOH is essential for tumor formation in Men1+/– mice. The pancreatic islets of Men1+/– mice show a pretumor stage of hyperplasia and dysplasia prior to LOH at the Men1 locus (25). Investigating the molecular aspects involved in these pretumor events can be helpful to understand tumor initiation and progression from tissue-specific menin haploinsufficiency and menin loss.

Tissue-specificity of the tumor suppressor role of menin has been shown in 2 conditional mouse models. First, mice with conditional loss of menin in the liver (Men1f/f;ALB-Cre) do not develop tumors in the liver (26). Second, mice with conditional loss of menin in the whole pancreas (Men1f/f;PDX1-Cre) develop tumors that originate from the β cells of endocrine pancreas (insulinoma), and not from any cells of exocrine pancreas (27). Interestingly, mice with conditional loss of menin in the glucagon-secreting pancreatic islet α cells (Men1f/f;GLU-Cre) do not develop glucagonomas, instead they predominantly develop β-cell tumors (insulinomas) (28, 29). It is possible that after menin loss, α cells may transdifferentiate into β cells, or paracrine signals from menin-null α cells induce β-cell proliferation (28, 29). As expected, parathyroid-specific Men1-knockout mice (Men1f/f;PTH-Cre) develop parathyroid hyperplasia and hypercalcemia, and pancreatic islet β-cell–specific Men1-knockout mice (Men1f/f;RIP-Cre) develop insulinomas (25, 30-32). The Men1f/f;RIP-Cre mice also develop prolactinomas due to the leaky expression of the RIP-Cre transgene in pituitary lactotroph cells. Similar to human MEN1, the prolactinomas in mice (Men1+/– or Men1f/f;RIP-Cre) are frequently observed in females. However, the reason for the sex bias of prolactinomas remains unknown. Although the conditional Men1-knockout mice do not depend on a spontaneous second hit for LOH of Men1, tumors develop 8 to 10 months after embryonic menin loss, and the basis for the delay in tumor formation is not known.

The functional characterization of menin has encountered various challenges due to the lack of any similarity to known proteins, lack of obvious functional motifs/domains, lack of normal or menin-null endocrine cell lines, and lack of ex vivo models of MEN1 tumors or their counterpart normal tissues (organoids or patient-derived xenograft [PDX]). Insights into how menin performs its tumor suppressor activity have been gained from the identification of interacting partners of menin in cell types unrelated to MEN1-associated tissues, followed by validation of some relevant targets in translational studies (33). Even though the sequence of menin does not reveal any functional attributes, direct or indirect interactions with more than 50 different proteins of known function have helped to provide clues about its role in various processes and pathways: cell adhesion, cell cycle progression, cell division, cell motility, cell signaling, cytoskeletal structure, DNA repair, genomic stability, and transcriptional regulation (34). Highly enriched among the interacting partners of menin are transcription factors and epigenetic regulators. Menin serves as a multifunctional protein through prominent functional contributions in transcriptional regulation as a coactivator or corepressor.

Functional contributions of menin in transcriptional regulation

The interactions of menin with various transcription factors and chromatin-modifying proteins have shown a significant functional contribution in cell signaling pathways mediated by transforming growth factor β (TGF-β)/bone morphogenetic protein (BMP), nuclear receptors, Wnt/β-catenin, and Hedgehog (Hh) (35, 36). These signaling pathways stimulate transcription factor recruitment to their cognate DNA binding sites to regulate gene expression. Menin interacts with SMAD3 or SMAD1/SMAD5 to promote their transcriptional activity, and loss of menin in these interactions inhibits TGF-β and BMP signaling pathways, respectively, thus antagonizing their proliferation-inhibitory effects. Nuclear receptors are transcription factors that are activated by binding to ligands such as steroid hormones. Menin has been shown to act as a coactivator of gene expression mediated by some nuclear receptors (AR, ERα, LXRα, PPARα, PPAR[Upsilon], RXR, and VDR), and loss of menin in these interactions predicts suppression of specific nuclear receptor signaling affecting cell growth and function. Conversely, menin interacts with β-catenin and its associated transcription factors TCF3 and TCF4 to suppress their activity, and loss of menin promotes Wnt/β-catenin signaling that is known to increase islet β-cell proliferation. Menin interacts with PRMT5 to antagonize Hh signaling by depositing a PRMT5-dependent repressive histone modification (H4R3me2s) to suppress the expression of genes in the Hh signaling pathway, GAS1 and GLI1. Loss of menin-PRMT5–mediated repressive marks in these genes would promote Hh signaling that can upregulate cell proliferation.

Among the AP1 family of JUN transcription factors (JUNB, cJUN, and JUND) that regulate gene expression downstream of various stimuli, menin interacts only with JUND and suppresses its transcriptional activity (37, 38). The menin interacting region of JUND maps to its N-terminus. Synthetic mutations in this region of JUND (human JUND amino acid residues 33-36) can disrupt interaction with menin. Such mutants of JUND that lack menin interaction are oncogenic because they promote cell proliferation, consistent with a tumor suppressor effect of the menin-JUND interaction (39).

Menin interacts with DAXX (a transcriptional repressor and component of chromatin remodeling complex) and SUV39H1, a histone methyltransferase that deposits a repressive histone modification H3K9me3, to repress the transcription of specific genes associated with the regulation of cell proliferation: MME, GBX2, and IL6 (40).

H3K4me3 is a histone modification that is primarily located in promoter regions near the transcriptional start site to activate gene transcription. Among the MLL family members that are responsible for depositing this histone mark, menin interacts with 2 histone methyltransferases MLL1 (KMT2A) and MLL2 (KMT2B). MLL1 and MLL2 are part of multisubunit protein complexes that contain ASH2L, hDPY30, HCF-2, RBBP5, and WDR5, and they also interact with the 140 kDa subunit of RNA-Pol-II (POLR2B) (41, 42). Loss of menin in this protein complex results in the transcriptional repression of specific genes due to gene-specific loss of H3K4me3 in the promoter region near the transcriptional start site (42).

The 3D structure of menin shows a central cavity that forms a binding pocket for protein interaction but no obvious DNA binding domain, indicating that to control gene expression menin does not directly bind to DNA and is dependent on its interactions with components of the transcriptional regulatory machinery (18, 19). Cocrystallization of menin with peptides from the interacting region of JUND or MLL1 have shown that their binding to the pocket region of menin is mutually exclusive (see Fig. 2) (18). Interestingly, the menin-binding peptides of JUND and MLL1 are almost identical at the 5 residues that are critical for binding to menin. Also, cocrystallization of menin with a peptide from the lens epithelium–derived growth factor (LEDGF) (amino acids 347-429) and MLL1 (amino acids 6-153, which contain motifs for menin-binding and LEDGF-binding) has shown that the interaction between menin and MLL1 creates an interface for binding LEDGF that is a protein that directs the MLL-complex to chromatin (see Fig. 2) (18). Similar structural studies of menin with other interacting partners may help to determine how menin can interact one on one with individual factors or simultaneously with multiple factors to control transcriptional regulation.

Characterization of menin’s target genes and role in cell proliferation

Genome-wide analysis of target genes using a menin antibody by techniques such as chromatin immunoprecipitation coupled with DNA microarray chips (ChIP-chip) or coupled with next-generation sequencing (ChIP-seq), or serial analysis of chromatin occupancy has shown that menin is localized to hundreds of genes near promoter regions and other regions in the genome (35, 43, 44). Whether all or some of these genes are relevant to the role of menin as a tumor suppressor has not been determined. One target gene that was identified in the study by Scacheri et al is Hlxb9/Mnx1, encoding an embryonic transcription factor responsible for islet β-cell differentiation (43). They compared ChIP-chip data of menin occupancy in human islets to gene expression data from islets of 15- and 25-week-old mice that were wild type (WT) or menin-null (Men1f/f;RIP-Cre). Hlxb9 was one of the genes identified among the few genes that were both bound by menin and altered in expression in menin-null islets. The expression of Hlxb9 was higher in the menin-null islets. This finding supports the notion that tissue-specificity of MEN1-associated tumors may be related to the regulation of tissue-specific target genes by menin such as Hlxb9 in islet β cells. Subsequent studies have shown menin-dependent regulation of a few β-cell differentiation factors (Foxa2, Nkx2.2, MafA, MafB, and Hlxb9) (45-48).

Given the association of menin in the MLL complex for depositing H3K4me3 in chromatin, another approach to characterize menin’s target genes is to analyze WT and menin-null cells for H3K4me3 profiles by ChIP-chip or ChIP-seq coupled with complementary DNA (cDNA) microarray analysis for differential gene expression. This approach in WT and menin-null mouse embryonic stem cells identified the long noncoding RNA Meg3 as a menin target gene that acts as a tumor suppressor, and loss of MEG3 expression has been reported in human sporadic pituitary adenomas (49-51). Similar analysis of WT or menin-null mouse embryonic fibroblasts (MEFs), and pancreatic islet-like endocrine cells derived by in vitro differentiation of WT or menin-null mouse embryonic stem cells, have shown Hox genes as targets of menin (42, 49). Menin loss suppresses the expression of Hox genes. The Hox genes encode essential transcription factors for embryonic development and tissue homeostasis, and their aberrant expression has been reported in various cancer types.

One of the obvious processes that could be dysregulated to promote increased cell proliferation in tumors on loss of menin is the cell cycle. Various proteins that are involved in the progression, maintenance and regulation of the cell cycle include cyclins, cyclin-dependent kinases (CDK), cyclin-dependent kinase inhibitors (CDKI), and tumor suppressors p53 and RB. Studies using menin-null MEFs have shown accelerated progression from G0/G1 to S phase, lower expression of 2 CDKI genes Cdkn2c (p18) and Cdkn1b (p27), and increased Cdk2 activity compared to WT MEFs or menin-null MEFs reconstituted with menin expression (52, 53). Similarly, lower expression of p18 and p27, and higher expression of Cdk4 has been observed in islet tumors of Men1+/– mice and in hyperplastic islets of mice with acute deletion of Men1 (Men1f/f;Cre-ER fed tamoxifen) compared to islets from WT mice (53, 54). Further studies in mouse models with knockout of each of these cell cycle genes (Rb1, Tp53, Cdk2, Cdk4, Cdkn2c, and Cdkn1b) in the Men1+/– background have shown that p18 inactivation and Cdk4 activation may be critical for islet tumor formation after menin loss (Table 1) (55-59).

Table 1.

Mouse models of menin loss to study functional interactionsa

Double knockout with conventional Men1+/–
GenotypePhenotype (islet tumorigenesis) Men1 LOH
Rb1  +/–Similar to Men1+/–Yes
Tp53  +/–Similar to Men1+/–Yes
Cdkn2c  –/–AcceleratedNo
Cdkn1b  –/–Similar to Men1+/–Yes
Cdk2  –/–Similar to Men1+/–Yes
Cdk4  –/–No tumors (islet and pituitary hypoplasia from Cdk4–/–)No
Double knockout with conditional menin loss in β cells (Men1f/f;RIP-Cre)
GenotypePhenotype (islet tumorigenesis and survival)
Pten  f/fAccelerated tumorigenesis and shorter lifespan
Kmt2a  f/fAccelerated tumorigenesis and shorter lifespan
Kdm5a  f/fDecreased tumorigenesis and prolonged survival
Ctnnb1  f/fDecreased tumorigenesis and prolonged survival
Inhbb  –/–No effect on tumorigenesis but prolonged survival
Oncogenic KRas mutant with conditional menin deficiency in β cells (Men1+/f;RIP-Cre)
GenotypePhenotype (islets of P5 neonates)
KRas(G12D) KRas(G12D) expression enhanced rather than inhibited β-cell proliferation

a References are cited in the main text.

Although the full spectrum of specific target genes of menin from interaction with various transcription factors that are relevant to MEN1-associated tumor cell proliferation remain to be determined, translational studies in mouse models have shown the importance of genetic interaction between Men1 and Pten, Kmt2a (Mll1), Kdm5a (Rbp2), Ctnnb1 (β-catenin), Inhbb (activin-B) or oncogenic Kras in islet tumors and β-cell proliferation (see Table 1) (60-65). Combined genetic manipulation of these genes in mice with β-cell-specific menin loss (Men1f/f;RIP-Cre) has shown that loss of Pten or Kmt2a accelerates islet tumor formation and reduces survival, loss of Ctnnb1 or a histone demethylase (Kdm5a) decreases islet tumor formation and prolongs survival, loss of Inhbb does not affect tumor formation but prolongs survival after age 10 months, and expression of activated Kras(G12D) enhances rather than inhibits β-cell proliferation.

One approach that has not been explored to identify target genes of menin dysregulated on menin loss in tumors is single-cell RNA sequencing analysis of MEN1-associated tumor cells and their counterpart normal cells.

Unexpected function of menin as a pro-oncogenic factor in MLL-rearranged leukemia and its potential therapeutic applications

The discovery of menin’s interaction with MLL1 exposed a surprising functional contribution of menin as an oncogenic cofactor of MLL-fusion proteins that drive an aggressive form of leukemia (41). The MLL1/KMT2A gene on chromosome 11q23 is involved in chromosomal translocations in 10% of acute leukemias with the N-terminus of MLL1 fused to the C-terminus of more than 80 different fusion partners. The most common among the translocations are t(4,11)(q21;q23), t(9,11)(p21;q23), and t(11,19)(q23;p13) leading to the expression of MLL-AF4, MLL-AF9, and MLL-ENL fusion proteins (66). The MLL rearranged (MLLr) leukemias are a distinct subset of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) that affect children and adults.

MLL1 is required to maintain the expression of HOX family genes, which regulate normal hematopoietic differentiation. Oncogenic MLL-fusion proteins cause acute leukemia because they upregulate the expression of HOX genes, including HOXA7, HOXA9, and a HOX cofactor, MEIS1, which enhances the proliferation of hematopoietic stem cells and blocks hematopoietic differentiation. MLL fusions that drive MLLr leukemia retain the menin-interacting part of MLL, and interaction of menin with the MLL-fusion protein is critical for the maintenance of the MLL-fusion–driven gene expression program (41). Therefore, blocking the interaction of MLL in the central cavity/pocket of menin with small molecules suggested an important therapeutic strategy for the treatment of MLLr leukemias. Over the years, structurally optimized chemical design has resulted in the development of several small molecule inhibitors of menin-MLL interaction that have greater potency and specificity, that are orally bioavailable, and with improved pharmaceutical properties (67-70). These inhibitors have been tested in various experimental model systems in which they block hematopoietic stem cell proliferation and promote differentiation—mouse models of MLLr leukemia, patient-derived leukemia cell lines, and PDX models of these cell lines and human primary leukemia cells. The promising results from these studies have translated into ongoing phase 1 and 2 clinical trials for 2 compounds: Kura Oncology (KO)-539, a structurally related analogue of MI-3454 (NCT04067336), and Syndax (SNDX)-5613, a close analogue of VTP-50469 (NCT04065399). These compounds can also block the interaction of menin and WT MLL, indicating a therapeutic role in non-MLLr leukemias. In a mouse model of non-MLLr leukemia in which AML development is dependent on a mutation in the nucleophosmin (NPM1) gene (seen in 30% of AML patients), VTP-50469 elicited a cytotoxic effect in preleukemia AML cells, suggesting the potential for preventive therapy (71).

Menin-MLL interaction inhibitors have also been tested in experimental models of solid tumors in which menin has been shown to act as a pro-oncogenic cofactor to block the interaction between menin and WT MLL. An earlier menin-MLL inhibitor, MI-2, has been shown to inhibit tumor cell growth of pediatric gliomas with a histone H3.3(p.Lys27Met) mutation (72). Antitumor effects of another menin-MLL inhibitor, MI-503, has been demonstrated in castration-resistant prostate cancer, Ewing sarcoma, and hepatocellular carcinoma (68). The utility of menin–MLL interaction inhibitors in MEN1-associated tumor cells are potentially irrelevant because menin acts as a tumor suppressor in the context of MEN1 and the tumors show biallelic menin loss or inactivation. However, these inhibitors have not been tested in sporadic endocrine tumors that retain WT menin, or sporadic tumors with specific menin missense mutations that retain interaction with MLL (and without MEN1 LOH). Availability of experimental models of such tumors could help to determine whether menin-MLL interaction inhibitors enhance or block the proliferation of endocrine tumor cells.

Role of menin as a tumor suppressor in nonMEN1 target tissues

Approximately, 45% to 50% of BRAF mutation–positive colorectal cancers show abnormal regulation of the WNT pathway. In a recent study, a somatic inactivating hotspot mutation at codon R516 in the MEN1 gene (R521 as per menin isoform 1) was detected in 4% of BRAF-mutant colorectal neoplasia samples (73). These data support a role of menin in colorectal tissues as a tumor suppressor and adds another WNT pathway-associated gene to the pathology of colorectal cancer, given that menin has been shown to participate in the regulation of the WNT pathway. In another recent study that investigated the germline susceptibility of patients with apparently sporadic osteosarcoma (malignant bone tumors), a higher than expected frequency (0.5%) of pathogenic/likely pathogenic variants of the MEN1 gene were observed in patients of European ancestry (74). These findings have important implications for the genetic testing of osteosarcoma patients and suggest a role of menin as a tumor suppressor in bone. However, osteogenic carcinomas are not described in MEN1 patients.

Epigenetic Regulation in the Tumorigenesis of MEN1–Associated Endocrine Tumors

DNA is wrapped around a histone octamer with 2 copies each of the 4 core histone proteins (H2A, H2B, H3, and H4), to form a nucleosome, which is the basic unit of chromatin. Epigenetic modifications to DNA and histone proteins can impart a closed or open chromatin structure for controlling access to the transcriptional machinery and to control other processes such as DNA replication and repair. Various epigenetic factors form multiprotein complexes, which may also include long noncoding RNAs, to function as enzymes or cofactors for “writing,” “reading,” or “erasing” the modifications on DNA and histones. DNA modifications include methylation, hydroxymethylation, and further oxidation. Posttranslational modifications of histones known as histone “marks” include methylation, acetylation, phosphorylation, ubiquitination, and other modifications. The precise regulation of these epigenetic modifications and their control mechanisms is essential to prevent abnormal cell proliferation and function that can lead to neoplasia and other conditions. Because epigenetic modifications can be written, read, and erased, they offer a therapeutic opportunity to restore aberrant epigenetic changes to the normal state with drugs that can block or enhance the enzymatic activity or critical interactions of epigenetic regulators.

Epigenetic events in MEN1–associated tumors

Various epigenetic changes have been reported in MEN1-associated tumors (Fig. 3). Evidence for a role of epigenetic regulation in the tumors of MEN1 is supported by the interaction of menin with histone-modifying proteins, particularly with histone lysine methyltransferases (KMTs) MLL1/KMT2A and MLL2/KMT2B in protein complexes that are responsible for writing the histone mark H3K4me3 (40, 41). Histone methyltransferases methylate lysine or arginine residues in the chain of amino acids that protrude from the nucleosome (histone tail). Histone H3 can be methylated on lysine residues at positions 4, 9, 27, 36, and 79 with 1, 2, or 3 methyl groups. Activation or silencing of gene expression is regulated by the level of methylation or demethylation of specific histone H3 lysine residues. H3K4me3 is a mark of actively transcribed genes, and H3K9me3 and H3K27me3 are associated with transcriptional silencing. Specific lysine demethylases (KDMs) can erase monomethylation, demethylation, or trimethylation of H3K4 or H3K9, such as lysine-specific demethylase 1 (LSD1/KDM1A), lysine-specific demethylase 2 (LSD2/KDM1B), and Jumonji AT-rich interacting domain 1A (JARID1A/KDM5A/RBP2).

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig3.jpg

Schematic representation of epigenetic regulation in multiple endocrine neoplasia type 1 (MEN1)-associated tumors. Epigenetic modification in normal cells is shown on the left. Aberrant epigenetic change observed or predicted after menin loss in tumors is shown in the middle. Consequence of the aberrant epigenetic change is shown on the right. Green and red indicates the nature of the specific histone or DNA epigenetic modification, active or repressive mark of gene expression, respectively. Open black oval with a slanted line indicates loss of that histone mark. Alternative lengthening of telomeres (ALT) is activated in tumors and it is absent in normal cells (indicated by the blue open oval with a slanted line). Not shown is miR-24–mediated epigenetic regulation.

Epigenetic regulation in MEN1-associated islet tumors from H3K4me3 has been explored in mice with targeted β-cell–specific menin loss (Men1f/f;RIP-Cre). Genome-wide distribution of the gene activation mark H3K4me3 and its counterpart recessive mark H3K27me3 has been examined in the pancreatic islets of 2-month-old Men1f/f;RIP-Cre and control RIP-Cre mice (75). Immunohistochemistry with anti-H3K4me3 showed no significant change in the global/overall level of H3K4me3 in menin-null islets compared with control islets. In menin-null islets, loss of H3K4me3 correlated with gain of H3K27me3 within a specific set of genes, and the expression of such genes was significantly decreased compared to control islets, particularly the gene encoding insulin-like growth factor 2 messenger RNA binding protein 2 (Igf2bp2). Interestingly, the altered epigenetic marks (loss of H3K4me3 and gain of H3K27me3) and lower expression of Igf2bp2 could be reversed in menin-null islets with simultaneous deletion of the H3K4me3 demethylase Rbp2 (Men1f/f;Kdm5af/f;RIP-Cre) (75). Immunohistochemistry with anti-H3K4me3 showed no significant change in the global/overall level of H3K4me3 in menin-Rbp2-null islets compared with control islets. The mice with β-cell–specific combined loss of menin and Rbp2 showed a decreased rate of islet tumor formation and prolonged survival (62). Therefore, specific epigenetic changes occurring as a consequence of menin loss could be reversed (by removing the Rbp2 histone demethylase), and the restoration of the epigenetic changes to the basal normal state could also reduce tumor formation.

The epigenetic regulation of MEN1-associated islet tumor cell proliferation has been explored for the interaction of menin with an arginine methyltransferase PRMT5 that deposits a repressive histone mark, H4R3me2s (36). In MEFs, menin together with PRMT5 was shown to repress the expression of the Gas1 gene, which is involved in the activation of the Hh signaling pathway. GAS1 is required for binding of the Sonic Hedgehog ligand to its receptor PTCH1 for the stimulation of Hh signaling. Therefore, in tumors with menin loss, GAS1 repression would be released to activate Hh signaling. Treatment of 8-month-old Men1f/f;RIP-Cre mice with the Hh inhibitor GDC-0449 for 4 weeks reduced the proliferation of islet β cells by approximately 60%. The effect on tumor size and overall survival was not investigated. This study showed that blocking aberrant signaling due to the loss of a menin-dependent epigenetic mark could inhibit cell proliferation.

Another histone modification that has been studied in MEN1-associated islet tumors is histone acetylation that is associated with active transcription. Acetylation marks at lysine residues in histone tails are deposited by histone acetyl transferases (HATs) and read by the bromodomain contained in the bromodomain and extraterminal (BET) proteins. JQ1 is a small-molecule inhibitor of bromodomain interactions with acetylated histones. Treatment of 30-week-old Men1f/f;RIP-Cre mice with twice-weekly injections of JQ1 for 1 month reduced the proliferation rate of islet β cells in the tumors by 49% to 55% and significantly increased apoptosis (76). The effect on tumor size and overall survival was not assessed in this study. Although the specific epigenetic changes in histone acetylation have not been investigated in MEN1-associated tumors, this study highlights the potential of targeting histone acetyl marks.

A few studies have investigated DNA methylation in MEN1-associated tumors, which is an epigenetic modification of CpG sites, particularly in gene promoter regions. DNA hypermethylation usually coincides with gene silencing. Also, H3K4me3 has been shown to protect CpG islands from DNA methylation to regulate gene transcription (77). DNA methylation can be blocked by directly inhibiting DNA methyltransferases (DNMTs) that establish, propagate, and maintain the stability of the DNA methylation mark. DNA hypermethylation was detected in a subset of MEN1 tumors. Global DNA hypermethylation was detected in parathyroid tumors and nonfunctioning pancreatic neuroendocrine tumors (pNETs) from MEN1 patients (78, 79). Also, promoter hypermethylation was observed as a frequent event in MEN1-associated advanced pNETs (80). Using a somatic gene transfer system in RIP-TVA mice, expression of DNMT1 increased β-cell proliferation, suggesting that DNMT1 could be targeted to inhibit DNA hypermethylation and β-cell proliferation (81).

MEN1-associated pNETs have been assessed for telomere length. Telomeres are specialized chromatin structures that protect chromosome ends. Alternative lengthening of telomeres (ALT) is a telomerase-independent process that is activated in cancer cells to prevent telomere shortening that accompanies normal proliferation of somatic cells. The correlation between ALT and prognosis is variable in different cancer types. Mutations in chromatin remodeling genes death domain-associated protein (DAXX) and α-thalassemia/mental retardation X-linked (ATRX) correlate with ALT activation in sporadic pNETs (82-85). In a study of nonfunctioning pNETs, 48% of sporadic and 25% of MEN1-associated pNETs were ALT positive, and ALT was associated with disease relapse (85).

Noncoding RNA (ncRNA)-mediated gene silencing is another epigenetic mechanism that has been investigated in MEN1-associated tumors. There are 2 types of ncRNAs: the short ncRNAs (fewer than 30 nucleotides) and the long ncRNAs (greater than 200 nucleotides). MicroRNAs are short ncRNAs that regulate gene expression at the transcriptional and posttranscriptional level. In the hyperplastic islets of 8-week-old Men1f/f;RIP-Cre mice, and human parathyroid tumors, miR-24 (and its immature form miR-24-1) has been shown to target menin because increased miR-24 level correlated with decreased menin level (86-88). This mechanism of silencing menin may also contribute to the second somatic “hit” of MEN1 inactivation in MEN1-associated tumors that do not show LOH at the MEN1 locus (86).

Exploring epigenetic diagnostic and therapeutic options

Epigenetic changes are stable in tumors and thus can be used as diagnostic markers. Also, epigenetic changes are reversible and can be targeted to restore normal epigenetic states such as by blocking the enzymatic activity of epigenetic regulators, disrupting specific interactions in chromatin-modifying protein complexes, interfering with the reading of epigenetic marks, or targeting specific epigenetic factors for degradation.

Evidence for epigenetic changes in MEN1-associated tumors in Men1f/f;RIP-Cre mice or human tumor samples indicates a potential for exploring epigenetic alterations as biomarkers for diagnostic and therapeutic options for these tumors. Among the epigenetic alterations that can occur in MEN1-associated tumors are loss of the active histone mark H3K4me3 in a subset of genes, gain of the repressive histone H3K27me3 in a subset of genes, enhanced Hh signaling due to loss of a repressive histone mark H4R3me2s in the promoter region of Gas, histone acetylation, DNA hypermethylation, ALT, and micro RNA-mediated silencing of menin expression (Fig. 3). Whether the epigenetic changes to DNA and histones, and ALT occur simultaneously in MEN1-associated tumors has not been investigated.

Experimental evidence for the reversal of changes to histone methylation in the tumors of Men1f/f;RIP-Cre from combined loss of a demethylase Rbp2 indicates a therapeutic opportunity with epigenetic drugs to inhibit this demethylase in MEN1-associated tumors (62). Similarly, the effect of a BETi, JQ1, to lower cell proliferation in the tumors of Men1f/f;RIP-Cre indicates that targeting histone acetylation can be further explored as a potential epigenetic therapeutic option in MEN1-associated tumors (76).

Epigenetic changes to DNA from advanced tumors can be measured in circulating cell free DNA (cfDNA) in blood and serum samples (89). Because epigenetic changes are stable in tumors and because of the noninvasive sample acquisition for cfDNA, this potential diagnostic assay (cfDNA liquid biopsy) is being explored for various cancers but has not yet been applied in a clinical setting for any type of cancer. Therapeutic options for targeting DNA hypermethylation in tumors are DNA hypomethylating agents, decitabine and azacytidine, that target DNA methylating enzymes. These drugs have been approved by the US Food and Drug Administration for the treatment of specific hematological malignancies and can be explored as potential therapeutic options in experimental models of MEN1-associated tumors that show DNA hypermethylation.

Telomere-specific fluorescence in situ hybridization has been used to determine ALT status in human tumor samples and may serve as a diagnostic assay in tissue biopsies (85). Potential therapeutics targeting ALT have been proposed (90). Results from miR-24–mediated silencing of the WT MEN1 allele in human parathyroid tumors without 11q13 LOH can be further investigated to develop RNA antagomir-based strategies to restore the expression of menin from the nonmutant copy of the MEN1 gene to control tumorigenesis (86).

Development and use of epigenetic-based therapeutics continue to face several issues and challenges such as cell-specific targeting in affected tissues, side effects, drug resistance, and achieving constant, consistent, and long-lasting effect on the target. Although several epigenetic drugs are in clinical trials, these challenges need to be overcome for translating drug discovery to human patients (91).

Novel Insights Into the Genetics of MEN1

The current germline or somatic MEN1 genetic testing consists of DNA sequence analysis to screen coding exons and splice junctions for mutations, and multiplex ligation-dependent probe amplification for deletion/duplication (del/dup) analysis to screen for larger alterations. Since its discovery, the availability of genetic testing of the MEN1 gene has become an essential part in the diagnosis and management of MEN1. Genetic screening in MEN1 is informative to confirm the clinical diagnosis, for carrier ascertainment, and for early monitoring for tumors. Also, in families with clinical and genetic MEN1, relatives with a negative MEN1 genetic test can be excluded from the burden of lifelong tumor monitoring.

Clinical practice consensus guidelines developed by a panel of experts including physicians, surgeons, geneticists, and other specialists from international centers outlined recommendations for genetic testing in MEN1 (3). The current guidelines recommend that genetic counseling must be available to patients before and after genetic testing. In terms of who should be tested, the guidelines state that the test should be offered to: 1) an index case with clinical MEN1 (presenting with 2 or more MEN1-associated endocrine tumors), 2) asymptomatic first-degree relatives of an individual with genetic MEN1 (known MEN1-mutation carrier) as early as before age 5 years, 3) symptomatic first-degree relatives of an individual with genetic MEN1 who are presenting with at least one MEN1-associated tumor, and 4) patients with multigland parathyroid disease or parathyroid adenomas before age 30 years, and gastrinoma or multiple pancreatic islet tumors at any age.

Sequencing and del/dup analysis can identify heterozygous MEN1 germline mutations in 70% to 90% of families with typical features of MEN1. A 2015 review of published germline mutations identified 576 unique mutations, and in 2019 the Universal Mutation Database of MEN1 mutations (UMD-MEN1 database) reported an additional 181 unique germline mutations (92, 93). These 757 unique MEN1 germline mutations cover the entire coding region with no hot spots.

The obviously pathogenic category of germline mutations (69%) predicts premature truncation of menin from nonsense mutations (14%), frame-shift mutations (42%), splice site mutations (10.5%), and large deletions (2.5%) (Fig. 4) (92). Missense mutations (25.5%) and in-frame insertion or deletion (indel) of one or more amino acids (5.5%) do not predict obvious inactivation of menin, and whether they are benign or pathogenic needs further investigation (92). Across multiple studies and among family members, no clear genotype-phenotype correlation has emerged from an analysis of mutation types or their location with the clinical manifestations of MEN1 (94). Similarly, somatic MEN1 mutations in sporadic tumors have not revealed any hot spots or a clear genotype-phenotype correlation with specific tumor types.

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig4.jpg

Percentage distribution of the different types of MEN1 mutations. Obvious inactivation of menin is predicted by nonsense, frameshift, and splice mutations, and large deletions that constitute 69% of the mutations. Missense and in-frame insertion or deletion (indel) mutations are sliced out of the pie chart to indicate the potential of variants of unknown significance among these 2 types of mutations.

Germline MEN1 mutations are not found in 10% to 30% of patients who develop clinical features consistent with MEN1. These MEN1-mutation–negative patients may carry a germline MEN1 mutation in regions that are not interrogated by current genetic testing methods (such as untranslated, intronic, and regulatory regions), or the tumors may show somatic mosaicism (postzygotic MEN1 mutation), or they carry germline mutations in other genes (such as CDKN1B [114]), or the clinical manifestation of multiple tumors is a sporadic coincidence with no underlying germline mutation. Candidate gene analysis, whole-genome sequencing (WGS) or whole-exome sequencing (WES) approaches have been applied to decipher the germline genetic defects in MEN1-mutation–negative cases.

In silico analysis of MEN1 missense mutations

One of the challenges of MEN1 genetic testing is the interpretation of missense and in-frame indel mutations that do not predict obvious damaging effects to the protein structure or function. Given that menin is a multifunctional protein with many interacting partners, missense variants and in-frame indels could disrupt the function of menin in various ways. However, reliable functional assays are not available to establish the impact of MEN1 missense mutations. The effect of amino acid substitutions on the structure or function of a protein without conducting functional studies can be assessed by computational (in silico) predictive tools: SIFT (Sorting Intolerant From Tolerant), PolyPhen-2 (Polymorphism Phenotyping V-2), MutationTaster, MutationAssessor, and other similar tools. The prediction programs are based on various criteria such as sequence homology, physicochemical similarity between the alternate amino acids, evolutionary conservation, or available 3D structures. However, these tools are only for predictions and their interpretation of pathogenic consequence should be used carefully.

The structure of menin has been used to evaluate the impact of missense mutations. In one study, mapping of 159 unique MEN1 missense mutations on the 3D structure of menin showed that 66% were located in buried residues that may destabilize the protein structure (19). The remaining 34% were located at solvent-exposed sites and might impair protein-protein interactions (19). This study did not compare differences between pathogenic and benign missense mutations. Another study performed an in silico thermodynamic analysis of 345 MEN1 missense mutations using various structures of menin alone or in a complex with peptides of interacting partners (MLL, JUND, or MLL/LEDGF) or with small-molecule inhibitors of menin-MLL interaction, from the Protein Data Bank (95). Thermodynamic destabilization of protein structure was measured as the change in free energy (ΔΔG) resulting from an amino acid substitution that was calculated by the FoldX program. A higher ΔΔG value (> 4 kcal/mol) corelated with a strong destabilizing effect, thus providing an in silico positive predictive value to discriminate between pathogenic and benign missense variants.

In 2015, the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) recommended a variant classification framework in their standards and guidelines (96). This framework suggested a 5-tier variant classification system: pathogenic, likely pathogenic, uncertain significance, likely benign, and benign. This classification was based on the allele frequency, segregation, de novo, protein expression, functional studies, and other factors. For the interpretation of MEN1 missense variants, the TENGEN network (French Oncogenetics Network of Neuroendocrine Tumors) has proposed adjustments to the ACMG-AMP framework (97). These recommendations can be useful for the classification of MEN1 missense variants and the genetic diagnosis of MEN1.

Advances in molecular genetic studies and their applications to genetic diagnosis of MEN1

One of the benefits of MEN1 genetic testing is to confirm the diagnosis of clinical MEN1. However, MEN1 genetic testing is negative in patients with clinical MEN1 who present with incomplete disease manifestations that represent phenocopies, or MEN1-like disease characterized by tumor in as few as 1 of the 3 main MEN1-associated endocrine tissues. The identification of susceptibility genes for endocrine tumor syndromes that have at least one overlapping feature with MEN1 has helped to extend the genetic diagnosis of MEN1 mutation-negative cases to include those additional genes in the genetic testing approach (Fig. 5). Among the 10% to 30% of germline MEN1 mutation–negative cases with or without a family history of clinical MEN1, a few may rarely test positive for germline mutations in genes for MEN1-like conditions (CDKN1B or other CDKI genes, CDC73, CASR, GNA11, AP2S1, GCM2, and AIP) (34, 98, 99). Therefore, the genetic predisposition in clinical MEN1-like cases should be further evaluated by genomic approaches to identify the responsible mutations and genes.

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig5.jpg

Schematic diagram for a suggested approach to germline genetic screening in MEN1 and MEN1-like disease. MEN1: a patient with 2 or more MEN1-associated endocrine tumors. MEN1-like: a patient with as few as any 1 of the 3 main MEN1-associated endocrine tumors. Clinical MEN1: patients with MEN1 or MEN1-like disease features. Genetic MEN1: germline MEN1 mutation-positive. WES, whole-exome sequencing; WGS, whole-genome sequencing; +ve, genetic test is positive; -ve, genetic test is negative, and +ve*, genetic test is positive as per American College of Medical Genetics and Genomics and Association for Molecular Pathology guidelines.

One approach to find the potentially missing MEN1 mutations in clinical MEN1 cases is to screen the noncoding regions of MEN1 (promoter, introns, and untranslated) that are not part of the current genetic testing methods. In one study, targeted next-generation sequencing of the entire 7.2-kilobase genomic region of MEN1 was performed, and no mutation was detected in 16 of 76 patients. Also, none of the 76 cases had a point mutation or short indel mutation in the noncoding regions of MEN1, indicating that such mutations may be very rare (100).

There is only one study that has performed WGS of constitutional and tumor DNA samples from patients who were mutation negative in prior MEN1 genetic testing (101). Among the 6 patients analyzed, surprisingly, pathogenic MEN1 germline heterozygous mutations were identified in 3 (2 splice-site variants c.1186-2A > G and p.Arg223Arg (CGG > CGC), and a missense variant p.Pro12Leu) that was missed in the prior routine genetic testing. One patient showed a pathogenic germline heterozygous missense mutation in CASR (p.Ile555Val), and one patient had a germline heterozygous deletion on chromosome 1q that included CDC73. In the same study, WGS of tumor DNA samples from 6 other mutation-negative patients did not detect any somatic variants in recurrent genes that may act as tumor suppressors (101). Therefore, the results of this WGS analysis raises the possibility of missing a germline mutation in prior routine genetic testing, perhaps due to older sequencing or variant classification methods. Thus it may be useful to repeat the MEN1 genetic testing of individuals who present with clinical MEN1 for whom finding an MEN1 mutation is highly likely (tumors of parathyroid, pituitary, and endocrine pancreas; or tumors of parathyroid and endocrine pancreas).

Collaborative efforts and other novel approaches can be considered to identify mutations in MEN1 or other genes in the 10% to 30% of mutation-negative cases of clinical MEN1. Blood transcriptome sequencing has been used for the identification of rare-disease genes, which is RNA sequencing analysis of RNA isolated from whole blood samples to detect any evidence of altered transcription as a consequence of DNA variants (102, 103). If the transcript is expressed in blood cells (eg, MEN1), the effect on splicing and expression level can be detected from missense, synonymous, and loss-of-function mutations within the coding exons. Loss-of-function mutations can lead to lower transcript levels through nonsense-mediated decay. Also, blood transcriptome analysis can identify the decreased expression of one allele due to a variant in the regulatory region. When combined with WGS, the corresponding DNA variants in genes of interest can be identified that are responsible for the altered transcription. One of the limitations of this approach is the inability to identify causal genes due to lack of expression in blood cells. Also, this approach may not successfully identify disease susceptibility genes if causal variants do not affect splicing or expression of the gene.

Clinical Course of Genetically Positive and Negative MEN1 Patients

MEN1 is mainly characterized by the occurrence of parathyroid tumors, DP-NETs, pituitary adenomas, and adrenal tumors (3, 104-107). A diagnosis of MEN1 can be made based on clinical, familial, or genetic criteria. Thus, for a clinical diagnosis patients should have 2 or more MEN1-associated tumors; for a familial diagnosis patients should have 1 MEN1-associated tumor plus a first-degree relative with MEN1; and for a genetic diagnosis a germline MEN1 mutation needs to be identified (105, 106). Across all these diagnostic criteria, up to 90% of patients will be found to have a MEN1 mutation. To date, however, no genotype-phenotype correlations have been reported, and even within the same family the tumor types and age of tumor onset can differ significantly (105). Therefore, long-term radiological and biochemical screening should be performed and appropriate treatment undertaken, as it has been reported that early diagnosis of tumors with appropriate interventions can significantly improve patient survival (3, 105, 108, 109). It is important to note, however, that MEN1 mutations can also give rise to familial isolated primary hyperparathyroidism (FIHP) (110). FIHP is characterized by the occurrence of hereditary PHPT without occurrence of other MEN1-associated tumors during a follow-up period of more than 10.4 years, and the development of PHPT at age 51.4 ± 14 years (110). Therefore, a diagnosis of FIHP should also be considered in patients who present with PHPT at an advanced age and show no further MEN1 manifestations after a 10-year follow-up.

In 10% to 30% of MEN1 patients who are diagnosed based on clinical criteria, MEN1 mutations are not identified (106, 111). These are referred to as phenocopies, and are clinically challenging because the manifestations and familial penetrance of the disease are not defined, and therefore the risk of tumor occurrence and subsequently the most appropriate screening protocols are debatable. Furthermore, analysis of a large MEN1 family cohort consisting of 152 members indicated that 10% of individuals in the family who were diagnosed as having MEN1 did not have the MEN1 mutation, thereby indicating that phenocopies can also occur in MEN1 patients with a family history of MEN1. Determining whether an individual is an MEN1 is of particular importance because it has been reported that MEN1-mutation–negative patients have less aggressive disease, with the median age of first tumor manifestation 13 years later, and median age of second manifestation 9 years later compared to MEN1-mutation–positive patients (5). Furthermore, no third manifestations were identified in mutation-negative patients compared to 76 patients in the mutation-positive group, and median survival was 14 years greater in the negative vs positive MEN1-mutation patient groups (age 87 vs 73 years) (106). The disease course for each MEN1-mutation–negative patient is however likely to differ, and subclassification of this group based on genetic analysis will help inform the most appropriate screening and treatment (112). To date, a number of genes have been reported as causing MEN1 phenocopies. These genes include the cyclin-dependent kinase inhibitor 1B (CDKN1B), which encodes the cell cycle regulating tumor suppressor protein p27Kip1; rearranged during transfection (RET) proto-oncogene, which encodes a receptor tyrosine kinase (RTK) for members of the glial cell line–derived neurotrophic factor family of extracellular signaling molecules; CDC73, which encodes parafibromin, a subunit of the polymerase II-associated factor protein complex that associates with the RNA polymerase II subunit POLR2A and with a histone methyltransferase complex; CASR, which encodes a G-protein coupled receptor; and aryl hydrocarbon receptor-interacting protein (AIP), which encodes a tumor suppressor protein that interacts and colocalizes with the protein kinase A subunits R1-α (PRKAR1A) and C-α (PRKACA) (3, 99, 101, 113).

CDKN1B mutations have been observed in patients with MEN1-associated tumors. These patients are classified as having MEN4, and to date more than 25 cases have been reported (114-116). The most common tumors to arise in MEN4 patients are parathyroid tumors, followed by pituitary adenomas and pNETs, and then occasional adrenal tumors and nonendocrine tumors such as lipomas and meningiomas (114-116). Furthermore, details of the age of onset of these tumors are limited; however, PHPT has been reported in a 15-year-old individual, indicating that, similar to MEN1, tumors may develop in MEN4 patients during childhood or adolescence (117). Although the clinical manifestations appear similar to MEN1, some key differences have been observed; for example, there are currently no reported cases of prolactinomas in MEN4, and the prevalence of DP-NETs in MEN4 patients is only approximately 25%, compared to up to 70% in MEN1 patients (114). In addition to CDKN1B mutations, mutations in other CDK family members including CDKN2B encoding p15INK4b, CDKN2C encoding p18INK4c, and CDKN1A encoding p21Cip1 have also been identified in MEN1 patients (118). The number of reported cases for these mutations is low, and therefore accurate predictions of tumor manifestation is difficult; however, parathyroid, pituitary, and adrenal tumors have been reported, as well as prostate and breast tumors in these patients (118). Moreover, it is predicted that patients with CDKN1B mutations account for only approximately 3% of MEN1-like individuals, with mutations in CDKN2B, CDKN2C, and CDKN1A estimated to account for up to 1%, 0.5%, and 0.5%, respectively (114, 118). Therefore, mutations in the CDK family may occur in only a small proportion of patients defined as being MEN1 phenocopies.

Analysis of patients and families diagnosed initially to have MEN1 based on clinical criteria has also highlighted additional genes that may represent MEN1 phenocopies, including RET, CDC73, CASR, and AIP (3, 99, 102, 113). RET mutations are usually associated with MEN2 (previously MEN2A) and MEN2B (previously MEN3). MEN2 is characterized by the occurrence of medullary thyroid carcinoma (MTC), pheochromocytoma and parathyroid tumors; while in MEN2 parathyroid tumors are rare, and the occurrence of MTC and pheochromocytoma is found in association with a marfanoid habitus, mucosal neuromas, medullated corneal fibers, and intestinal autonomic ganglion dysfunction leading to megacolon (3). However, a patient presenting with Cushing disease due to a corticotrophinoma at age 48 years, who later developed PHPT and was diagnosed clinically to have MEN1, but in whom a MEN1 mutation was not identified, was subsequently found to have MTC and pheochromocytoma at age 66 years, and an RET mutation (116 RET mutation) (119), consistent with a diagnosis of MEN2. MEN1 phenocopies associated with CDC73 mutations, a gene in which mutations usually cause hyperparathyroidism–jaw tumor (HPT-JT) syndrome characterized by parathyroid tumors, ossifying jaw fibromas, renal tumors, and uterine neoplasms (120), have been reported in 2 unrelated patients who had an initial diagnosis of MEN1, but on genetic analysis were shown to have a CDC73 mutation (99, 100); one had PHPT and a prolactinoma (100), and the other had acromegaly, PHPT, and a pNET (100). MEN1 phenocopies associated with CASR mutations, which usually give rise to familial hypocalciuric hypercalcemia type 1 and FIHP (121), have been reported in 2 unrelated patients; one had acromegaly and hypercalcemia possibly due to PHPT (99), and the other had a NET hepatic metastasis from an unknown primary tumor, and PHPT (102). Thus, these patients represent MEN1 phenocopies because they have developed MEN1 tumor manifestations on a background of hereditary disorders associated with parathyroid tumors. MEN1 phenocopy associated with an AIP mutation, which usually gives rise to familial isolated pituitary adenomas, has been reported in a patient with acromegaly and PHPT, thereby illustrating the occurrence of a MEN1 phenocopy in which parathyroid tumor development arose on a background of a hereditary disorder associated with pituitary adenomas (3, 113). These findings indicate that if a patient is diagnosed with MEN1 based on the combined occurrence of PHPT and a pituitary tumor, then testing for MEN1, RET, CDKN1B, CDKN2B, CDKN2C, CDKN1A, CDC73, CASR, or AIP mutations would be advisable to determine if the patient has MEN1, or could potentially have MEN2, MEN4, HPT-JT, FHH, FIHP, or familial isolated pituitary adenoma. In addition, the prevalence of both PHPT and pituitary adenomas is rapidly rising, with PHPT increasing from 76 to 233 per 100 000 women, and 30 to 85 per 100 000 men over the past 2 decades (122), and pituitary tumors identified in more than 25% of unselected autopsies and 20% of the population undergoing intracranial imaging (123). Therefore, the potential of patients to develop coincidental parathyroid and pituitary tumors, and thus meeting the MEN1 clinical criteria, is also increasing. This again highlights the need for detailed germline genetic testing as well as familial investigation of MEN1 patients (112).

Cohorts of patients who have MEN1-like syndrome but tested negative for MEN1, CDKN1A, CDKN1B, CDKN2B, CDKN2C, CDC73, CASR, RET, and AIP mutations have also been reported (100, 102). It is possible that these patients may have mutations in noncoding regions of the MEN1 gene that affect menin expression, for example, in promotor or enhance regions. Sequencing of cDNA may therefore be of benefit to identify, for example, splicing changes. It is, however, also probable that additional, yet unreported genes are involved in the pathogenesis of MEN1 phenocopies. To identify novel MEN-causing genes will likely require genetic analysis of large cohorts of patients (102). This is likely because novel genes will be occurring in less than 20% of clinically diagnosed MEN1 patients. Thus, in summary the clinical course of patients who are MEN1-mutation–negative differs from that of MEN1-mutation–positive patients. Currently, genetic testing for genes including CDKN1A, CDKN1B, CDKN2B, CDKN2C, CDC73, CASR, RET, and AIP may highlight MEN1 phenocopies; however, these still account for only approximately 5% to 10% of MEN1-mutation–negative cases. Therefore, future studies identifying novel genes will be important for determining the treatment and screening of MEN1-mutation–negative MEN1 patients.

Clinical Presentation of MEN1 in Children and Adolescents

Approximately 12% to 17% of MEN1 patients are diagnosed with the disease in the first 2 decades of life (124-126). Clinically evident disease appears uncommon before adolescence, with consensus guidelines currently recommending phenotype screening of confirmed MEN1 carriers commencing by age 5 years (3). Even if penetrance of MEN1 mutations is age dependent, clinical manifestations of MEN1 have occurred in some patients by age 5 years. Therefore, clinical guidelines suggest the performance of genetic testing in asymptomatic relatives of MEN1-mutated patients as soon as possible, certainly within the first decade of life. MEN1 germline-mutation analysis should be recommended in individuals presenting at an early age with a single, apparently sporadic MEN1-associated tumor (3).

Early manifestation of the classical MEN1 endocrine disorders in young patients can be the first manifestation of the syndrome and thus their recognition may help not only the close monitoring of patient’s treatment, but also direct the screening for other endocrinopathies.

The seminal paper on the role of a germline MEN1 mutation causing pituitary adenomas in children and adolescents described prolactin (PRL)-secreting tumors as the most frequent manifestation, with growth hormone (GH) excess being rare and possibly related to GH-releasing hormone–secreting pancreatic tumor (127). The age at diagnosis of MEN1 syndrome varies according to the clinical, familial, or genetic diagnosis. Clinically a functional tumor is diagnosed much earlier than a nonfunctional tumor (128). The familial diagnosis allows the recognition of gene carriers at birth with the possibility of following the natural history of the disease in a given patient. Future tests based on wide genome analysis will allow the identification of gene mutations even in sporadic asymptomatic carriers. Diagnosis in children has to do with the presymptomatic screening of at-risk patients, which allows for earlier detection and intervention, with a resultant decrease in mortality and morbidity associated with these tumors (129). Presymptomatic screening recommendations for MEN1 management have been based on the youngest age at which disease manifestations have been reported, and this cannot be considered a precise optimal timing.

Diagnosis and therapy

Primary hyperparathyroidism

PHPT is the earliest laboratory and/or clinical manifestation in patients with MEN1. The youngest reported cases have been reported at age 4 years and with a pediatric prevalence of 75% in a study evaluating 122 patients with MEN1 younger than 21 years (125). These data were confirmed in another cohort with a prevalence of 58% (130). PHPT is mainly asymptomatic in young patients with MEN1 (125, 130), even though a higher incidence of rickets and osteomalacia was demonstrated in the pediatric population with PHPT compared with adult patients (131). A single case of a severe complication of MEN1-associated PHPT has been described: A 14-year-old boy with MEN1 experienced a stroke in the absence of other recognized causes, but with only PHPT (132).

Development of hypercalcemia during surveillance is suggestive of PHPT and should be followed up by the simultaneous measurement of calcium and intact parathyroid hormone. Diagnostic management is usually the same as for adult patients.

The therapy for PHPT in MEN1 is surgical parathyroidectomy. In adult patients this should not be postponed because bone complications are more severe in MEN1-associated PHPT than in its sporadic counterpart (133). The published data with 19 MEN1 adolescents developing PHPT before age 20 years and undergoing parathyroidectomy before age 25 years to control calcemia support the use of surgery to avoid PHPT complications, even though 2 patients developed postsurgical hypoparathyroidism (134). Subtotal parathyroidectomy is always recommended.

Treatment with the calcimimetic cinacalcet has been demonstrated to be successful in children affected by neonatal severe hyperparathyroidism (135), but reports on the response to monotherapy with cinacalcet in children with MEN1 are not available.

Pituitary tumors

Pituitary adenomas are the second in frequency in young MEN1 patients (> 30%) with an age at diagnosis as early as 10 years (125, 130). Adult MEN1-associated pituitary adenomas are known to be more frequent in women than men (124). Conversely, in children macroadenomas and severity of symptoms are prevalent in young boys rather than young girls (124, 125, 130, 136-140). Similar to adults, prolactinomas are the most frequent MEN1-associated pituitary tumors in children (3, 125, 130). Pituitary-associated Cushing disease is more frequent than Cushing syndrome caused by adrenal lesions in pediatric MEN1 patients (125, 137, 141). Pituitary tumors have been described in several publications as aggressive because they are larger than in sporadic cases and may be multiple at the time of diagnosis with reduced response to standard therapy (124). A case of a boy with MEN1 who developed a thyrotropin-secreting pituitary carcinoma at age 19 years has been reported in the literature (142). The aggressive behavior of MEN1-associated pituitary tumors is an important reason for anticipating the screening at age 5 years (3).

The treatment of pituitary tumors in young patients should follow the standard therapy for the sporadic forms of the disease. For macroprolactinomas the presence of MEN1 mutation was independently associated with resistance to dopamine therapy in up to 16% of patients (143).

Neuroendocrine tumors

NETs are the rarest lesion in young MEN1 patients, with the youngest patient diagnosed at age 8 years (125, 130, 144). Interestingly, the systematic use of endoscopic ultrasound (EUS) in young MEN1 patients has provided evidence for the prevalence of clinically occult (nonfunctioning) pNETs in up to 42% of pediatric cases (145). Differently from adults, gastrinomas are rare in children with MEN1, but when present can be very aggressive (125, 130). The parents of MEN1 children are educated to recognize symptoms related to Zollinger-Ellison syndrome (ZES). Moreover, MEN1-associated insulinoma, the most frequent NETs in the juvenile MEN1 French cohort, is very precocious in onset and its diagnosis is delayed (125). A possible explanation for this delay is the overlapping of symptoms related to hypoglycemia and epilepsy, a much less rare disorder in infancy. Finally, for the few young MEN1 patients affected by thymic NETs, the disease was lethal (125, 146). Altogether these are important elements for deciding when to start clinical surveillance in young MEN1 patients.

Duodenopancreatic disease is a major cause of mortality in adult patients with MEN1, but although rare is also present in the pediatric and young adult age groups and thus requires active surveillance. Conditions like insulinoma are cured by surgery, thereby avoiding severe hypoglycemia and brain damage (147).

General recommendations

The different studies carried out after the publication of the clinical guidelines in 2012 demonstrate that mortality is rare in children and young adults, whereas morbidity is not uncommon for the manifestation of PHPT, DP-NETs, and pituitary disease. MEN1-associated endocrine tumors in children and adolescents appear to have clinical manifestations that differ from those observed in adults with increased severity. Identification of the endocrine tumors at an earlier stage in children could potentially reduce morbidity in MEN1 as observed in other hereditary tumor syndromes. It is important to remember that any decision related to active early tumor surveillance and intervention should take into account patient preference and ascertain any financial and psychological burden. An important consideration in supporting and managing these younger patients is a risk assessment of the likelihood of aggressive or incidental functional disease vs the burden of frequent medical surveillance.

Adverse Fertility, Pregnancy Outcomes, and Impact of Parental MEN1

Women affected by MEN1 typically show classical endocrinopathies during their reproductive years. Some of these endocrine manifestations are known to potentially affect reproductive health. In a report describing a family genotyped by linkage analysis with 2 of the siblings found to be homozygotes, homozygosity did not result in a more severe phenotype, while resulting in unexplained infertility possibly at the time of conception (148). Surprisingly, until very recently there was little published research regarding the impact of MEN1 on pregnancy and a corresponding lack of data to guide antenatal management, except for a few case reports. There are, however, case reports that outline the management of PHPT and sporadic pituitary tumors and DP-NETs in pregnancy in MEN1. Data concerning the impact of MEN1 on fertility and pregnancy is based on the accumulated experience with more common sporadic, single-organ dysfunction, such as isolated PHPT, pituitary tumors, and NETs.

Adverse fertility

Prolactinoma, which represents 70% of pituitary adenomas occurring before age 21 years (125, 126), is recognized to reduce fertility, whereas PHPT does not affect fertility per se (149, 150). In a historical population-based analysis in a multigenerational kindred in Tasmania, named Tasman 1 MEN1 kindred, fertility and pregnancy outcomes were analyzed, with no adverse impact of MEN1 on patient fertility and stillbirth (151). The most likely explanation for this finding is the lack of symptomatology in parathyroid and pituitary tumors commonly occurring before age 20 years in MEN1 (125). Indeed, in the Tasman population–based analysis the majority of pregnancies should have occurred in the context of subclinical pituitary or parathyroid disease. Similarly, in a Finnish MEN1 kindred, reproductive fitness was not affected by the disease (152). The analysis of the large Tasman kindred also made it possible to exclude any adverse impact of the maternal MEN1 status on offspring sex or offspring MEN1 (151). In a recent retrospective Australian analysis that evaluated 96 pregnancies relating to 26 women within the Tasman 1 cohort, emergency cesarean deliveries and miscarriage rate were not significantly different in MEN1-positive women (153).

Overall the limited published data suggest no adverse impact of MEN1 on patient fertility; however pituitary disease in MEN1 could impair reproductive potential. Therefore, MEN1 patients should be informed about the need of a careful antenatal investigation of any potential problem, with target intervention only in selected patients.

Pregnancy outcomes

Surprisingly, there is little published research on the impact of MEN1 on pregnancy outcomes, making it difficult to guide doctors and patients on the management of the pregnant MEN1 patient. Case reports published offer a limited experience to conclude that endocrine disorders in MEN1 pregnant women are variable with a variable expression of maternofetal complications (154, 155).

Outcomes of pregnancy in MEN1 women could be weighted indirectly on the basis of limited publications reporting on the impact of sporadic, single-organ endocrinopathies. Complications of PHPT in pregnancy are associated with a high incidence of maternal, fetal, and neonatal complications directly proportionate to the degree of maternal calcium levels (156). Maternal complications include kidney stones, pancreatitis, acute hypercalcemia, hypertensive crisis, cardiac arrhythmias, preeclampsia, and miscarriage (153, 156). PHPT in pregnancy can cause intrauterine growth restriction, preterm delivery, intrauterine fetal death, neonatal low birth weight, and hypocalcemia (157). Adverse outcomes appear to relate to the degree of hypercalcemia rather than to the presence of PHPT (149, 150).

Calcemia needs to be monitored during pregnancy in MEN1 women and kept in the mild to moderate levels. Parathyroidectomy is usually avoided when possible and when necessary is timed in the second trimester. Neonates should be monitored for hypocalcemia.

Nonparathyroid MEN1–related endocrinopathies occur less frequently during pregnancy (158). Pituitary disease was the second most common endocrinopathy in MEN1 pregnant women, with a prevalence of prolactinomas and nonfunctioning adenomas, and maternal and neonatal consequences (ie, hypertension and low birth weight) (153, 158).

Gestational diabetes mellitus was found at a higher rate in MEN1 pregnant women (156) as a consequence of a high prevalence of hypoglycemia in infants with an MEN1-positive mother. Therefore, blood glucose should be actively tested in MEN1 pregnant women and in their offspring.

Impact of parental MEN1

In a large survey conducted in the Tasman 1 cohort, parental MEN1 was shown to be associated with a high vulnerability to neonates post partum, and this is not fully explained by the most common metabolic alteration, hypercalcemia (158). Main neonatal complications encompassed lower birth weight, longer stay and admission to a higher dependency nursery, hypoglycemia, and more rarely hypocalcemia (158). Children with an MEN1 parent have an elevated risk of postnatal mortality, and this was attributable not solely to the offspring of MEN1 mothers, but was also present in the offspring of MEN1 fathers (158). Infections appear to be an important cause of mortality in these children, and the potential for a role of menin in the control of the immune system has been proposed (158).

Genetic and Clinical Signatures of Functioning and Nonfunctioning Duodenopancreatic Neuroendocrine Tumors in MEN1 and Their Impacts on Diagnosis and Therapy of These Neoplasia

In MEN1 patients, DP-NETs are highly prevalent and are today the major cause of premature MEN1-related death because of metastasized disease (124, 159, 160). As early as in the first decade of life, a few MEN1 patients have been reported to develop DP-NETs (125). At age 50 years, approximately 50% of patients suffer or have suffered from a DP-NET, rising to a prevalence of almost 90% at older than 80 years (108). Within MEN1 families, there are some indications that tumors arise at an earlier age in patients within successive generations (161). During the lives of patients, multiple functioning as well as nonfunctioning tumors occur throughout the pancreas, which further complicates the management for individual patients (162). Therefore, MEN1 carriers are intensively screened presymptomatically from a young age to enable timely interventions for preventing morbidity and metastasized disease (3). Insights into the clinical and genetic signatures of the different types of DP-NETs and the impact on the management of patients is very important for an adequate management of patients. Because of the complexity, and for well informed and up-to-date decision making tailored to individual patient needs, MEN1 care within an experienced multidisciplinary team dedicated to collaborative research is of the utmost importance (163).

Functioning (hormone-producing) DP-NETs are often diagnosed because of elevation of plasma biochemical markers and the endocrine syndrome that is caused by the hormone produced by the NET. The management of functioning DP-NETs entails both the treatment of the functional syndrome caused by hormonal hypersecretion as well as mitigating the oncologic risk of distant metastases, while minimizing treatment-related morbidity and mortality. This is particularly challenging in gastrinoma, the most frequently occurring functioning DP-NET in MEN1. Gastrinomas occur in approximately 30% of patients with MEN1 (110, 164-168). These tumors produce gastrin, which, if unopposed, leads to acid hypersecretion of the stomach with subsequent severe peptic ulceration and gastrointestinal bleeding, the so-called Zollinger-Ellison syndrome (169). Gastrinomas in MEN1 are often small and in most cases located in the submucosa of the duodenum (170, 171). Proton pump inhibitors are effective for the treatment of peptic ulcer disease and therefore suggested for the treatment of the majority of patients with gastrinomas (3, 172). The prognosis of MEN1-related gastrinomas is hard to interpret because there is a wide variation among studies with regard to diagnosis and treatment (173-178). In a French cohort study, gastrinomas were associated with an increased risk of distant metastases irrespective of tumor size. In this study metastasized gastrinomas were not significantly associated with patient survival (160). However, in a subgroup of MEN1 patients, gastrinomas appeared to have an aggressive course of disease with distant metastases and early death. In this study aggressive tumor behavior was associated with larger tumor size and higher gastrin levels (173). In a recent population-based study the life expectancy of MEN1 patients with a gastrinoma was shown to be reduced. Also, in this study fasting serum gastrin levels were associated with overall survival and could therefore provide a justification for selecting those patients who might benefit from surgery (179). However, since surgery for gastrinomas is often extensive, not always curative, and is associated with morbidity, controversies exist about indication and timing (3, 172, 180).

Interestingly, past Helicobacter pylori exposure was associated with increased prevalence and severity of hypergastrinemia in MEN1 patients (181). Based on these findings, routine H pylori serotyping is suggested in all MEN1 patients, with eradication therapy for those who demonstrate evidence of active infection.

The most frequently occurring functioning pNETs in patients with MEN1 are insulinomas, with an incidence of 10% to 15% (33, 108, 182). Insulinomas lead to symptomatic and potentially life-threatening hypoglycemia. Patients are often young and localization of the insulinoma in the presence of multiple other pNETs is difficult. Therefore, deciding on the type and extent of surgery is complex (183). Although studied only in 6 MEN1 patients with evidence of an insulinoma, the recently developed 68Ga-exendin-4 positron emission tomography–computed tomography (PET-CT) scan appears to help guide selective pancreas surgery (184). This functional imaging technique makes use of the glucagon-like peptide-1 (GLP-1) receptor combined with exendin-4, a synthetic GLP-1 analogue. Although different surgical strategies are followed, resection of a MEN1-related insulinoma is often effective but depends on the individual patient’s characteristics (185). The same dilemma applies to the even more seldom occurring functioning pNETs such as glucagonoma, VIPoma, and GHRH-oma, which also lead to symptoms because of overproduction of hormones and can have a poor prognosis (186).

Nonfunctioning pNETs (NF-pNETs) are the most commonly occurring pNETs in patients with MEN1 (133, 187, 188). Therefore, the clinical practice guidelines advise screening for new tumors and monitoring of already existing tumors by plasma biochemical tumor markers combined with radiological examinations or EUS (3). Based on the recent literature, the annual use of tumor markers can no longer be recommended for diagnosing NF-pNET (189-191). The preferred radiological examination is the magnetic resonance imaging scan, not only because of the lower risk of cumulative ionizing radiation exposure but also because of the better sensitivity compared with CT scans (189). EUS is the most sensitive imaging modality; however, it is operator dependent and invasive. Functional imaging using 68Ga-DOTA PET-CT seems to be most useful for the detection of metastasis of prevalent NF-pNET (191). Up to now, there has been no agreement on the optimal follow-up schedule and the timing of surgical intervention, which is the only curative option. Given the paucity of high-quality evidence, appraisal of the existing literature still leads to different opinions on the timing and extent of interventions for surgical interventions (192). Among the multidisciplinary teams taking care for patients with MEN1, the management of NF-pNETs is one of the greatest dilemmas. On the one hand, surgery for NF-pNET often leads to major short- and long-term complications that should be taken into account in the decision making before proceeding to surgery (193). On the other hand, up to now, surgery has been the only curative option for NF-pNET. In one small nonrandomized and nonblinded study, the somatostatin analogue (SSA) lanreotide appeared to improve the progression-free survival (PFS) of patients with nonmetastatic NF-pNET of less than 2 cm (194). However, before this can be translated to clinical practice, more robust evidence is needed.

In MEN1-related NF-pNETs, well-informed decision making toward an optimal follow-up and treatment plan requires insight into the natural course and prognostic factors (163). NF-pNET was scarce and often at the risk of bias (195). This emphasizes the caution that is needed before applying these scientific findings to clinical practice.

In the follow-up of patients, the size of pNETs is often used for deciding on the follow-up schedule and when to proceed to surgery. From the available studies it can be concluded that a larger tumor size is associated with a higher chance for metastases and a worse survival (195). In addition, from the studies comparing the effect of surgical resection with a watchful-waiting strategy, it can be concluded that patients with tumors smaller than 2 cm have an overall low absolute risk for metastases and death (196-198). Therefore, a watchful-waiting strategy for those patients seems to be oncologically safe, with surgery not lowering the risk for metastases enough when outweighing the risk for complications.

In daily clinical practice tumor growth over time is often used as a predictor for the course of the disease in individual patients. In the aforementioned review, 2 studies assessing the growth of NF-pNETs were identified (195, 199, 200). Overall, from the current available literature it can be concluded that NF-pNETs smaller than 2 cm have a generally stable course, and a watch-and-wait strategy appears to be safe in those patients (195). However, metastasis of NF-pNETs smaller than 2 cm do seldom occur and reliable predictors for the course of disease in individual patients are urgently needed to enable decision making for individual patients (163).

Several studies were undertaken in MEN1 patients assessing the prognostic value of tumor tissue–based markers as assessed by the pathological examination of surgically resected NF-pNETs (80, 85, 201, 202). From research in non-MEN1–related NETs, it is clear that tumors with a higher World Health Organization grade according to the mitotic index and higher Ki67 labeling index are associated with worse patient survival (203). In only one study, tumor grade was assessed as a predictor for survival of MEN1 patients who were operated because of a NF-pNET (201). A higher tumor grade as assessed by mitotic index, in contrast to Ki 67 labeling index, was associated with a higher risk of liver metastasis. These patients generally underwent surgery for tumors larger than 2 cm.

Epigenetic changes contribute to tumor development. This mechanism is potentially reversible and might therefore be a new therapeutic target. Promotor hypermethylation is reported in sporadically occurring pNETs (204). In MEN1-related pNET, the association between promotor hypermethylation expressed as cumulative methylation index and clinical outcome was also studied. Patients with distant metastasis of an NF-pNET appeared to have a higher cumulative methylation index (80).

Cejas et al showed that within the group of NF-pNETs, a distinction can be made between α-cell tumors expressing the transcription factor ARX and β-cell tumors expressing the transcription factor PDX1 (85). In this study, both transcription factors were assessed by immunohistochemistry. The occurrence of distant metastasis after surgical resection of MEN1-related NF-pNETs that were often larger than 2 cm was strongly associated with the expression of ARX and not PDX1. Interestingly, distant metastasis almost exclusively occurred in cases with ALT within this subgroup of tumors expressing ARX positivity.

Of the currently available markers, the WHO grade of tumors can be used to assess the risk for metastasis. Because grade 2 tumors are at a higher risk for metastasis, after resection of these tumors patients should be carefully followed. In addition, tissue-based prognostic factors such as the transcription factors ARX and PDX1 can be assessed by immunohistochemistry. Furthermore, ALT can be an important addition as a biomarker for predicting the risk for metastasis in individual patients with tumors expressing ARX. New biomarkers such as multianalyte circulating transcriptomas, tissue-based molecular factors, and image-based markers are needed for predicting the course of tumors in individual MEN1 patients (195). In the coming decade the goals for MEN1-related DP-NETs should be to attain the ability to predict aggressive behavior early on in the disease course and develop new therapies to prevent metastatic disease. Where patients with sporadic pNET often present with metastatic disease and research is aimed at devising novel treatment options for advanced disease and prediction of treatment response, in MEN1 there is a window of opportunity to prevent metastatic disease. Since MEN1 is a rare condition and high-quality research is urgently needed, patients should be treated in centers of expertise, in which multidisciplinary dedicated care is combined with collaborative research to search for new insights into more individualized care for patients (163).

Novelties in the Surgical Approaches in MEN1 Endocrine Tumors

Surgical management of MEN1 is complex and controversial, given the multifocal and multiglandular nature of the disease and the high risk of tumor recurrence even after surgical intervention. Establishing the diagnosis of MEN1 before making surgical decisions and referring affected individuals to a surgeon with experience in treating MEN1 can be critical in preventing unnecessary operations or inappropriate surgical approaches.

Treatment for parathyroid tumors

The timing and the extent of surgery for MEN1 PHPT remains controversial. Once surgical intervention is necessary, subtotal parathyroidectomy (removal of 3-3.5 glands) is often suggested as the initial treatment (205). If 3.5 or more glands are removed, the rate of persistent disease is 5% to 6%. Preoperative imaging is not sufficiently reliable to justify unilateral exploration, with 86% of patients having enlarged contralateral parathyroid tumors that were missed. Fifty percent of the remaining patients had the largest parathyroid gland identified intraoperatively on the contralateral side (206). Reoperation is often necessary because hyperplastic parathyroid tissue can be stimulated to grow from embryologic locations along its path along the neck and mediastinum (205). Total parathyroidectomy with autotransplantation of parathyroid tissue to the forearm risks the devastating sequelae of rendering the patient hypoparathyroid (206). Concomitant transcervical thymectomy decreases the rate of recurrence and theoretically lessens the risk of thymic neuroendocrine tumor development (which can occur in males) and is suggested at the initial operation (207). The standard cervical exploration remains the procedure of choice. At the initial operation, all 4 parathyroid glands are identified. Occasionally supernumerary parathyroid tissue will be identified. The size of the parathyroid glands can be different and gland weight can vary. Factors for choice of remnant creation include relative macroscopic normality, the accessibility of the preserved gland for subsequent reoperation (inferior parathyroid may be more suitable lying more anteriorly, away from the recurrent laryngeal nerve), and vascular viability. The authors choose the remnant early in the exploration to confirm viability before resection of the other glands. A remnant approximately twice the size of a normal gland (60 mg) is ideal. Robotic and videoscopic techniques have been described but the resource allocation and value have not been proven to be better. However, for recurrent disease or disease noted within the mediastinum that cannot be accessed through the neck, video-assisted thorascopic approaches are useful.

Treatment for duodenopancreatic neuroendocrine tumors

The timing and extent of surgery for DP-NETs are controversial and depend on many factors, including the severity of symptoms, extent of disease, functional component, location and necessity of simple enucleation, subtotal or total pancreatectomy, and pancreaticoduodenectomy (PD; Whipple procedure). Pancreatoduodenectomy has been associated with higher cure rates and improved overall survival; it also has higher rates of postoperative complications and long-term morbidity (208). The risks and benefits should be carefully considered, and surgical decisions should be made on a case-by-case basis. With regard to open or minimally invasive (laparoscopic or robotic) approaches, minimally invasive pancreatectomy appears to be safe and associated with a shorter length of stay and fewer complications in selected patients (209).

Minimally invasive surgery techniques have evolved remarkably over the past few decades in the field of surgical oncology, including minimally invasive pancreatectomy. Because of the relative simplicity of distal pancreatectomy, minimally invasive distal pancreatectomy has been widely accepted and increasingly performed with reported safety (210). Large retrospective analyses as well as recent systematic reviews have shown that minimally invasive distal pancreatectomy resulted in less blood loss and shorter hospital stays compared with open distal pancreatectomy, and there were no significant differences in incidence rates of short-term complications, including postoperative pancreatic fistula, or in mortality rates or complete gross tumor resection rates between the 2 techniques (211, 212). Alfieri and colleagues reported a large Italian multicenter comparative study comparing laparoscopic and robotic distal pancreatectomy for pNETs. The study included a total of 181 patients (96 robotic and 85 laparoscopic distal pancreatectomies), and the authors reported that both approaches are safe and efficacious for pNET treatment, with a similar conversion rate, postoperative morbidity, and pancreatic fistula rate between groups (213). The robotic approach, reported to have a higher spleen-preservation rate and less blood loss compared with laparoscopic PD, is a technically demanding surgical procedure associated with high mortality and morbidity rates. Techniques for minimally invasive PD have continued to evolve (214, 215), most significantly with the emergence of the robotic surgery platform (216).

The LEOPARD-2 trial was conducted in the Netherlands. This multicenter national study was terminated prematurely after accrual of 99 patients owing to safety concerns, with reported 10% 90-day mortality in the laparoscopic group compared with 2% in the open group (217). The results of the LEOPARD-2 trial clearly demonstrate the safety concerns of laparoscopic PD. Augmented surgical dexterity, particularly the wide range of instrument articulation provided by the robotic surgery platform, may improve the safety and generalizability of robotic PD, and several retrospective cohort studies have reported promising results (218). The accumulating reports support the use of robotic minimally invasive PD (Fig. 6); however, prospective randomized, controlled trials are warranted to determine the safety and noninferior oncologic outcomes of robotic PD compared with open PD for patients with pNETs.

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig6.jpg

Intraoperative view after robotic pancreaticoduodenectomy of a pancreatic neuroendocrine tumor (pNET) in the head of the pancreas before reconstruction. CHA, common hepatic artery; IVC, inferior vena cava; SMA, superior mesenteric artery; SMV, superior mesenteric vein.

Recent advances in interventional gastroenterology have allowed for new treatments as emerging adjuncts to standard care in patients with pNETs. Multiple applications of different techniques have been demonstrated to locally treat pNETs. The studies are small and none have had large cohorts of MEN1 patients, but several deserve mention. Local therapies include ethanol ablation using 1 to 2 or more cycles of ethanol lavage at various concentrations for local pNET treatment (219, 220). In 2015, Park et al (221) published results from a series of 11 patients with 14 lesions. Ten patients had nonfunctional NETs and 2 patients had symptomatic insulinomas (1 with 3 lesions). Endoscopic ethanol ablation was performed with 98% ethanol over multiple sessions, and patients were followed for 1 year. Postprocedure complete resolution was seen in 8 of 13 lesions (61.5%); both insulinoma patients were asymptomatic. Three patients had mild pancreatitis and required stent placement for PD stricture. All 3 patients with acute pancreatitis had more than 2-mL ethanol lavage in a single procedure. EUS-guided brachytherapy, photodynamic therapy (222), laser ablation therapy (223), CyberKnife frameless radiosurgery, regarded as image-guided radiotherapy, have also been described as emerging technologies.

Radiofrequency ablation (RFA) is perhaps the most common endoscopic treatment considered safe. The RFA technique emits thermal energy resulting in coagulative necrosis of the surrounding tissue. Recent studies have shown promising results with RFA in patients with unresectable pancreatic cancer in the open, laparoscopic, or percutaneous setting. EUS-guided RFA (EUS-RFA) allows real-time imaging of pancreatic tumors and may result in safe tissue ablation. In 2015, Armellini et al (224) demonstrated the safety and feasibility of EUS-RFA in a patient with pNET who refused surgery. Post ablation, the patient remained asymptomatic and the CT after 1 month showed complete radiological ablation—suggesting RFA can be a potential alternative to surgery in select cases. Lakhtakia and Seo (225) and Waung et al (226) reported that EUS-RFA used in 4 cases of insulinoma resulted in complete clinical resolution in all patients (complete morphological resolution in 2 patients) following EUS-RFA with 12 and 10 months of follow-up, respectively.

Benefits of affecting the systemic immunomodulatory response have also been studied with RFA of pancreas lesions. The initial results assessing a systemic response from RFA have proved feasibility for locally advanced pancreatic tumors (not specifically pNETs) with regard to adding the benefit of evidence of immunomodulation. One study group observed a general activation of an adaptive response decrease of immunosuppression. The plausibility of this occurring with pNETs needs further study (227).

Treatment of nonfunctioning pancreatic neuroendocrine tumors

Approximately 50% of individuals with MEN1 will develop NF-pNETs. These are often identified incidentally during assessment and exploration for functioning tumors. As with gastrinomas, the metastatic rate is correlated with larger tumor size. Tumors smaller than 1.5 cm are not likely to have lymph node metastases (228), although the presence of metastatic disease has been associated with earlier age at death than in those without DP-NETs (127).

Treatment of pituitary tumors

Medical therapy to suppress hypersecretion is often the first line of therapy for MEN1-associated pituitary tumors. Surgery is often necessary for patients who are resistant to this treatment. The indications for surgical removal of pituitary tumors associated with MEN1 are similar to those for nonsyndromic tumors. They include hormone hypersecretion unresponsive to medical therapy, compression of the optic nerves and/or chiasm endangering vision, and uncertainty of diagnosis requiring biopsy. Adenomas that secrete GH (causing acromegaly) or adrenocorticotropin (ACTH) (causing Cushing disease) are typically addressed surgically because medical strategies for those tumor types tend to palliate rather than cure. Prolactinomas are usually treated with dopamine agonists, with surgery reserved for patients in whom side effects limit their ability to take those medications consistently over the long term, or in whom they are incompletely effective in achieving the dual goals of hormone normalization and control of tumor growth. In one series of 136 patients, medical therapy was successful in approximately one-half of patients with secreting tumors (49 of 116, 42%), and successful suppression was correlated with smaller tumor size (229). For the nonfunctional adenomas that form the majority of pituitary tumors seen in MEN1, surgery is the main method for tumor control when such control is needed. Small nonfunctional adenomas (microadenomas, < 10 mm) are usually watched, and intervention is necessary only if they grow close enough to the optic chiasm to warrant removal. The pituitary tumor should be given priority for treatment only in MEN1 patients with pituitary apoplexy (hemorrhage into an adenoma causing loss of vision and/or hormonal collapse) or who present with significant chiasmal compression and risk of blindness.

Most pituitary surgery today is performed by the endonasal transsphenoidal approach, which can be achieved through visualization of the sphenoid sinus and sella by either microscope or endoscope. The microscope confers the advantage of binocular vision with depth of field; the endoscope provides a wider field of view. Both techniques are minimally invasive in nature, and such surgery typically requires short postoperative stays of 1 to 3 days in hospital. Pituitary tumors are typically soft and friable, and can be removed selectively with specially designed curettes that protect the adjacent normal gland, and allow its preservation. Although such removals are typically piecemeal, in the past decade en bloc resection has become possible thanks to the recognition that tumors larger than 2 mm are surrounded by a pseudocapsule that provides a plane for the surgeon (230). For ACTH-negative and GH-positive tumors in particular, en bloc resection provides higher rates of durable remission. Only 1% to 2% of patients will need craniotomy, which carries more risk of neurological injury and is reserved for those with tumors that extend laterally from the sella, encase suprasellar arteries, invade the skull base, or have a firm consistency that makes transsphenoidal removal too difficult and less safe.

Rates of success in the surgical removal of MEN1-associated adenomas are similar to those seen in large general series of pituitary tumors in the general population. Although pituitary tumors in MEN1 patients have been reported as more invasive or aggressive than in those without MEN1, one recent surgical series showed that 82% of the tumors were confined to the sella (Knosp grades 0, 1, and 2), and that rates of remission of hormone hypersecretion and of tumor control matched those in patients without MEN1 (231). The lack of growth usually seen over time in asymptomatic unoperated-on microadenomas in MEN1 is congruent with those findings (232). Applying general principles of pituitary surgery to MEN1-associated adenomas yields excellent control of the majority of cases, and that modern techniques (en bloc removal, endoscopic assistance) have improved the safety and efficacy of such surgery in this group, with significant benefit to such patients. Radiation therapy is reserved for patients with incomplete surgical resection (3).

Pharmacological Therapies in MEN1-associated Endocrine Tumors

MEN1 patients often have multiple, multifocal tumors that occur at a younger age and have a higher metastatic potential compared to the single sporadic counterpart tumors (104, 233). This means that surgery may not be possible, especially in the case of pNETs. In addition, with the increase in screening and subsequent earlier diagnosis, pharmacological treatments may also be administered to delay surgical intervention, or to control symptoms in functioning tumors (3). Selecting the optimal pharmacological therapy is however challenging because clinical trials are often not undertaken in MEN1 patients, and instead results are extrapolated from those undertaken in patients with a single endocrine tumor, or based on descriptive, small-cohort studies. Nevertheless, there are a number of pharmacological, or medical, treatments available for MEN1-associated tumors, that can be broadly divided into biotherapies and chemotherapies.

Biotherapies are agents that target specific tumor-associated receptors, or signaling pathways. For NETs, these predominately include agents that target somatostatin receptor (SSTR), mechanistic target of rapamycin (mTOR), and receptor tyrosine kinase (RTK) signaling (109). The first-line pharmacological treatment for MEN1-associated NETs is commonly a SSA (234). SSAs bind to SSTRs, a family of G protein–coupled receptors consisting of SSTR1-5, to activate their downstream signaling pathways (235). MEN1-associated NETs can express all 5 SSTRs; however, they most commonly express SSTR2 and SSTR5 (234, 236). Targeting SSTR signaling, which affects antisecretory and antiproliferative pathways, is effective in facilitating symptom control due to hormone hypersecretion and in reducing tumor burden (104, 234, 235). Three SSAs, octreotide, lanreotide, and pasireotide, have been used clinically for treating NETs. Octreotide and lanreotide, which primarily bind to SSTR2, have been demonstrated to have efficacy in pituitary and gastroenteropancreatic tumors NETs (237-242). Specific evidence for the efficacy of octreotide and lanreotide in MEN1 patients has also been demonstrated in a study of 5 patients with gastroenteropancreatic NETs associated with hypergastrinemia, in whom 3 months’ treatment reduced gastrin hypersecretion, and reduced the size of liver metastases (243); a retrospective study of 40 individuals with MEN1-associated DP-NETs, in whom 12 to 15 months’ octreotide treatment resulted in tumor response in 10%, and stable disease in 80% of patients (244); in a longitudinal, open-label study of 8 MEN1 patients with pNETs smaller than 2 cm, in whom octreotide treatment resulted in a decrease in GEP hormones and stable disease in approximately 80% of patients (245); and in a prospective observational study, in which lanreotide treatment in 23 MEN1 patients with pNETs smaller than 2 cm significantly improved PFS (203). Pasireotide, which binds to SSTRs 1–3 and 5, has been reported to be effective in treating pituitary adenomas and pNETs (246-249). Thus, pasireotide has been reported to ameliorate hypoglycemia in insulinoma patients, and to be effective in patients who are non-responsive to octreotide and lanreotide (246, 247, 250-252). However, clinical trials have indicated that the efficacy of pasireotide is not significantly superior to octreotide and lanreotide in controlling disease progression, and that it is associated with a higher frequency of hyperglycemia that requires medical intervention, thereby limiting its use (242, 246, 249). Furthermore, due to the lack of detailed clinical trials in MEN1 patients the evidence for the use of pasireotide for MEN1-associated tumors is currently unclear. SSAs have also been used in peptide receptor radionuclide therapy (PRRT), whereby they are labeled with a β-emitting radionuclide, for example 177lutetium (253). One prospective randomized study indicated an increase in PFS in PRRT vs SSA alone in metastatic midgut NETs, and currently PRRT is recommended as a third-line treatment for metastatic midgut and pNETs (111). Small retrospective case series have also highlighted the benefit of PRRT for metastatic MEN1-associated tumors (254, 255).

In addition to SSTRs, other receptors can be targeted for NET treatment. For example, NETs are often highly vascularized and express RTKs including vascular endothelial growth factor receptor (VEGFR), insulin-like growth factor 1 receptor, and platelet-derived growth factor receptor (104, 109). The RTK inhibitors sunitinib and pazopanib have been reported to increase PFS from approximately 5.5 to 11.4 months in patients with pNETs (107, 256). However, the evidence of the efficacy of these inhibitors in MEN1 patients is inconclusive, as only one study specifically examined sunitinib treatment in MEN1 patients, and no improvement in PFS was observed in the 2 patients included in the study (257). In addition to SSTRs and RTKs, pituitary tumors can also express G protein–coupled dopaminergic type 2 receptors, and dopamine agonists (eg, cabergoline) are well known to lower PRL levels and decrease tumor size in prolactinomas in non-MEN1 and MEN1 patients (258). As well as receptors, molecules targeting downstream signaling pathways have also been shown to have efficacy in NETs, including inhibitors of mTOR signaling (83). Everolimus, an mTOR inhibitor approved for the treatment of advanced NETs, has been demonstrated to increase PFS of metastatic pancreatic and lung NET patients from approximately 4 to approximately 11 months (259, 260). Evidence for everolimus for the treatment of locally advanced or metastatic pNETs in MEN1 patients has also been demonstrated in a retrospective study of 6 patients in which PFS was higher in individuals with MEN1 mutations (33.1 months) compared to those with sporadic non-MEN1 disease (12.3 months) (257, 261).

Finally, surgical removal of the multiple parathyroid tumors that occur in patients with MEN1 is the definitive treatment (3). However, in patients in whom surgery has either failed or is contraindicated, drugs acting as CaSR agonists (calcimimetics, eg, cinacalcet) may be successfully used to treat the hypercalcemia and PHPT (262). Thus, in 8 patients up to 30 mg twice daily cinacalcet treatment has been reported to reduce median serum calcium by 0.35 mmol/L, and decrease serum parathyroid hormone by a median of 5.05 pmol/L, with no change in urinary calcium; none of the patients demonstrated recurrent stone formation (262).

Chemotherapies, including alkylating agents, antimicrotubule agents, topoisomerase inhibitors, antimetabolites, and cytotoxic antibodies, have been used to treat NETs. These are, however, reserved for pancreatic, thymic, and bronchopulmonary NET patients with metastatic disease, high tumor burden, or high proliferative index (3, 109). Commonly used agents include the alkylating agents streptozocin and temozolomide, the antimicrotubule agent docetaxel, the topoisomerase inhibitor doxorubicin, and the antimetabolites capecitabine and gemcitabine, either alone or in combination (3, 109). Thus, for high-grade, poorly differentiated pNETs, it has been reported that streptozocin-based chemotherapy is effective (109); however, the benefits for non-pNETs or for MEN1-associated NETs is unclear because of the lack of detailed clinical trials in these patients.

Over the last few decades the increased knowledge of menin function and the development of robust MEN1 tumor models has led to the preclinical evaluation of a number of novel pharmacological therapies specifically targeting MEN1-associated tumor pathways. This includes gene replacement therapies, epigenetic targeting therapies, Wnt pathway inhibitors, and novel mTOR and RTK inhibitors. Furthermore, these and existing pharmacological agents have also been evaluated for their efficacy in chemoprevention. These preclinical studies are discussed subsequently and are shown in Fig. 7.

An external file that holds a picture, illustration, etc.
Object name is bnaa031_fig7.jpg

Emerging therapies for MEN1. Menin, encoded by the MEN1 gene, has roles in multiple pathways associated with cell proliferation. These can be targeted by emerging compounds, including receptor tyrosin kinase (RTKs) inhibitors, novel mechanistic target of rapamycin (mTOR) inhibitors, β-catenin antagonists, epigenetic modulators, and thrombospondin analogues. In addition, preclinical studies indicate MEN1 gene replacement may have efficacy in MEN1 patients, and somatostatin (SST) analogues may have chemopreventive efficacy.

Menin is a tumor suppressor, and loss of menin expression or function results in NET tumor formation. Therefore, MEN1 gene replacement therapy has the potential as a therapy for all MEN1-associated tumors. Thus, injection of a recombinant nonreplicating adenoviral serotype 5 vector containing Men1 cDNA under the control of a cytomegalovirus promoter into pituitary tumors of a conventional heterozygous Men1 knockout mouse (Men1+/–) resulted in increased menin expression, and decreased proliferation without inducing an immune response (263). Furthermore, systemic delivery of a hybrid adeno-associated virus and phage vector that displays active octreotide on the viral surface that allows targeted delivery of a tumor necrosis factor transgene, significantly reduced the size of pNETs in a conditional Men1 pancreatic knockout mouse model (Men1f/f;RIP-Cre) (264). Despite the promise of gene therapy representing a potentially curative treatment for MEN1-associated NETs, its translation into clinical trials may be complex because challenges around delivery, specificity, and off-target effects still need to be clarified.

One of the key functions of menin is the regulation of gene transcription through epigenetic mechanisms, including the modification of histones via methyltransferase and deacetylase complexes (described earlier). The use of epigenetic-targeting compounds may therefore have utility in treating MEN1-associated tumors. Preclinical studies have shown that an inhibitor, JQ1, of the BET protein family that binds to acetylated histone residues to promote gene transcription may have efficacy in MEN1-associated pNETs as it significantly reduced proliferation and increased apoptosis of pNETs in a conditional Men1 knockout mouse model (76). Furthermore, JQ1 has also been shown to reduce secretion of ACTH from the pituitary tumor cell line, AtT20 (265). In addition, another BET inhibitor, CPI203, was able to significantly reduce proliferation of a BON-1 xenograft model, further indicating the likely utility of BET inhibition for pNETs (266). The utility of histone deacetylases inhibitors (HDACis) has also been evaluated in preclinical NET models. The HDAC5 inhibitor LMK-235 significantly reduced proliferation and increased apoptosis of pNET cell lines (267), and etinostat, an HDAC1/3 inhibitor, could significantly inhibit the expression of master regulator proteins of metastatic gastroenteropancreatic NETs, as well as reduce tumor growth of the midgut H-STS cell line xenograft mouse model (268). In addition, the HDACi SAHA has been reported to significantly decrease proliferation, and increase apoptosis of a growth hormone and PRL-secreting GH3 rat pituitary tumor cell line, and to decrease cell viability and ACTH secretion from AtT20 and human-derived corticotroph tumor cells (269, 270). Similarly, the HDACi trichostatin also significantly decreased proliferation and inhibited ACTH production in AtT20 cells (271). A number of BET and HDACis are already in clinical trials, and thus these may offer a promising novel therapeutic approach for MEN1-associated NETs.

Menin has also been reported to promote β-catenin phosphorylation, resulting in inhibition of Wnt signaling; therefore, when menin function is lost β-catenin accumulates in the nucleus and promotes gene transcription (104). The β-catenin inhibitor PRI-724 has been shown to significantly decrease pNET cell line viability (272), and a preclinical study in conditional Men1 knockout mice indicated that the β-catenin antagonist PKF115-584 decreased the number and size of pNETs, as well as increased overall mouse survival (63). PRI-724 is already in clinical trials for other conditions (272), and therefore this may provide a novel pharmacological agent for assessment in MEN1-NETs.

RTK and mTOR inhibitors are already in clinical use, although they may have limited efficacy and patients often stop responding to these inhibitors. Therefore, a number of preclinical studies have investigated different approaches for targeting RTK and mTOR signaling pathways. This includes targeting angiogenic pathways with an anti-VEGF (the cytokine that binds to the RTK, VEGFR) monoclonal antibody. However, studies in a RipTag2 insulinoma mouse model indicated that although these compounds reduced tumor burden, they increased invasiveness and metastasis (273). This increased invasiveness may be overcome by targeting multiple RTK pathways simultaneously. For example, concurrent inhibition of VEGF and cMET using sunitibib and crizotinib, respectively, reduced invasion and metastasis in the RipTag2 pNET model (274). Furthermore, the VEGF, PDGF, and fibroblast growth factor targeting the small kinase inhibitor nindetanib has been reported to decrease tumor growth and prolong survival of Rip1Tag2 insulinomas without increasing local invasiveness or metastatic spread (275). In addition, inhibition of nitric oxide synthase may also prove a novel angiogenic-targeting pharmacological approach, as the nitric oxide synthase inhibitor L-NAME could increase constriction of tumor-supplying arterioles in pNETs of a conventional Men1 knockout mouse model (276). Also, functionally active angiogenic peptides, including for thrompospondin 1 have been reported to suppress angiogenesis and tumor growth in RipTag2 pNETs, and as menin interacts with SMAD3, which is downstream of thrompospondin 1 receptor signaling, this may also provide a menin-targeted therapy for MEN1 patients (104, 277). Finally, novel mTOR inhibitors are being investigated; for example, sapanisertib may provide a novel pharmacological agent for everolimus-resistant pNETs. Thus, sapanisertib caused tumor shrinkage of MEN1-mutant patient xenografts that were implanted in female athymic nude mice, and these included tumors that were nonresponsive to everolimus (278).

Pharmacological agents have efficacy in reducing proliferation and hormone secretion; however, they may also have utility for chemoprevention, which would be of particular importance in MEN1 patients, to prevent or delay tumor occurrence. SSAs may have a role in chemoprevention as paseriotide treatment of a conventional Men1 knockout mouse model and a Pdx1-Cre Men1 knockout model (Men1f/f;Pdx1-Cre) significantly reduced pNET occurrence (279, 280). Furthermore, lanreotide treatment of a conventional Men1 knockout mouse model also significantly decreased the number of new pNETs, and pNET size compared to vehicle-treated mice (281). In addition, clinical trials indicate that patients treated with octreotide have stable disease (244, 245). Therefore, the administration of SSAs to familial or genetically diagnosed MEN1 patients may provide a novel approach for preventing or controlling tumor development.

Preclinical studies of novel agents targeting menin specific pathways that are perturbed on menin loss in tumors may provide a wealth of pharmacological approaches for the treatment of MEN1-associated tumors. These will, however, need to be carefully evaluated in prospective clinical trials specifically recruiting MEN1 patients.

Prognosis and Quality of Life

MEN1 is linked to a higher mortality when compared to a normal population or to nonaffected members in MEN1 families, with DP-NETs and thymic NETs representing the main cause (70%) of death (3, 282). Even if uncommon, other tumors such as adrenal carcinoma and parathyroid carcinoma are fatal (13, 283). A predisposition to breast cancer, mainly of the luminal type, has been reported in female MEN1 patients at a mean age of 45 years, earlier than in the general population, thereby prompting the recommendation for breast cancer screening in MEN1 patients around age 40 years (284).

In females with MEN1, the mortality is lower than in males with MEN1, probably due to the lower incidence of thymic NETs in females. Moreover, sporadic MEN1 cases show higher mortality than familial cases. All these observations affect the surveillance strategies for MEN1 patients.

The publication of the last guidance paper (3) has certainly contributed to the decrease in percentage of deaths related to MEN1 tumors, with a significant improvement in the management of MEN1 patients.

The oncological nature of MEN1, the multiplicity of tumors, the aggressiveness of some of the neoplasms, the lack of solid data on prognosis and of phenotype genotype correlations, increase the potential for a considerable influence of the disease in the health-related quality of life (HRQoL) of the affected patients. Recent studies support this hypothesis, showing a role of the disease and its management in the lower QoL scores in adults with MEN1 (285-288). Important variables that influence HRQoL in MEN1 patients are the possibility of being referred to a dedicated multidisciplinary center and being supported by a dedicated patient advocacy group.

Unfortunately, specific questionnaires measuring HRQoL for MEN1 or other hereditary cancers have not yet been developed, contributing to an underestimation or overestimation of specific traits of the syndrome

Conclusions and Future Prospects

Considerable progress has been made in the past decade to study the natural history, diagnosis, and management of MEN1, and in basic and preclinical research to understand the pathophysiology of MEN1-associated tumors. This review has outlined the major advances since the publication of the most recent MEN1 guidance paper (3). The combined efforts of basic researchers and clinicians have been instrumental to alleviate the morbidity and mortality of affected individuals. National registries and databases have been developed that can facilitate access to relatively large well-characterized patient populations for diagnostic and therapeutic developments. Patient associations and federations have been developed worldwide that can reinforce future screening or intervention programs. In parallel, centers for excellence are now being recognized and these represent quality contacts for reliable information and clinical care for any type of patients’ needs. Basic research on the characterization of menin and mouse models on Men1 loss has revealed protein interactions and pathways affected in tumors that can be targeted for potential therapeutic options.

However, many important questions are still unanswered, and appropriate and timely recommendations must be offered to the clinicians involved in the care of MEN1 patients.

MEN1 is a complex disorder predisposing to more than 20 benign and malignant endocrine and nonendocrine neoplasms. The multidisciplinary team involved in the clinical care of these patients should represent experts in endocrinology, radiology, gastroenterology, surgery, and genetics working in tight connection. This is an essential requirement for the best care of this difficult disease.

Periodic reassessment of the biomedical literature and raw genetic data by experts is encouraged to help to identify new genes or as yet unrecognized syndromes in individuals with specific syndromic features but who lack known genetic susceptibility mutations. Further work is needed to identify epigenetic or modifying factors that may explain even rare associations with uncommon tumors for MEN1. Biobanks of MEN1 biological materials should be established.

Preclinical and clinical evidences point to the efficacy of potential pharmacological treatments for MEN1 tumors. These promising data are a clear indication for the development of multicenter clinical trials nationally and internationally. These studies can benefit from the development of liquid biopsy assays that could predict specific therapeutic options for patients and efficacy of the therapy.

Additionally, the design of a dedicated HRQoL questionnaire will represent a necessary step to increase the analytical effectiveness of any given treatment in MEN1. Advocacy associations will be instrumental to carry on these studies.

Acknowledgments

Images, text, and cognitive input were graciously contributed by Drs Naru Ikoma, Ian McCutchen, and Nirav Thosani. Dr Ikoma (The MD Anderson Cancer Center, Houston, Texas) performed the robotic pancreatectomy and provided images, and Dr Thosani (University of Texas Health Science Center, Houston, Texas), performed the EUS ablation and provided images. Their work has made this review better. We are grateful to Dr Alison B. Hickman (Laboratory of Molecular Biology/National Institute of Diabetes and Digestive and Kidney Diseases/National Institutes of Health) for preparing Fig. 2.

Financial Support: This work was supported by the FIRMO Foundation (to M.L.B.) and the Intramural Research Program of the National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases (to S.K.A.). R.V.T. is a Wellcome Trust and National Institute for Health Research (NIHR) senior investigator and receives funding from the NIHR Oxford Biomedical Research Centre Programme.

Glossary

Abbreviations

3D3-dimensional
ACMGAmerican College of Medical Genetics and Genomics
ACTHadrenocorticotropin
ALLacute lymphoblastic leukemia
ALTalternative lengthening of telomeres
AMLacute myeloid leukemia
AMPAssociation for Molecular Pathology
BETbromodomain and extraterminal
BMPbone morphogenetic protein
CDKcyclin-dependent kinase
CDKIcyclin-dependent kinase inhibitor
cDNAcomplementary DNA
cfDNAcirculating cell free DNA
ChIP-chipchromatin immunoprecipitation coupled with DNA microarray chips
ChIP-seqchromatin immunoprecipitation coupled with next-generation sequencing
del/dupdeletion/duplication
DNMTDNA methyltransferase
DP-NETduodenopancreatic neuroendocrine tumor
EUSendoscopic ultrasound
FIHPfamilial isolated primary hyperparathyroidism
GHgrowth hormone
HDACihistone deacetylases inhibitor
HhHedgehog
HPT-JThyperparathyroidism–jaw tumor syndrome
HRQoLhealth-related quality of life
KDMlysine demethylase
KMThistone lysine methyltransferase
LEDGFlens epithelium–derived growth factor
LOHloss of heterozygosity
MEFmouse embryonic fibroblast
MEN1multiple endocrine neoplasia type 1
MLLrMLL rearranged
MTCmedullary thyroid carcinoma
mTORmechanistic target of rapamycin
ncRNAnoncoding RNA
NF-pNETnonfunctioning pancreatic neuroendocrine tumor
NLSnuclear localization signal
PDpancreaticoduodenectomy
PDXpatient-derived xenograft
PET-CTpositron emission tomography–computed tomography
PFSprogression-free survival
PHPTprimary hyperparathyroidism
pNETpancreatic neuroendocrine tumor
PRLprolactin
PRRTpeptide receptor radionuclide therapy
RFAradiofrequency ablation
RTKreceptor tyrosine kinase
SSAsomatostatin analogue
SSTRsomatostatin receptor
TGF-βtransforming growth factor β
VEGFRvascular endothelial growth factor receptor
WESwhole-exome sequencing
WGSwhole-genome sequencing
WTwild type

Additional Information

Disclosure Statement: The authors have nothing to disclose.

Data Availability

All data generated or analyzed during this study are included in this published article or in the data repositories listed in “References.”

References

1. Chandrasekharappa SC, Guru SC, Manickam P, et al.  Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science. 1997;276(5311):404-407. [Abstract] [Google Scholar]
2. Lemmens I, Van de Ven WJ, Kas K, et al.  Identification of the multiple endocrine neoplasia type 1 (MEN1) gene. The European Consortium on MEN1. Hum Mol Genet. 1997;6(7):1177-1183. [Abstract] [Google Scholar]
3. Thakker RV, Newey PJ, Walls GV, et al. ; Endocrine Society. Clinical practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012;97(9):2990-3011. [Abstract] [Google Scholar]
4. Giusti F, Cianferotti L, Boaretto F, et al.  Multiple endocrine neoplasia syndrome type 1: institution, management, and data analysis of a nationwide multicenter patient database. Endocrine. 2017;58(2):349-359. [Abstract] [Google Scholar]
5. Carroll  RW. Multiple endocrine neoplasia type 1 (MEN1). Asia Pac J Clin Oncol. 2013;9(4):297-309. [Abstract] [Google Scholar]
6. Marchand L, Decaussin-Petrucci M, Giraud S, Cotton F, Thivolet C, Simon C.. Hibernoma and multiple endocrine neoplasia type 1 syndrome: a non-fortuitous association? A case report and literature review. Ann Endocrinol (Paris). 2017;78(3):194-197. [Abstract] [Google Scholar]
7. Cuevas-Ocampo AK, Bollen AW, Goode B, et al.  Genetic confirmation that ependymoma can arise as part of multiple endocrine neoplasia type 1 (MEN1) syndrome. Acta Neuropathol. 2017;133(4):661-663. [Europe PMC free article] [Abstract] [Google Scholar]
8. McKeeby JL, Li X, Zhuang Z, et al.  Multiple leiomyomas of the esophagus, lung, and uterus in multiple endocrine neoplasia type 1. Am J Pathol. 2001;159(3):1121-1127. [Europe PMC free article] [Abstract] [Google Scholar]
9. Guru SC, Goldsmith PK, Burns AL, et al.  Menin, the product of the MEN1 gene, is a nuclear protein. Proc Natl Acad Sci U S A. 1998;95(4):1630-1634. [Europe PMC free article] [Abstract] [Google Scholar]
10. Giusti F, Cianferotti L, Boaretto F, et al.  Multiple endocrine neoplasia syndrome type 1: institution, management, and data analysis of a nationwide multicenter patient database. Endocrine. 2017;58(2):349-359. [Abstract] [Google Scholar]
11. Dreijerink KM, Goudet P, Burgess JR, Valk GD.; International Breast Cancer in MEN1 Study Group . Breast-cancer predisposition in multiple endocrine neoplasia type 1. N Engl J Med. 2014;371(6):583-584. [Europe PMC free article] [Abstract] [Google Scholar]
12. Tepede AA, Welch J, Lee M, et al.  18F-FDOPA PET/CT accurately identifies MEN1-associated pheochromocytoma. Endocrinol Diab Metab. 2020;2020:19-0156. [Europe PMC free article] [Abstract] [Google Scholar]
13. Perrier ND, Arnold A, Costa-Guda J, et al.  New and future perspectives for parathyroid carcinoma. Endocr Relat Cancer. 2020;27(8):T53-T63. [Abstract] [Google Scholar]
14. La P, Desmond A, Hou Z, Silva AC, Schnepp RW, Hua X.. Tumor suppressor menin: the essential role of nuclear localization signal domains in coordinating gene expression. Oncogene. 2006;25(25):3537-3546. [Abstract] [Google Scholar]
15. Francis J, Lin W, Rozenblatt-Rosen O, Meyerson M.. The menin tumor suppressor protein is phosphorylated in response to DNA damage. PloS One. 2011;6(1):e16119. [Europe PMC free article] [Abstract] [Google Scholar]
16. Feng ZJ, Gurung B, Jin GH, Yang XL, Hua XX.. SUMO modification of menin. Am J Cancer Res. 2013;3(1):96-106. [Europe PMC free article] [Abstract] [Google Scholar]
17. He X, Wang L, Yan J, Yuan C, Witze ES, Hua X.. Menin localization in cell membrane compartment. Cancer Biol Ther. 2016;17(1):114-122. [Europe PMC free article] [Abstract] [Google Scholar]
18. Huang J, Gurung B, Wan B, et al.  The same pocket in menin binds both MLL and JUND but has opposite effects on transcription. Nature. 2012;482(7386):542-546. [Europe PMC free article] [Abstract] [Google Scholar]
19. Murai MJ, Chruszcz M, Reddy G, Grembecka J, Cierpicki T.. Crystal structure of menin reveals binding site for mixed lineage leukemia (MLL) protein. J Biol Chem. 2011;286(36):31742-31748. [Europe PMC free article] [Abstract] [Google Scholar]
20. Crabtree JS, Scacheri PC, Ward JM, et al.  A mouse model of multiple endocrine neoplasia, type 1, develops multiple endocrine tumors. Proc Natl Acad Sci U S A. 2001;98(3):1118-1123. [Europe PMC free article] [Abstract] [Google Scholar]
21. Bertolino P, Tong WM, Galendo D, Wang ZQ, Zhang CX.. Heterozygous Men1 mutant mice develop a range of endocrine tumors mimicking multiple endocrine neoplasia type 1. Mol Endocrinol. 2003;17(9):1880-1892. [Abstract] [Google Scholar]
22. Loffler KA, Biondi CA, Gartside M, et al.  Broad tumor spectrum in a mouse model of multiple endocrine neoplasia type 1. Int J Cancer. 2007;120(2):259-267. [Abstract] [Google Scholar]
23. Harding B, Lemos MC, Reed AA, et al.  Multiple endocrine neoplasia type 1 knockout mice develop parathyroid, pancreatic, pituitary and adrenal tumours with hypercalcaemia, hypophosphataemia and hypercorticosteronaemia. Endocr Relat Cancer. 2009;16(4):1313-1327. [Europe PMC free article] [Abstract] [Google Scholar]
24. Bertolino P, Radovanovic I, Casse H, Aguzzi A, Wang ZQ, Zhang CX.. Genetic ablation of the tumor suppressor menin causes lethality at mid-gestation with defects in multiple organs. Mech Dev. 2003;120(5):549-560. [Abstract] [Google Scholar]
25. Crabtree JS, Scacheri PC, Ward JM, et al.  Of mice and MEN1: insulinomas in a conditional mouse knockout. Mol Cell Biol. 2003;23(17):6075-6085. [Europe PMC free article] [Abstract] [Google Scholar]
26. Scacheri PC, Crabtree JS, Kennedy AL, et al.  Homozygous loss of menin is well tolerated in liver, a tissue not affected in MEN1. Mamm Genome. 2004;15(11):872-877. [Abstract] [Google Scholar]
27. Shen HC, He M, Powell A, et al.  Recapitulation of pancreatic neuroendocrine tumors in human multiple endocrine neoplasia type I syndrome via Pdx1-directed inactivation of Men1. Cancer Res. 2009;69(5):1858-1866. [Europe PMC free article] [Abstract] [Google Scholar]
28. Shen HC, Ylaya K, Pechhold K, et al.  Multiple endocrine neoplasia type 1 deletion in pancreatic alpha-cells leads to development of insulinomas in mice. Endocrinology. 2010;151(8):4024-4030. [Europe PMC free article] [Abstract] [Google Scholar]
29. Lu J, Herrera PL, Carreira C, et al.  α Cell-specific Men1 ablation triggers the transdifferentiation of glucagon-expressing cells and insulinoma development. Gastroenterology. 2010;138(5):1954-1965. [Abstract] [Google Scholar]
30. Libutti SK, Crabtree JS, Lorang D, et al.  Parathyroid gland-specific deletion of the mouse Men1 gene results in parathyroid neoplasia and hypercalcemic hyperparathyroidism. Cancer Res. 2003;63(22):8022-8028. [Abstract] [Google Scholar]
31. Bertolino P, Tong WM, Herrera PL, Casse H, Zhang CX, Wang ZQ.. Pancreatic beta-cell-specific ablation of the multiple endocrine neoplasia type 1 (MEN1) gene causes full penetrance of insulinoma development in mice. Cancer Res. 2003;63(16):4836-4841. [Abstract] [Google Scholar]
32. Biondi CA, Gartside MG, Waring P, et al.  Conditional inactivation of the MEN1 gene leads to pancreatic and pituitary tumorigenesis but does not affect normal development of these tissues. Mol Cell Biol. 2004;24(8):3125-3131. [Europe PMC free article] [Abstract] [Google Scholar]
33. Pieterman CR, Conemans EB, Dreijerink KM, et al.  Thoracic and duodenopancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1: natural history and function of menin in tumorigenesis. Endocr Relat Cancer. 2014;21(3):R121-R142. [Abstract] [Google Scholar]
34. Agarwal  SK. The future: genetics advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer. 2017;24(10):T119-T134. [Europe PMC free article] [Abstract] [Google Scholar]
35. Dreijerink KMA, Timmers HTM, Brown M.. Twenty years of menin: emerging opportunities for restoration of transcriptional regulation in MEN1. Endocr Relat Cancer. 2017;24(10):T135-T145. [Europe PMC free article] [Abstract] [Google Scholar]
36. Feng Z, Ma J, Hua X.. Epigenetic regulation by the menin pathway. Endocr Relat Cancer. 2017;24(10):T147-T159. [Europe PMC free article] [Abstract] [Google Scholar]
37. Agarwal SK, Guru SC, Heppner C, et al.  Menin interacts with the AP1 transcription factor JunD and represses JunD-activated transcription. Cell. 1999;96(1):143-152. [Abstract] [Google Scholar]
38. Gobl AE, Berg M, Lopez-Egido JR, Oberg K, Skogseid B, Westin G.. Menin represses JunD-activated transcription by a histone deacetylase-dependent mechanism. Biochim Biophys Acta. 1999;1447(1):51-56. [Abstract] [Google Scholar]
39. Agarwal SK, Novotny EA, Crabtree JS, et al.  Transcription factor JunD, deprived of menin, switches from growth suppressor to growth promoter. Proc Natl Acad Sci U S A. 2003;100(19):10770-10775. [Europe PMC free article] [Abstract] [Google Scholar]
40. Hughes CM, Rozenblatt-Rosen O, Milne TA, et al.  Menin associates with a trithorax family histone methyltransferase complex and with the hoxc8 locus. Mol Cell. 2004;13(4):587-597. [Abstract] [Google Scholar]
41. Yokoyama A, Somervaille TC, Smith KS, Rozenblatt-Rosen O, Meyerson M, Cleary ML.. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell. 2005;123(2):207-218. [Abstract] [Google Scholar]
42. Wang P, Lin C, Smith ER, et al.  Global analysis of H3K4 methylation defines MLL family member targets and points to a role for MLL1-mediated H3K4 methylation in the regulation of transcriptional initiation by RNA polymerase II. Mol Cell Biol. 2009;29(22):6074-6085. [Europe PMC free article] [Abstract] [Google Scholar]
43. Scacheri PC, Davis S, Odom DT, et al.  Genome-wide analysis of menin binding provides insights into MEN1 tumorigenesis. Plos Genet. 2006;2(4):e51. [Europe PMC free article] [Abstract] [Google Scholar]
44. Agarwal SK, Impey S, McWeeney S, et al.  Distribution of menin-occupied regions in chromatin specifies a broad role of menin in transcriptional regulation. Neoplasia. 2007;9(2):101-107. [Europe PMC free article] [Abstract] [Google Scholar]
45. Zhang HL, Li WY, Zhang CP, et al.  Differentially expressed genes in Men1 knockout and wild-type embryoid bodies for pancreatic islet development. Mol Med Rep. 2011;4(2):301-305. [Abstract] [Google Scholar]
46. Hamze Z, Vercherat C, Bernigaud-Lacheretz A, et al.  Altered MENIN expression disrupts the MAFA differentiation pathway in insulinoma. Endocr Relat Cancer. 2013;20(6):833-848. [Europe PMC free article] [Abstract] [Google Scholar]
47. Lu J, Hamze Z, Bonnavion R, et al.  Reexpression of oncoprotein MafB in proliferative β-cells and Men1 insulinomas in mouse. Oncogene. 2012;31(31):3647-3654. [Abstract] [Google Scholar]
48. Shi K, Parekh VI, Roy S, Desai SS, Agarwal SK.. The embryonic transcription factor Hlxb9 is a menin interacting partner that controls pancreatic β-cell proliferation and the expression of insulin regulators. Endocr Relat Cancer. 2013;20(1):111-122. [Europe PMC free article] [Abstract] [Google Scholar]
49. Agarwal SK, 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(5):e37952. [Europe PMC free article] [Abstract] [Google Scholar]
50. Zhou Y, Zhang X, Klibanski A.. MEG3 noncoding RNA: a tumor suppressor. J Mol Endocrinol. 2012;48(3):R45-R53. [Europe PMC free article] [Abstract] [Google Scholar]
51. Modali SD, Parekh VI, Kebebew E, Agarwal SK.. Epigenetic regulation of the lncRNA MEG3 and its target c-MET in pancreatic neuroendocrine tumors. Mol Endocrinol. 2015;29(2):224-237. [Europe PMC free article] [Abstract] [Google Scholar]
52. Milne TA, Hughes CM, Lloyd R, et al.  Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors. Proc Natl Acad Sci U S A. 2005;102(3):749-754. [Europe PMC free article] [Abstract] [Google Scholar]
53. Schnepp RW, Chen YX, Wang H, et al.  Mutation of tumor suppressor gene Men1 acutely enhances proliferation of pancreatic islet cells. Cancer Res. 2006;66(11):5707-5715. [Europe PMC free article] [Abstract] [Google Scholar]
54. Karnik SK, Hughes CM, Gu X, et al.  Menin regulates pancreatic islet growth by promoting histone methylation and expression of genes encoding p27Kip1 and p18INK4c. Proc Natl Acad Sci U S A. 2005;102(41):14659-14664. [Europe PMC free article] [Abstract] [Google Scholar]
55. Loffler KA, Biondi CA, Gartside MG, et al.  Lack of augmentation of tumor spectrum or severity in dual heterozygous Men1 and Rb1 knockout mice. Oncogene. 2007;26(27):4009-4017. [Abstract] [Google Scholar]
56. Matoso A, Zhou Z, Hayama R, Flesken-Nikitin A, Nikitin AY.. Cell lineage-specific interactions between Men1 and Rb in neuroendocrine neoplasia. Carcinogenesis. 2008;29(3):620-628. [Abstract] [Google Scholar]
57. Loffler KA, Mould AW, Waring PM, Hayward NK, Kay GF.. Menin and p53 have non-synergistic effects on tumorigenesis in mice. BMC Cancer. 2012;12:252. [Europe PMC free article] [Abstract] [Google Scholar]
58. Gillam MP, Nimbalkar D, Sun L, et al.  MEN1 tumorigenesis in the pituitary and pancreatic islet requires Cdk4 but not Cdk2. Oncogene. 2015;34(7):932-938. [Europe PMC free article] [Abstract] [Google Scholar]
59. Bai F, Pei XH, Nishikawa T, Smith MD, Xiong Y.. p18Ink4c, but not p27Kip1, collaborates with Men1 to suppress neuroendocrine organ tumors. Mol Cell Biol. 2007;27(4):1495-1504. [Europe PMC free article] [Abstract] [Google Scholar]
60. Wong C, Tang LH, Davidson C, et al.  Two well-differentiated pancreatic neuroendocrine tumor mouse models. Cell Death Differ. 2020;27(1):269-283. [Europe PMC free article] [Abstract] [Google Scholar]
61. Lin W, Francis JM, Li H, et al.  Kmt2a cooperates with menin to suppress tumorigenesis in mouse pancreatic islets. Cancer Biol Ther. 2016;17(12):1274-1281. [Europe PMC free article] [Abstract] [Google Scholar]
62. Lin W, Cao J, Liu J, et al.  Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proc Natl Acad Sci U S A. 2011;108(33):13379-13386. [Europe PMC free article] [Abstract] [Google Scholar]
63. Jiang X, Cao Y, Li F, et al.  Targeting β-catenin signaling for therapeutic intervention in MEN1-deficient pancreatic neuroendocrine tumours. Nat Commun. 2014;5:5809. [Europe PMC free article] [Abstract] [Google Scholar]
64. Ripoche D, Charbord J, Hennino A, et al.  ActivinB is induced in insulinoma to promote tumor plasticity through a β-cell-induced dedifferentiation. Mol Cell Biol. 2015;36(5):756-764. [Europe PMC free article] [Abstract] [Google Scholar]
65. Chamberlain CE, Scheel DW, McGlynn K, et al.  Menin determines K-RAS proliferative outputs in endocrine cells. J Clin Invest. 2014;124(9):4093-4101. [Europe PMC free article] [Abstract] [Google Scholar]
66. Meyer C, Burmeister T, Gröger D, et al.  The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32(2):273-284. [Europe PMC free article] [Abstract] [Google Scholar]
67. Grembecka J, He S, Shi A, et al.  Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol. 2012;8(3):277-284. [Europe PMC free article] [Abstract] [Google Scholar]
68. Borkin D, Klossowski S, Pollock J, et al.  Complexity of blocking bivalent protein-protein interactions: development of a highly potent inhibitor of the Menin-mixed-lineage leukemia interaction. J Med Chem. 2018;61(11):4832-4850. [Europe PMC free article] [Abstract] [Google Scholar]
69. Klossowski S, Miao H, Kempinska K, et al.  Menin inhibitor MI-3454 induces remission in MLL1-rearranged and NPM1-mutated models of leukemia. J Clin Invest. 2020;130(2):981-997. [Europe PMC free article] [Abstract] [Google Scholar]
70. Krivtsov AV, Evans K, Gadrey JY, et al.  A Menin-MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL-rearranged leukemia. Cancer Cell. 2019;36(6):660-673.e11. [Europe PMC free article] [Abstract] [Google Scholar]
71. Uckelmann HJ, Kim SM, Wong EM, et al.  Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science. 2020;367(6477):586-590. [Europe PMC free article] [Abstract] [Google Scholar]
72. Funato K, Major T, Lewis PW, Allis CD, Tabar V.. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science. 2014;346(6216):1529-1533. [Europe PMC free article] [Abstract] [Google Scholar]
73. Fennell LJ, Kane A, Liu C, et al.  APC mutation marks an aggressive subtype of BRAF mutant colorectal cancers. Cancers (Basel). 2020;12(5):1171-1193. [Europe PMC free article] [Abstract] [Google Scholar]
74. Mirabello L, Zhu B, Koster R, et al.  Frequency of pathogenic germline variants in cancer-susceptibility genes in patients with osteosarcoma. JAMA Oncol. 2020;6(5):724-734. [Europe PMC free article] [Abstract] [Google Scholar]
75. Lin W, Watanabe H, Peng S, et al.  Dynamic epigenetic regulation by menin during pancreatic islet tumor formation. Mol Cancer Res. 2015;13(4):689-698. [Abstract] [Google Scholar]
76. Lines KE, Stevenson M, Filippakopoulos P, et al.  Epigenetic pathway inhibitors represent potential drugs for treating pancreatic and bronchial neuroendocrine tumors. Oncogenesis. 2017;6(5):e332. [Europe PMC free article] [Abstract] [Google Scholar]
77. Cedar H, Bergman Y.. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10(5):295-304. [Abstract] [Google Scholar]
78. Yuan Z, Sánchez Claros C, Suzuki M, et al.  Loss of MEN1 activates DNMT1 implicating DNA hypermethylation as a driver of MEN1 tumorigenesis. Oncotarget. 2016;7(11):12633-12650. [Europe PMC free article] [Abstract] [Google Scholar]
79. Tirosh A, Mukherjee S, Lack J, et al.  Distinct genome-wide methylation patterns in sporadic and hereditary nonfunctioning pancreatic neuroendocrine tumors. Cancer. 2019;125(8):1247-1257. [Europe PMC free article] [Abstract] [Google Scholar]
80. Conemans EB, Lodewijk L, Moelans CB, et al.  DNA methylation profiling in MEN1-related pancreatic neuroendocrine tumors reveals a potential epigenetic target for treatment. Eur J Endocrinol. 2018;179(3):153-160. [Abstract] [Google Scholar]
81. Yuan Z, Gardiner JC, Maggi EC, et al.  Tissue-specific induced DNA methyltransferase 1 (Dnmt1) in endocrine pancreas by RCAS-TVA-based somatic gene transfer system promotes β-cell proliferation. Cancer Gene Ther. 2019;26(3-4):94-102. [Europe PMC free article] [Abstract] [Google Scholar]
82. Jiao Y, Shi C, Edil BH, et al.  DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331(6021):1199-1203. [Europe PMC free article] [Abstract] [Google Scholar]
83. Scarpa A, Chang DK, Nones K, et al. ; Australian Pancreatic Cancer Genome Initiative . Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65-71. [Abstract] [Google Scholar]
84. Chan CS, Laddha SV, Lewis PW, et al.  ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat Commun. 2018;9:4158. [Europe PMC free article] [Abstract] [Google Scholar]
85. Cejas P, Drier Y, Dreijerink KMA, et al.  Enhancer signatures stratify and predict outcomes of non-functional pancreatic neuroendocrine tumors. Nat Med. 2019;25(8):1260-1265. [Europe PMC free article] [Abstract] [Google Scholar]
86. Luzi E, Marini F, Giusti F, Galli G, Cavalli L, Brandi ML.. The negative feedback-loop between the oncomir Mir-24-1 and menin modulates the Men1 tumorigenesis by mimicking the “Knudson’s second hit”. PloS One. 2012;7(6):e39767. [Europe PMC free article] [Abstract] [Google Scholar]
87. Vijayaraghavan J, Maggi EC, Crabtree JS.. miR-24 regulates menin in the endocrine pancreas. Am J Physiol Endocrinol Metab. 2014;307(1):E84-E92. [Abstract] [Google Scholar]
88. Luzi E, Ciuffi S, Marini F, Mavilia C, Galli G, Brandi ML.. Analysis of differentially expressed microRNAs in MEN1 parathyroid adenomas. Am J Transl Res. 2017;9(4):1743-1753. [Europe PMC free article] [Abstract] [Google Scholar]
89. Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV.; on behalf of the CCGA Consortium; STRIVE Investigators; GRAIL Inc. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol. 2020;31(6):P745-P759. [Europe PMC free article] [Abstract] [Google Scholar]
90. Kent T, Gracias D, Shepherd S, Clynes D.. Alternative lengthening of telomeres in pediatric cancer: mechanisms to therapies. Front Oncol. 2019;9:1518. [Europe PMC free article] [Abstract] [Google Scholar]
91. Mohammad HP, Barbash O, Creasy CL.. Targeting epigenetic modifications in cancer therapy: erasing the roadmap to cancer. Nat Med. 2019;25(3):403-418. [Abstract] [Google Scholar]
92. Concolino P, Costella A, Capoluongo E.. Multiple endocrine neoplasia type 1 (MEN1): an update of 208 new germline variants reported in the last nine years. Cancer Genet. 2016;209(1-2):36-41. [Abstract] [Google Scholar]
93. Romanet P, Mohamed A, Giraud S, et al.  UMD-MEN1 database: an overview of the 370 MEN1 variants present in 1676 patients from the French population. J Clin Endocrinol Metab. 2019;104(3):753-764. [Abstract] [Google Scholar]
94. Marini F, Giusti F, Fossi C, et al.  Multiple endocrine neoplasia type 1: analysis of germline MEN1 mutations in the Italian multicenter MEN1 patient database. Endocrine. 2018;62(1):215-233. [Abstract] [Google Scholar]
95. Caswell RC, Owens MM, Gunning AC, Ellard S, Wright CF.. Using structural analysis in silico to assess the impact of missense variants in MEN1. J Endocr Soc. 2019;3(12):2258-2275. [Europe PMC free article] [Abstract] [Google Scholar]
96. Richards S, Aziz N, Bale S, et al. ; ACMG Laboratory Quality Assurance Committee . Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405-424. [Europe PMC free article] [Abstract] [Google Scholar]
97. Romanet P, Odou MF, North MO, et al.  Proposition of adjustments to the ACMG-AMP framework for the interpretation of MEN1 missense variants. Hum Mutat. 2019;40(6):661-674. [Abstract] [Google Scholar]
98. Turner JJ, Christie PT, Pearce SH, Turnpenny PD, Thakker RV.. Diagnostic challenges due to phenocopies: lessons from multiple endocrine neoplasia type1 (MEN1). Hum Mutat. 2010;31(1):E1089-E1101. [Abstract] [Google Scholar]
99. Nachtigall LB, Guarda FJ, Lines KE, et al.  Clinical MEN-1 among a large cohort of patients with acromegaly. J Clin Endocrinol Metab. 2020;105(6):e2271-e2281. [Europe PMC free article] [Abstract] [Google Scholar]
100. Carvalho RA, Urtremari B, Jorge AAL, et al.  Germline mutation landscape of multiple endocrine neoplasia type 1 using full gene next-generation sequencing. Eur J Endocrinol. 2018;179(6):391-407. [Abstract] [Google Scholar]
101. Backman S, Bajic D, Crona J, Hellman P, Skogseid B, Stålberg P.. Whole genome sequencing of apparently mutation-negative MEN1 patients. Eur J Endocrinol. 2020;182(1):35-45. [Abstract] [Google Scholar]
102. Frésard L, Smail C, Ferraro NM, et al. ; Undiagnosed Diseases Network; Care4Rare Canada Consortium . Identification of rare-disease genes using blood transcriptome sequencing and large control cohorts. Nat Med. 2019;25(6):911-919. [Europe PMC free article] [Abstract] [Google Scholar]
103. Lee H, Huang AY, Wang LK, et al. ; Undiagnosed Diseases Network . Diagnostic utility of transcriptome sequencing for rare Mendelian diseases. Genet Med. 2020;22(3):490-499. [Europe PMC free article] [Abstract] [Google Scholar]
104. Frost M, Lines KE, Thakker RV.. Current and emerging therapies for PNETs in patients with or without MEN1. Nat Rev Endocrinol. 2018;14(4):216-227. [Europe PMC free article] [Abstract] [Google Scholar]
105. Kamilaris CDC, Stratakis CA.. Multiple endocrine neoplasia type 1 (MEN1): an update and the significance of early genetic and clinical diagnosis. Front Endocrinol (Lausanne). 2019;10:339. [Europe PMC free article] [Abstract] [Google Scholar]
106. de Laat JM, van der Luijt RB, Pieterman CR, et al.  MEN1 redefined, a clinical comparison of mutation-positive and mutation-negative patients. BMC Med. 2016;14(1):182. [Europe PMC free article] [Abstract] [Google Scholar]
107. Ahn HK, Choi JY, Kim KM, et al.  Phase II study of pazopanib monotherapy in metastatic gastroenteropancreatic neuroendocrine tumours. Br J Cancer. 2013;109(6):1414-1419. [Europe PMC free article] [Abstract] [Google Scholar]
108. Pieterman CR, Schreinemakers JM, Koppeschaar HP, et al.  Multiple endocrine neoplasia type 1 (MEN1): its manifestations and effect of genetic screening on clinical outcome. Clin Endocrinol (Oxf). 2009;70(4):575-581. [Abstract] [Google Scholar]
109. Pavel M, O’Toole D, Costa F, et al. ; Vienna Consensus Conference participants . ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site. Neuroendocrinology. 2016;103(2):172-185. [Abstract] [Google Scholar]
110. Hannan FM, Nesbit MA, Christie PT, et al.  Familial isolated primary hyperparathyroidism caused by mutations of the MEN1 gene. Nat Clin Pract Endocrinol Metab. 2008;4(1):53-58. [Abstract] [Google Scholar]
111. Lemos MC, Thakker RV.. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008;29(1):22-32. [Abstract] [Google Scholar]
112. Pieterman CRC, Hyde SM, Wu SY, et al.  Understanding the clinical course of genotype-negative MEN1 patients can inform management strategies. Surgery. Published online July 20, 2020. 10.1016/j.surg.2020-04.067 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
113. Belar O, De La Hoz C, Pérez-Nanclares G, Castaño L, Gaztambide S.; Spanish MEN1 Group . Novel mutations in MEN1, CDKN1B and AIP genes in patients with multiple endocrine neoplasia type 1 syndrome in Spain. Clin Endocrinol (Oxf). 2012;76(5):719-724. [Abstract] [Google Scholar]
114. Frederiksen A, Rossing M, Hermann P, Ejersted C, Thakker RV, Frost M.. Clinical features of multiple endocrine neoplasia type 4: novel pathogenic variant and review of published cases. J Clin Endocrinol Metab. 2019;104(9):3637-3646. [Europe PMC free article] [Abstract] [Google Scholar]
115. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, et al.  Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci U S A. 2006;103(42):15558-15563. [Europe PMC free article] [Abstract] [Google Scholar]
116. Georgitsi M, Raitila A, Karhu A, et al.  Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J Clin Endocrinol Metab. 2007;92(8):3321-3325. [Abstract] [Google Scholar]
117. Elston MS, Meyer-Rochow GY, Dray M, Swarbrick M, Conaglen JV.. Early onset primary hyperparathyroidism associated with a novel germline mutation in CDKN1B. Case Rep Endocrinol. 2015;2015:510985. [Europe PMC free article] [Abstract] [Google Scholar]
118. Agarwal SK, Mateo CM, Marx SJ.. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. J Clin Endocrinol Metab. 2009;94(5):1826-1834. [Europe PMC free article] [Abstract] [Google Scholar]
119. Naziat A, Karavitaki N, Thakker R, et al.  Confusing genes: a patient with MEN2A and Cushing’s disease. Clin Endocrinol (Oxf). 2013;78(6):966-968. [Abstract] [Google Scholar]
120. Newey PJ, Bowl MR, Cranston T, Thakker RV.. Cell division cycle protein 73 homolog (CDC73) mutations in the hyperparathyroidism-jaw tumor syndrome (HPT-JT) and parathyroid tumors. Hum Mutat. 2010;31(3):295-307. [Abstract] [Google Scholar]
121. Hannan FM, Kallay E, Chang W, Brandi ML, Thakker RV.. The calcium-sensing receptor in physiology and in calcitropic and noncalcitropic diseases. Nat Rev Endocrinol. 2018;15(1):33-51. [Europe PMC free article] [Abstract] [Google Scholar]
122. Yeh MW, Ituarte PH, Zhou HC, et al.  Incidence and prevalence of primary hyperparathyroidism in a racially mixed population. J Clin Endocrinol Metab. 2013;98(3):1122-1129. [Europe PMC free article] [Abstract] [Google Scholar]
123. Daly AF, Tichomirowa MA, Beckers A.. The epidemiology and genetics of pituitary adenomas. Best Pract Res Clin Endocrinol Metab. 2009;23(5):543-554. [Abstract] [Google Scholar]
124. Goudet P, Murat A, Binquet C, et al.  Risk factors and causes of death in MEN1 disease. A GTE (Groupe d’Etude des Tumeurs Endocrines) cohort study among 758 patients. World J Surg. 2010;34(2):249-255. [Abstract] [Google Scholar]
125. Goudet P, Dalac A, Le Bras M, et al.  MEN1 disease occurring before 21 years old: a 160-patient cohort study from the Groupe d’étude des Tumeurs Endocrines. J Clin Endocrinol Metab. 2015;100(4):1568-1577. [Abstract] [Google Scholar]
126. Herath M, Parameswaran V, Thompson M, Williams M, Burgess J.. Paediatric and young adult manifestations and outcomes of multiple endocrine neoplasia type 1. Clin Endocrinol (Oxf). 2019;91(5):633-638. [Abstract] [Google Scholar]
127. Stratakis CA, Tichomirowa MA, Boikos S, et al.  The role of germline AIP, MEN1, PRKAR1A, CDKN1B and CDKN2C mutations in causing pituitary adenomas in a large cohort of children, adolescents, and patients with genetic syndromes. Clin Genet. 2010;78(5):457-463. [Europe PMC free article] [Abstract] [Google Scholar]
128. Machens A, Schaaf L, Karges W, et al.  Age-related penetrance of endocrine tumours in multiple endocrine neoplasia type 1 (MEN1): a multicentre study of 258 gene carriers. Clin Endocrinol (Oxf.). 2007;67(4):613-622. [Abstract] [Google Scholar]
129. van Leeuwaarde RS, van Nesselrooij BP, Hermus AR, et al.  Impact of delay in diagnosis in outcomes in MEN1: results from the Dutch MEN1 Study Group. J Clin Endocrinol Metab. 2016;101(3):1159-1165. [Abstract] [Google Scholar]
130. Vannucci L, Marini F, Giusti F, Ciuffi S, Tonelli F, Brandi ML.. MEN1 in children and adolescents: data from patients of a regional referral center for hereditary endocrine tumors. Endocrine. 2018;59(2):438-448. [Abstract] [Google Scholar]
131. Wang W, Kong J, Nie M, et al.  Primary hyperparathyroidism in Chinese children and adolescents: a single-center experience at Peking Union Medical College Hospital. Clin Endocrinol. 2017;87:865-873. [Abstract] [Google Scholar]
132. Mitre N, Mack K, Babovic-Vuksanovic D, Thompson G, Kumar S.. Ischemic stroke as the presenting symptom of primary hyperparathyroidism due to multiple endocrine neoplasia type 1. J Pediatr. 2008;153(4):582-585. [Abstract] [Google Scholar]
133. Maraghelli D, Giusti F, Marini F, Brandi ML.. Bone tissue and mineral metabolism in hereditary endocrine tumors: clinical manifestations and genetic bases. Orphanet J Rare Dis. 2020;15(1):102. [Europe PMC free article] [Abstract] [Google Scholar]
134. Marini F, Giusti F, Tonelli F, Brandi ML.. When parathyroidectomy should be indicated or postponed in adolescents with MEN1-related primary hyperparathyroidism. Front Endocrinol (Lausanne). 2018;9:597. [Europe PMC free article] [Abstract] [Google Scholar]
135. Gannon AW, Monk HM, Levine MA.. Cinacalcet monotherapy in neonatal severe hyperparathyroidism: a case study and review. J Clin Endocrinol Metab. 2014;99(1):7-11. [Europe PMC free article] [Abstract] [Google Scholar]
136. Gan HW, Bulwer C, Jeelani O, Levine MA, Korbonits M, Spoudeas HA.. Treatment-resistant pediatric giant prolactinoma and multiple endocrine neoplasia type 1. Int J Pediatr Endocrinol. 2015;2015(1):15. [Europe PMC free article] [Abstract] [Google Scholar]
137. Simonds WF, Varghese S, Marx SJ, Nieman LK.. Cushing’s syndrome in multiple endocrine neoplasia type 1. Clin Endocrinol (Oxf). 2012;76(3):379-386. [Europe PMC free article] [Abstract] [Google Scholar]
138. Karageorgiadis AS, Papadakis GZ, Biro J, et al.  Ectopic adrenocorticotropic hormone and corticotropin-releasing hormone co-secreting tumors in children and adolescents causing cushing syndrome: a diagnostic dilemma and how to solve it. J Clin Endocrinol Metab. 2015;100(1):141-148. [Europe PMC free article] [Abstract] [Google Scholar]
139. Tatsi C, Stratakis CA.. Aggressive pituitary tumors in the young and elderly. Rev Endocr Metab Disord. 2020;21(2):213-223. [Abstract] [Google Scholar]
140. Delemer  B. MEN1 and pituitary adenomas. Ann Endocrinol (Paris). 2012;73(2):59-61. [Abstract] [Google Scholar]
141. Rix M, Hertel NT, Nielsen FC, et al.  Cushing’s disease in childhood as the first manifestation of multiple endocrine neoplasia syndrome type 1. Eur J Endocrinol. 2004;151(6):709-715. [Abstract] [Google Scholar]
142. Scheithauer BW, Kovacs K, Nose V, et al.  Multiple endocrine neoplasia type 1-associated thyrotropin-producing pituitary carcinoma: report of a probable de novo example. Hum Pathol. 2009;40(2):270-278. [Abstract] [Google Scholar]
143. Salenave S, Ancelle D, Bahougne T, et al.  Macroprolactinomas in children and adolescents: factors associated with the response to treatment in 77 patients. J Clin Endocrinol Metab. 2015;100(3):1177-1186. [Abstract] [Google Scholar]
144. Fabbri HC, Mello MP, Soardi FC, et al.  Long-term follow-up of an 8-year-old boy with insulinoma as the first manifestation of a familial form of multiple endocrine neoplasia type 1. Arq Bras Endocrinol Metabol. 2010;54(8):754-760. [Abstract] [Google Scholar]
145. Gonçalves TD, Toledo RA, Sekiya T, et al.  Penetrance of functioning and nonfunctioning pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1 in the second decade of life. J Clin Endocrinol Metab. 2014;99(1):E89-E96. [Abstract] [Google Scholar]
146. Goudet P, Murat A, Cardot-Bauters C, et al. ; GTE network (Groupe des Tumeurs Endocrines) . Thymic neuroendocrine tumors in multiple endocrine neoplasia type 1: a comparative study on 21 cases among a series of 761 MEN1 from the GTE (Groupe des Tumeurs Endocrines). World J Surg. 2009;33(6):1197-1207. [Abstract] [Google Scholar]
147. Adzick NS, De Leon DD, States LJ, et al.  Surgical treatment of congenital hyperinsulinism: results from 500 pancreatectomies in neonates and children. J Pediatr Surg. 2019;54(1):27-32. [Europe PMC free article] [Abstract] [Google Scholar]
148. Brandi ML, Weber G, Svensson A, et al.  Homozygotes for the autosomal dominant neoplasia syndrome (MEN1). Am J Hum Genet. 1993;53(6):1167-1172. [Europe PMC free article] [Abstract] [Google Scholar]
149. Abood A, Vestergaard P.. Pregnancy outcomes in women with primary hyperparathyroidism. Eur J Endocrinol. 2014;171(1):69-76. [Abstract] [Google Scholar]
150. Hirsch D, Kopel V, Nadler V, Levy S, Toledano Y, Tsvetov G.. Pregnancy outcomes in women with primary hyperparathyroidism. J Clin Endocrinol Metab. 2015;100(5):2115-2122. [Abstract] [Google Scholar]
151. Thompson M, Burgess J.. No evidence of adverse fertility and pregnancy outcome in patients with unrecognized and untreated multiple 1endocrine neoplasia type 1. Clin Endocrinol (Oxf). 2019;90(2):312-319. [Abstract] [Google Scholar]
152. Ebeling T, Vierimaa O, Kytölä S, Leisti J, Salmela PI.. Effect of multiple endocrine neoplasia type 1 (MEN1) gene mutations on premature mortality in familial MEN1 syndrome with founder mutations. J Clin Endocrinol Metab. 2004;89(7):3392-3396. [Abstract] [Google Scholar]
153. Hogg P, Thompson M, Burgess J.. The clinical expression and impact of multiple endocrine neoplasia type 1 during pregnancy. Clin Endocrinol (Oxf). Published online May 17, 2020. 10.1111/cen.14252 [Abstract] [CrossRef] [Google Scholar]
154. Daglar HK, Kirbas A, Biberoglu E, Laleli B, Danisman N.. Management of a multiple endocrine neoplasia type 1 during pregnancy: a case report and review of the literature. J Exp Ther Oncol. 2016;11(3):217-220. [Abstract] [Google Scholar]
155. Mistry M, Gupta M, Kaler M.. Pregnancy in multiple endocrine neoplasia type 1 equals multiple complications. Obstet Med. 2014;7(3):123-125. [Europe PMC free article] [Abstract] [Google Scholar]
156. McCarthy A, Hogarth S, Khoi S, et al.  Management of primary hyperparathyroidism in pregnancy: a case series. Endocrinol Diab Metab. 2019;2019:19-0039. [Europe PMC free article] [Abstract] [Google Scholar]
157. Diaz-Soto G, Linglart A, Sénat MV, Kamenicky P, Chanson P.. Primary hyperparathyroidism in pregnancy. Endocrine. 2013;44(3):591-597. [Abstract] [Google Scholar]
158. Thompson M, Hogg P, De Paoli A, Burgess J.. Parental multiple endocrine neoplasia type 1 (MEN1) is associated with increased offspring childhood mortality. J Clin Endocrinol Metab. 2019;105(4):e1106-e1114. [Abstract] [Google Scholar]
159. Conemans EB, Nell S, Pieterman CRC, et al.  Prognostic factors for survival of MEN1 patients with duodenopancreatic tumors metastatic to the liver: results from the DMSG. Endocr Pract. 2017;23(6):641-648. [Abstract] [Google Scholar]
160. Vinault S, Mariet AS, Le Bras M, et al.  Metastatic potential and survival of duodenal and pancreatic tumors in multiple endocrine neoplasia type 1: a GTE and AFCE cohort study (Groupe d’Étude des Tumeurs Endocrines and Association Francophone de Chirurgie Endocrinienne). Ann Surg. 2020;272(6):1094-1101. [Abstract] [Google Scholar]
161. van den Broek MFM, van Nesselrooij BPM, Pieterman CRC, et al.  Clues for genetic anticipation in multiple endocrine neoplasia type 1. J Clin Endocrinol Metab. 2020;105(7):dgaa257. [Abstract] [Google Scholar]
162. Anlauf M, Schlenger R, Perren A, et al.  Microadenomatosis of the endocrine pancreas in patients with and without the multiple endocrine neoplasia type 1 syndrome. Am J Surg Pathol. 2006;30(5):560-574. [Abstract] [Google Scholar]
163. Pieterman CRC, Sadowski SM, Maxwell JE, et al.  Hereditary endocrine tumours: current state-of-the-art and research opportunities: MEN1-related pancreatic NETs: identification of unmet clinical needs and future directives. Endocr Relat Cancer. 2020;27(8):T9-T25. [Abstract] [Google Scholar]
164. Goudet P, Bonithon-Kopp C, Murat A, et al.  Gender-related differences in MEN1 lesion occurrence and diagnosis: a cohort study of 734 cases from the Groupe d’etude des Tumeurs Endocrines. Eur J Endocrinol. 2011;165(1):97-105. [Abstract] [Google Scholar]
165. Lourenço DM Jr, Toledo RA, Coutinho FL, et al.  The impact of clinical and genetic screenings on the management of the multiple endocrine neoplasia type 1. Clinics (Sao Paulo). 2007;62(4):465-476. [Abstract] [Google Scholar]
166. Sakurai A, Suzuki S, Kosugi S, et al. ; MEN Consortium of Japan . Multiple endocrine neoplasia type 1 in Japan: establishment and analysis of a multicentre database. Clin Endocrinol (Oxf). 2012;76(4):533-539. [Abstract] [Google Scholar]
167. Vierimaa O, Ebeling TM, Kytölä S, et al.  Multiple endocrine neoplasia type 1 in Northern Finland; clinical features and genotype phenotype correlation. Eur J Endocrinol. 2007;157(3):285-294. [Abstract] [Google Scholar]
168. Waldmann J, Fendrich V, Habbe N, et al.  Screening of patients with multiple endocrine neoplasia type 1 (MEN-1): a critical analysis of its value. World J Surg. 2009;33(6):1208-1218. [Abstract] [Google Scholar]
169. Zollinger RM, Ellison EH.. Primary peptic ulcerations of the jejunum associated with islet cell tumors of the pancreas. Ann Surg. 1955;142(4):709-723; discussion, 724. [Abstract] [Google Scholar]
170. Anlauf M, Garbrecht N, Henopp T, et al.  Sporadic versus hereditary gastrinomas of the duodenum and pancreas: distinct clinico-pathological and epidemiological features. World J Gastroenterol. 2006;12(34):5440-5446. [Europe PMC free article] [Abstract] [Google Scholar]
171. Pipeleers-Marichal M, Somers G, Willems G, et al.  Gastrinomas in the duodenums of patients with multiple endocrine neoplasia type 1 and the Zollinger-Ellison syndrome. N Engl J Med. 1990;322(11):723-727. [Abstract] [Google Scholar]
172. Falconi M, Eriksson B, Kaltsas G, et al. ; Vienna Consensus Conference participants. ENETS Consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology. 2016;103(2):153-171. [Europe PMC free article] [Abstract] [Google Scholar]
173. Cadiot G, Vuagnat A, Doukhan I, et al.  Prognostic factors in patients with Zollinger-Ellison syndrome and multiple endocrine neoplasia type 1. Groupe d’Etude des Néoplasies Endocriniennes Multiples (GENEM and groupe de Recherche et d’Etude du Syndrome de Zollinger-Ellison (GRESZE). Gastroenterology. 1999;116(2):286-293. [Abstract] [Google Scholar]
174. Gibril F, Venzon DJ, Ojeaburu JV, Bashir S, Jensen RT.. Prospective study of the natural history of gastrinoma in patients with MEN1: definition of an aggressive and a nonaggressive form. J Clin Endocrinol Metab. 2001;86(11):5282-5293. [Abstract] [Google Scholar]
175. Lopez CL, Falconi M, Waldmann J, et al.  Partial pancreaticoduodenectomy can provide cure for duodenal gastrinoma associated with multiple endocrine neoplasia type 1. Ann Surg. 2013;257(2):308-314. [Abstract] [Google Scholar]
176. Norton JA, Alexander HR, Fraker DL, Venzon DJ, Gibril F, Jensen RT.. Comparison of surgical results in patients with advanced and limited disease with multiple endocrine neoplasia type 1 and Zollinger-Ellison syndrome. Ann Surg. 2001;234(4):495-505; discussion 505. [Abstract] [Google Scholar]
177. Singh MH, Fraker DL, Metz DC.. Importance of surveillance for multiple endocrine neoplasia-1 and surgery in patients with sporadic Zollinger-Ellison syndrome. Clin Gastroenterol Hepatol. 2012;10(11):1262-1269. [Abstract] [Google Scholar]
178. Tonelli F, Fratini G, Nesi G, et al.  Pancreatectomy in multiple endocrine neoplasia type 1-related gastrinomas and pancreatic endocrine neoplasias. Ann Surg. 2006;244(1):61-70. [Abstract] [Google Scholar]
179. van Beek DJ, Nell S, Pieterman CRC, et al.  Prognostic factors and survival in MEN1 patients with gastrinomas: results from the DutchMEN Study Group (DMSG). J Surg Oncol. 2019;120(6):966-975. [Europe PMC free article] [Abstract] [Google Scholar]
180. Albers MB, Manoharan J, Bartsch DK.. Contemporary surgical management of the Zollinger-Ellison syndrome in multiple endocrine neoplasia type 1. Best Pract Res Clin Endocrinol Metab. 2019;33(5):101318. [Abstract] [Google Scholar]
181. Endall R, Thompson M, Parameswaran V, Burgess J.. The relationship of gastrinoma in MEN1 to Helicobacter pylori infection. J Clin Endocrinol Metab. 2020;105(3):e676-e682. [Abstract] [Google Scholar]
182. Norton JA, Fang TD, Jensen RT.. Surgery for gastrinoma and insulinoma in multiple endocrine neoplasia type 1. J Natl Compr Canc Netw. 2006;4(2):148-153. [Abstract] [Google Scholar]
183. O’Riordain DS, O’Brien T, van Heerden JA, Service FJ, Grant CS.. Surgical management of insulinoma associated with multiple endocrine neoplasia type I. World J Surg. 1994;18(4):488-493; discussion 493-494. [Abstract] [Google Scholar]
184. Antwi K, Nicolas G, Fani M, et al.  68Ga-Exendin-4 PET/CT detects insulinomas in patients with endogenous hyperinsulinemic hypoglycemia in MEN-1. J Clin Endocrinol Metab. 2019;104(12):5843-5852. [Abstract] [Google Scholar]
185. van Beek DJ, Nell S, Verkooijen HM, Borel Rinkes IHM, Valk GD, Vriens MR; on behalf of the DutchMEN Study Group; International MEN1 Insulinoma Study Group. Surgery for multiple endocrine neoplasia type 1–related insulinoma: long-term outcomes in a large international cohort. Br J Surg. 2020;107(11):1489-1499. [Europe PMC free article] [Abstract] [Google Scholar]
186. Lévy-Bohbot N, Merle C, Goudet P, et al. ; Groupe des Tumeurs Endocrines. Prevalence, characteristics and prognosis of MEN 1-associated glucagonomas, VIPomas, and somatostatinomas: study from the GTE (Groupe des Tumeurs Endocrines) registry. Gastroenterol Clin Biol. 2004;28(11):1075-1081. [Abstract] [Google Scholar]
187. Gonçalves TD, Toledo RA, Sekiya T, et al.  Penetrance of functioning and nonfunctioning pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1 in the second decade of life. J Clin Endocrinol Metab. 2014;99(1):E89-E96. [Abstract] [Google Scholar]
188. Triponez F, Dosseh D, Goudet P, et al.  Epidemiology data on 108 MEN 1 patients from the GTE with isolated nonfunctioning tumors of the pancreas. Ann Surg. 2006;243(2):265-272. [Abstract] [Google Scholar]
189. de Laat JM, Pieterman CR, Weijmans M, et al.  Low accuracy of tumor markers for diagnosing pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1 patients. J Clin Endocrinol Metab. 2013;98(10):4143-4151. [Abstract] [Google Scholar]
190. Qiu W, Christakis I, Silva A, et al.  Utility of chromogranin A, pancreatic polypeptide, glucagon and gastrin in the diagnosis and follow-up of pancreatic neuroendocrine tumors in multiple endocrine neoplasia type 1 patients. Clin Endocrinol (Oxf). 2016;85(3):400-407. [Europe PMC free article] [Abstract] [Google Scholar]
191. van Treijen MJC, van Beek DJ, van Leeuwaarde RS, Vriens MR, Valk GD.. Diagnosing nonfunctional pancreatic NETs in MEN1: the evidence base. J Endocr Soc. 2018;2(9):1067-1088. [Europe PMC free article] [Abstract] [Google Scholar]
192. Challis BG, Casey RT, Grossman A, Newell-Price J, Newey P, Thakker RV.. What is the appropriate management of nonfunctioning pancreatic neuroendocrine tumors disclosed on screening in adult patients with multiple endocrine neoplasia type 1? Clin Endocrinol (Oxf). 2019;91(6):708-715. [Abstract] [Google Scholar]
193. Nell S, Borel Rinkes IHM, Verkooijen HM, et al. ; DMSG. Early and late complications after surgery for MEN1-related nonfunctioning pancreatic neuroendocrine tumors. Ann Surg. 2018;267(2):352-356. [Abstract] [Google Scholar]
194. Faggiano A, Modica R, Lo Calzo F, et al.  Lanreotide therapy vs active surveillance in MEN1-related pancreatic neuroendocrine tumors < 2 centimeters. J Clin Endocrinol Metab. 2020;105(1):78-84. [Abstract] [Google Scholar]
195. Sadowski SM, Pieterman CRC, Perrier ND, Triponez F, Valk GD.. Prognostic factors for the outcome of nonfunctioning pancreatic neuroendocrine tumors in MEN1: a systematic review of literature. Endocr Relat Cancer. 2020;27(6):R145-R161. [Europe PMC free article] [Abstract] [Google Scholar]
196. Nell S, Verkooijen HM, Pieterman CRC, et al.  Management of MEN1 related nonfunctioning pancreatic NETs: a shifting paradigm: results from the DutchMEN1 Study Group. Ann Surg. 2018;267(6):1155-1160. [Abstract] [Google Scholar]
197. Partelli S, Tamburrino D, Lopez C, et al.  Active surveillance versus surgery of nonfunctioning pancreatic neuroendocrine neoplasms ≤ 2 cm in MEN1 patients. Neuroendocrinology. 2016;103(6):779-786. [Abstract] [Google Scholar]
198. Triponez F, Goudet P, Dosseh D, et al. ; French Endocrine Tumor Study Group. Is surgery beneficial for MEN1 patients with small (< or = 2 cm), nonfunctioning pancreaticoduodenal endocrine tumor? An analysis of 65 patients from the GTE. World J Surg. 2006;30(5):654-662; discussion 663. [Abstract] [Google Scholar]
199. Dʼsouza SL, Elmunzer BJ, Scheiman JM.. Long-term follow-up of asymptomatic pancreatic neuroendocrine tumors in multiple endocrine neoplasia type I syndrome. J Clin Gastroenterol. 2014;48(5):458-461. [Abstract] [Google Scholar]
200. Pieterman CRC, de Laat JM, Twisk JWR, et al.  Long-term natural course of small nonfunctional pancreatic neuroendocrine tumors in MEN1-results from the Dutch MEN1 Study Group. J Clin Endocrinol Metab. 2017;102(10):3795-3805. [Abstract] [Google Scholar]
201. Conemans EB, Brosens LAA, Raicu-Ionita GM, et al.  Prognostic value of WHO grade in pancreatic neuro-endocrine tumors in multiple endocrine neoplasia type 1: results from the DutchMEN1 Study Group. Pancreatology. 2017;17(5):766-772. [Abstract] [Google Scholar]
202. Conemans EB, Raicu-Ionita GM, Pieterman CRC, et al.  Expression of p27Kip1 and p18Ink4c in human multiple endocrine neoplasia type 1-related pancreatic neuroendocrine tumors. J Endocrinol Invest. 2018;41(6):655-661. [Abstract] [Google Scholar]
203. Lee L, Ito T, Jensen RT.. Prognostic and predictive factors on overall survival and surgical outcomes in pancreatic neuroendocrine tumors: recent advances and controversies. Expert Rev Anticancer Ther. 2019;19(12):1029-1050. [Europe PMC free article] [Abstract] [Google Scholar]
204. Karpathakis A, Dibra H, Thirlwell C.. Neuroendocrine tumours: cracking the epigenetic code. Endocr Relat Cancer. 2013;20(3):R65-R82. [Abstract] [Google Scholar]
205. Romero Arenas MA, Morris LF, Rich TA, et al.  Preoperative multiple endocrine neoplasia type 1 diagnosis improves the surgical outcomes of pediatric patients with primary hyperparathyroidism. J Pediatr Surg. 2014;49(4):546-550. [Abstract] [Google Scholar]
206. Nilubol N, Weinstein LS, Simonds WF, Jensen RT, Marx SJ, Kebebew E.. Limited parathyroidectomy in multiple endocrine neoplasia type 1-associated primary hyperparathyroidism: a setup for failure. Ann Surg Oncol. 2016;23(2):416-423. [Abstract] [Google Scholar]
207. Pieterman CR, van Hulsteijn LT, den Heijer M, et al. ; DutchMEN1 Study Group. Primary hyperparathyroidism in MEN1 patients: a cohort study with longterm follow-up on preferred surgical procedure and the relation with genotype. Ann Surg. 2012;255(6):1171-1178. [Abstract] [Google Scholar]
208. Lopez CL, Waldmann J, Fendrich V, Langer P, Kann PH, Bartsch DK.. Long-term results of surgery for pancreatic neuroendocrine neoplasms in patients with MEN1. Langenbecks Arch Surg. 2011;396(8):1187-1196. [Abstract] [Google Scholar]
209. Drymousis P, Raptis DA, Spalding D, et al.  Laparoscopic versus open pancreas resection for pancreatic neuroendocrine tumours: a systematic review and meta-analysis. HPB (Oxford). 2014;16(5):397-406. [Europe PMC free article] [Abstract] [Google Scholar]
210. Asbun HJ, Moekotte AL, Vissers FL, et al. ; International Study Group on Minimally Invasive Pancreas Surgery (I-MIPS). The Miami International evidence-based guidelines on minimally invasive pancreas resection. Ann Surg. 2020;271(1):1-14. [Abstract] [Google Scholar]
211. van Hilst J, de Rooij T, Klompmaker S, et al. ; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Minimally invasive versus open distal pancreatectomy for ductal adenocarcinoma (DIPLOMA): a Pan-European propensity score matched study. Ann Surg. 2019;269(1):10-17. [Abstract] [Google Scholar]
212. Riviere D, Gurusamy KS, Kooby DA, et al.  Laparoscopic versus open distal pancreatectomy for pancreatic cancer. Cochrane Database Syst Rev. 2016;4(4):CD011391. [Europe PMC free article] [Abstract] [Google Scholar]
213. Alfieri S, Butturini G, Boggi U, et al. ; Italian Robotic pNET Group. Short-term and long-term outcomes after robot-assisted versus laparoscopic distal pancreatectomy for pancreatic neuroendocrine tumors (pNETs): a multicenter comparative study. Langenbecks Arch Surg. 2019;404(4):459-468. [Abstract] [Google Scholar]
214. Croome KP, Farnell MB, Que FG, et al.  Total laparoscopic pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: oncologic advantages over open approaches?  Ann Surg.  2014;260(4):633-638; discussion 638-640. [Abstract] [Google Scholar]
215. Kendrick ML, van Hilst J, Boggi U, et al. ; Minimally Invasive Pancreatic Resection Organizing Committee. Minimally invasive pancreatoduodenectomy. HPB (Oxford). 2017;19(3):215-224. [Abstract] [Google Scholar]
216. Watkins AA, Kent TS, Gooding WE, et al.  Multicenter outcomes of robotic reconstruction during the early learning curve for minimally-invasive pancreaticoduodenectomy. HPB (Oxford). 2018;20(2):155-165. [Abstract] [Google Scholar]
217. van Hilst J, de Rooij T, Bosscha K, et al. ; Dutch Pancreatic Cancer Group. Laparoscopic versus open pancreatoduodenectomy for pancreatic or periampullary tumours (LEOPARD-2): a multicentre, patient-blinded, randomised controlled phase 2/3 trial. Lancet Gastroenterol Hepatol. 2019;4(3):199-207. [Abstract] [Google Scholar]
218. Shi Y, Wang W, Qiu W, et al.  Learning curve from 450 cases of robot-assisted pancreaticoduocectomy in a high-volume pancreatic center: optimization of operative procedure and a retrospective study. Ann Surg. Published online October 22, 2019. 10.1097/SLA.0000000000003664 [Abstract] [CrossRef] [Google Scholar]
219. Paik WH, Seo DW, Dhir V, Wang HP.. Safety and efficacy of EUS-guided ethanol ablation for treating small solid pancreatic neoplasm. Medicine (Baltimore). 2016;95(4):e2538. [Europe PMC free article] [Abstract] [Google Scholar]
220. Yang D, Inabnet WB III, Sarpel U, DiMaio CJ.. EUS-guided ethanol ablation of symptomatic pancreatic insulinomas. Gastrointest Endosc. 2015;82(6):1127. [Abstract] [Google Scholar]
221. Park DH, Choi JH, Oh D, et al.  Endoscopic ultrasonography-guided ethanol ablation for small pancreatic neuroendocrine tumors: results of a pilot study. Clin Endosc. 2015;48(2):158-164. [Europe PMC free article] [Abstract] [Google Scholar]
222. Choi JH, Oh D, Lee JH, et al.  Initial human experience of endoscopic ultrasound-guided photodynamic therapy with a novel photosensitizer and a flexible laser-light catheter. Endoscopy. 2015;47(11):1035-1038. [Abstract] [Google Scholar]
223. Di Matteo FM, Saccomandi P, Martino M, et al.  Feasibility of EUS-guided Nd:YAG laser ablation of unresectable pancreatic adenocarcinoma. Gastrointest Endosc. 2018;88(1):168-174.e1. [Abstract] [Google Scholar]
224. Armellini E, Crinò SF, Ballarè M, Occhipinti P.. Endoscopic ultrasound-guided radiofrequency ablation of a pancreatic neuroendocrine tumor. Endoscopy. 2015;47(Suppl 1 UCTN):E600-E601. [Abstract] [Google Scholar]
225. Lakhtakia S, Seo DW.. Endoscopic ultrasonography-guided tumor ablation. Dig Endosc. 2017;29(4):486-494. [Abstract] [Google Scholar]
226. Waung JA, Todd JF, Keane MG, Pereira SP.. Successful management of a sporadic pancreatic insulinoma by endoscopic ultrasound-guided radiofrequency ablation. Endoscopy. 2016;48(Suppl 1):E144-E145. [Abstract] [Google Scholar]
227. Giardino A, Innamorati G, Ugel S, et al.  Immunomodulation after radiofrequency ablation of locally advanced pancreatic cancer by monitoring the immune response in 10 patients. Pancreatology. 2017;17(6):962-966. [Abstract] [Google Scholar]
228. Zhang IY, Zhao J, Fernandez-Del Castillo C, et al.  Operative versus nonoperative management of nonfunctioning pancreatic neuroendocrine tumors. J Gastrointest Surg. 2016;20(2):277-283. [Abstract] [Google Scholar]
229. Vergès B, Boureille F, Goudet P, et al.  Pituitary disease in MEN type 1 (MEN1): data from the France-Belgium MEN1 multicenter study. J Clin Endocrinol Metab. 2002;87(2):457-465. [Abstract] [Google Scholar]
230. Oldfield EH, Vortmeyer AO.. Development of a histological pseudocapsule and its use as a surgical capsule in the excision of pituitary tumors. J Neurosurg. 2006;104(1):7-19. [Abstract] [Google Scholar]
231. Wu Y, Gao L, Guo X, et al.  Pituitary adenomas in patients with multiple endocrine neoplasia type 1: a single-center experience in China. Pituitary. 2019;22(2):113-123. [Abstract] [Google Scholar]
232. de Laat JM, Dekkers OM, Pieterman CR, et al.  Long-term natural course of pituitary tumors in patients with MEN1: results from the DutchMEN1 Study Group (DMSG). J Clin Endocrinol Metab. 2015;100(9):3288-3296. [Abstract] [Google Scholar]
233. Yates CJ, Newey PJ, Thakker RV.. Challenges and controversies in management of pancreatic neuroendocrine tumours in patients with MEN1. Lancet Diabetes Endocrinol. 2015;3(11):895-905. [Abstract] [Google Scholar]
234. Grozinsky-Glasberg S, Shimon I, Korbonits M, Grossman AB.. Somatostatin analogues in the control of neuroendocrine tumours: efficacy and mechanisms. Endocr Relat Cancer. 2008;15(3):701-720. [Abstract] [Google Scholar]
235. Theodoropoulou M, Stalla GK.. Somatostatin receptors: from signaling to clinical practice. Front Neuroendocrinol. 2013;34(3):228-252. [Abstract] [Google Scholar]
236. Mizutani G, Nakanishi Y, Watanabe N, et al.  Expression of somatostatin receptor (SSTR) subtypes (SSTR-1, 2A, 3, 4 and 5) in neuroendocrine tumors using real-time RT-PCR method and immunohistochemistry. Acta Histochem Cytochem. 2012;45(3):167-176. [Europe PMC free article] [Abstract] [Google Scholar]
237. Rinke A, Wittenberg M, Schade-Brittinger C, et al. ; PROMID Study Group. 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 (PROMID): results of long-term survival. Neuroendocrinology. 2017;104(1):26-32. [Abstract] [Google Scholar]
238. Melmed  S. New therapeutic agents for acromegaly. Nat Rev Endocrinol. 2016;12(2):90-98. [Abstract] [Google Scholar]
239. Caplin ME, Pavel M, Ćwikła JB, et al. ; CLARINET Investigators . Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224-233. [Abstract] [Google Scholar]
240. Caplin ME, Pavel M, Ćwikła JB, et al. ; CLARINET Investigators . Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer. 2016;23(3):191-199. [Europe PMC free article] [Abstract] [Google Scholar]
241. Hofland J, Herrera-Martínez AD, Zandee WT, de Herder WW.. Management of carcinoid syndrome: a systematic review and meta-analysis. Endocr Relat Cancer. 2019;26(3):R145-R156. [Abstract] [Google Scholar]
242. Yao JC, Chan JA, Mita AC, et al.  Phase I dose-escalation study of long-acting pasireotide in patients with neuroendocrine tumors. Onco Targets Ther. 2017;10:3177-3186. [Europe PMC free article] [Abstract] [Google Scholar]
243. Burgess JR, Greenaway TM, Parameswaran V, Shepherd JJ.. Octreotide improves biochemical, radiologic, and symptomatic indices of gastroenteropancreatic neoplasia in patients with multiple endocrine neoplasia type 1 (MEN-1). Implications for an integrated model of MEN-1 tumorigenesis. Cancer. 1999;86(10):2154-2159. [Abstract] [Google Scholar]
244. Ramundo V, Del Prete M, Marotta V, et al. ; Multidisciplinary Group for Neuroendocrine Tumors of Naples . Impact of long-acting octreotide in patients with early-stage MEN1-related duodeno-pancreatic neuroendocrine tumours. Clin Endocrinol (Oxf). 2014;80(6):850-855. [Abstract] [Google Scholar]
245. Cioppi F, Cianferotti L, Masi L, Giusti F, Brandi ML.. The LARO-MEN1 study: a longitudinal clinical experience with octreotide long-acting release in patients with multiple endocrine neoplasia type 1 syndrome. Clin Cases Miner Bone Metab. 2017;14(2):123-130. [Europe PMC free article] [Abstract] [Google Scholar]
246. Kvols LK, Oberg KE, O’Dorisio TM, et al.  Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer. 2012;19(5):657-666. [Abstract] [Google Scholar]
247. Hendren NS, Panach K, Brown TJ, et al.  Pasireotide for the treatment of refractory hypoglycaemia from malignant insulinoma. Clin Endocrinol (Oxf). 2018;88(2):341-343. [Abstract] [Google Scholar]
248. Cives M, Kunz PL, Morse B, et al.  Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer. 2015;22(1):1-9. [Europe PMC free article] [Abstract] [Google Scholar]
249. Wolin EM, Jarzab B, Eriksson B, et al.  Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Devel Ther. 2015;9:5075-5086. [Europe PMC free article] [Abstract] [Google Scholar]
250. Tirosh A, Stemmer SM, Solomonov E, et al.  Pasireotide for malignant insulinoma. Hormones (Athens). 2016;15(2):271-276. [Abstract] [Google Scholar]
251. Colao A, Bronstein MD, Freda P, et al. ; Pasireotide C2305 Study Group . Pasireotide versus octreotide in acromegaly: a head-to-head superiority study. J Clin Endocrinol Metab. 2014;99(3):791-799. [Europe PMC free article] [Abstract] [Google Scholar]
252. Gadelha MR, Bronstein MD, Brue T, et al. ; Pasireotide C2402 Study Group . Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, phase 3 trial. Lancet Diabetes Endocrinol. 2014;2(11):875-884. [Abstract] [Google Scholar]
253. Ezziddin S, Khalaf F, Vanezi M, et al.  Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2014;41(5):925-933. [Abstract] [Google Scholar]
254. Naik C, Basu S.. Peptide receptor radionuclide therapy with 177Lu-DOTATATE for metastatic neuroendocrine tumor occurring in association with multiple endocrine neoplasia type 1 and Cushing’s syndrome. World J Nucl Med. 2017;16(2):126-132. [Europe PMC free article] [Abstract] [Google Scholar]
255. Gezer E, Çetinarslan B, Cantürk Z, Tarkun İ, Sözen M, Selek A.. Metastatic MEN1 syndrome treated with Lutetium-177—a case report. Eur Endocrinol. 2019;15(2):92-94. [Europe PMC free article] [Abstract] [Google Scholar]
256. Raymond E, Dahan L, Raoul JL, et al.  Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364(6):501-513. [Abstract] [Google Scholar]
257. Nuñez JE, Donadio M, Filho DR, et al.  The efficacy of everolimus and sunitinib in patients with sporadic or germline mutated metastatic pancreatic neuroendocrine tumors. J Gastrointest Oncol. 2019;10(4):645-651. [Europe PMC free article] [Abstract] [Google Scholar]
258. Gillam MP, Molitch ME, Lombardi G, Colao A.. Advances in the treatment of prolactinomas. Endocr Rev. 2006;27(5):485-534. [Abstract] [Google Scholar]
259. Yao JC, Lombard-Bohas C, Baudin E, et al.  Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol. 2010;28(1):69-76. [Europe PMC free article] [Abstract] [Google Scholar]
260. Yao JC, Fazio N, Singh S, et al. ; RAD001 in Advanced Neuroendocrine Tumours, Fourth Trial (RADIANT-4) Study Group . Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387(10022):968-977. [Europe PMC free article] [Abstract] [Google Scholar]
261. Maia MC, Muniz Lourenço D Jr, Riechelmann R.. Efficacy and long-term safety of everolimus in pancreatic neuroendocrine tumor associated with multiple endocrine neoplasia type I: case report. Oncol Res Treat. 2016;39(10):643-645. [Abstract] [Google Scholar]
262. Moyes VJ, Monson JP, Chew SL, Akker SA.. Clinical use of cinacalcet in MEN1 hyperparathyroidism. Int J Endocrinol. 2010;2010:906163. [Europe PMC free article] [Abstract] [Google Scholar]
263. Walls GV, Lemos MC, Javid M, et al.  MEN1 gene replacement therapy reduces proliferation rates in a mouse model of pituitary adenomas. Cancer Res. 2012;72(19):5060-5068. [Europe PMC free article] [Abstract] [Google Scholar]
264. Smith TL, Yuan Z, Cardó-Vila M, et al.  AAVP displaying octreotide for ligand-directed therapeutic transgene delivery in neuroendocrine tumors of the pancreas. Proc Natl Acad Sci U S A. 2016;113(9):2466-2471. [Europe PMC free article] [Abstract] [Google Scholar]
265. Lines KE, Filippakopoulos P, Stevenson M, et al.  Effects of epigenetic pathway inhibitors on corticotroph tumour AtT20 cells. Endocr Relat Cancer. 2020;27(3):163-174. [Europe PMC free article] [Abstract] [Google Scholar]
266. Wong C, Laddha SV, Tang L, et al.  The bromodomain and extra-terminal inhibitor CPI203 enhances the antiproliferative effects of rapamycin on human neuroendocrine tumors. Cell Death Dis. 2014;5(10):e1450. [Europe PMC free article] [Abstract] [Google Scholar]
267. Wanek J, Gaisberger M, Beyreis M, et al.  Pharmacological inhibition of class IIA HDACs by LMK-235 in pancreatic neuroendocrine tumor cells. Int J Mol Sci. 2018;19(10):3128. [Europe PMC free article] [Abstract] [Google Scholar]
268. Alvarez MJ, Subramaniam PS, Tang LH, et al.  A precision oncology approach to the pharmacological targeting of mechanistic dependencies in neuroendocrine tumors. Nat Genet. 2018;50(7):979-989. [Europe PMC free article] [Abstract] [Google Scholar]
269. Sangeetha SR, Singh N, Vender JR, Dhandapani KM.. Suberoylanilide hydroxamic acid (SAHA) induces growth arrest and apoptosis in pituitary adenoma cells. Endocrine. 2009;35(3):389-396. [Abstract] [Google Scholar]
270. Lu J, Chatain GP, Bugarini A, et al.  Histone deacetylase inhibitor SAHA is a promising treatment of Cushing disease. J Clin Endocrinol Metab. 2017;102(8):2825-2835. [Europe PMC free article] [Abstract] [Google Scholar]
271. Nakada Y, Kageyama K, Sugiyama A, et al.  Inhibitory effects of trichostatin A on adrenocorticotropic hormone production and proliferation of corticotroph tumor AtT-20 cells. Endocr J. 2015;62(12):1083-1090. [Abstract] [Google Scholar]
272. Jin XF, Spoettl G, Maurer J, Nölting S, Auernhammer CJ.. Inhibition of Wnt/β-catenin signaling in neuroendocrine tumors in vitro: antitumoral effects. Cancers (Basel). 2020;12(2):345. [Europe PMC free article] [Abstract] [Google Scholar]
273. Casanovas O, Hicklin DJ, Bergers G, Hanahan D.. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8(4):299-309. [Abstract] [Google Scholar]
274. Sennino B, Ishiguro-Oonuma T, Wei Y, et al.  Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2012;2(3):270-287. [Europe PMC free article] [Abstract] [Google Scholar]
275. Bill R, Fagiani E, Zumsteg A, et al.  Nintedanib is a highly effective therapeutic for neuroendocrine carcinoma of the pancreas (PNET) in the Rip1Tag2 transgenic mouse model. Clin Cancer Res. 2015;21(21):4856-4867. [Abstract] [Google Scholar]
276. Chu X, Gao X, Jansson L, Quach M, Skogseid B, Barbu A.. Multiple microvascular alterations in pancreatic islets and neuroendocrine tumors of a Men1 mouse model. Am J Pathol. 2013;182(6):2355-2367. [Abstract] [Google Scholar]
277. Xie L, Duncan MB, Pahler J, et al.  Counterbalancing angiogenic regulatory factors control the rate of cancer progression and survival in a stage-specific manner. Proc Natl Acad Sci U S A. 2011;108(24):9939-9944. [Europe PMC free article] [Abstract] [Google Scholar]
278. Chamberlain CE, German MS, Yang K, et al.  A patient-derived xenograft model of pancreatic neuroendocrine tumors identifies sapanisertib as a possible new treatment for everolimus-resistant tumors. Mol Cancer Ther. 2018;17(12):2702-2709. [Europe PMC free article] [Abstract] [Google Scholar]
279. Quinn TJ, Yuan Z, Adem A, et al.  Pasireotide (SOM230) is effective for the treatment of pancreatic neuroendocrine tumors (PNETs) in a multiple endocrine neoplasia type 1 (MEN1) conditional knockout mouse model. Surgery. 2012;152(6):1068-1077. [Europe PMC free article] [Abstract] [Google Scholar]
280. Walls GV, Stevenson M, Soukup BS, et al.  Pasireotide therapy of multiple endocrine neoplasia type 1-associated neuroendocrine tumors in female mice deleted for an Men1 allele improves survival and reduces tumor progression. Endocrinology. 2016;157(5):1789-1798. [Europe PMC free article] [Abstract] [Google Scholar]
281. Lopez CL, Joos B, Bartsch DK, et al.  Chemoprevention with Somatuline delays the progression of pancreatic neuroendocrine neoplasms in a mouse model of multiple endocrine neoplasia type 1 (MEN1). World J Surg. 2019;43(3):831-838. [Abstract] [Google Scholar]
282. Ito T, Igarashi H, Uehara H, Berna MJ, Jensen RT.. Causes of death and prognostic factors in multiple endocrine neoplasia type 1: a prospective study: comparison of 106 MEN1/Zollinger-Ellison syndrome patients with 1613 literature MEN1 patients with or without pancreatic endocrine tumors. Medicine (Baltimore). 2013;92(3):135-181. [Europe PMC free article] [Abstract] [Google Scholar]
283. Gatta-Cherifi B, Chabre O, Murat A, et al.  Adrenal involvement in MEN1. Analysis of 715 cases from the Groupe d’etude des Tumeurs Endocrines database. Eur J Endocrinol. 2012;166(2):269-279. [Abstract] [Google Scholar]
284. van Leeuwaarde RS, Dreijerink KM, Ausems MG, et al.  MEN1-dependent breast cancer: indication for early screening? Results from the Dutch MEN1 Study Group. J Clin Endocrinol Metab. 2017;102(6):2083-2090. [Abstract] [Google Scholar]
285. Goswami S, Peipert BJ, Helenowski I, Yount SE, Sturgeon C.. Disease and treatment factors associated with lower quality of life scores in adults with multiple endocrine neoplasia type I. Surgery. 2017;162(6):1270-1277. [Abstract] [Google Scholar]
286. Peipert BJ, Goswami S, Yount SE, Sturgeon C.. Health-related quality of life in MEN1 patients compared with other chronic conditions and the United States general population. Surgery. 2018;163(1):205-211. [Abstract] [Google Scholar]
287. Peipert BJ, Goswami S, Helenowski I, Yount SE, Sturgeon C.. Financial burden is associated with worse health-related quality of life in adults with multiple endocrine neoplasia type 1. Surgery. 2017;162(6):1278-1285. [Abstract] [Google Scholar]
288. van Leeuwaarde RS, Pieterman CRC, Bleiker EMA, et al.  High fear of disease occurrence is associated with low quality of life in patients with multiple endocrine neoplasia type 1: results from the Dutch MEN1 Study Group. J Clin Endocrinol Metab. 2018;103(6):2354-2361. [Abstract] [Google Scholar]

Articles from Endocrine Reviews are provided here courtesy of The Endocrine Society

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/95182537
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/95182537

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1210/endrev/bnaa031

Supporting
Mentioning
Contrasting
0
70
0

Article citations


Go to all (53) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

NIHR Oxford Biomedical Research Centre

    National Institute for Health Research (NIHR)

      National Institute of Diabetes and Digestive and Kidney Diseases

        National Institutes of Health

          Wellcome Trust