ArticlePDF AvailableLiterature Review

Current understanding of pathogenetic mechanisms in neuroendocrine neoplasms

Authors:

Figures

Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=iere20
Expert Review of Endocrinology & Metabolism
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iere20
Current understanding of pathogenetic
mechanisms in neuroendocrine neoplasms
Roberta Modica, Alessia Liccardi, Roberto Minotta, Giuseppe Cannavale, Elio
Benevento & Annamaria Colao
To cite this article: Roberta Modica, Alessia Liccardi, Roberto Minotta, Giuseppe Cannavale,
Elio Benevento & Annamaria Colao (03 Nov 2023): Current understanding of pathogenetic
mechanisms in neuroendocrine neoplasms, Expert Review of Endocrinology & Metabolism,
DOI: 10.1080/17446651.2023.2279540
To link to this article: https://doi.org/10.1080/17446651.2023.2279540
Published online: 03 Nov 2023.
Submit your article to this journal
Article views: 146
View related articles
View Crossmark data
REVIEW
Current understanding of pathogenetic mechanisms in neuroendocrine neoplasms
Roberta Modica
a
, Alessia Liccardi
a
, Roberto Minotta
a
, Giuseppe Cannavale
a
, Elio Benevento
a
and Annamaria Colao
a,b
a
Endocrinology Unit, Department of Clinical Medicine and Surgery, Federico II University, Naples, Napoli, Italy;
b
UNESCO Chair “Education for
Health and Sustainable Development, ” Federico II University, Naples, Italy
ABSTRACT
Introduction: Despite the fact that important advances in research on neuroendocrine neoplasms
(NENs) have been made, consistent data about their pathogenetic mechanism are still lacking.
Furthermore, different primary sites may recognize different pathogenetic mechanisms.
Areas covered: This review analyzes the possible biological and molecular mechanisms that may lead
to NEN onset and progression in different organs. Through extensive research of the literature, risk
factors including hypercholesterolemia, inflammatory bowel disease, chronic atrophic gastritis are
evaluated as potential pathogenetic mechanisms. Consistent evidence is available regarding sporadic
gastric NENs and MEN1 related duodenopancreatic NENs precursor lesions, and genetic-epigenetic
mutations may play a pivotal role in tumor development and bone metastases onset. In lung neuroen-
docrine tumors (NETs), diffuse proliferation of neuroendocrine cells on the bronchial wall (DIPNECH) has
been proposed as a premalignant lesion, while in lung neuroendocrine carcinoma nicotine and smoke
could be responsible for carcinogenic processes. Also, rare primary NENs such as thymic (T-NENs) and
Merkel cell carcinoma (MCC) have been analyzed, finding different possible pathogenetic mechanisms.
Expert opinion: New technologies in genomics and epigenomics are bringing new light to the
pathogenetic landscape of NENs, but further studies are needed to improve both prevention and
treatment in these heterogeneous neoplasms.
ARTICLE HISTORY
Received 25 May 2023
Accepted 1 November 2023
KEYWORDS
Cancer;
gastroenteropancreatic;
medullary thyroid
carcinoma; Merkel cell
carcinoma; neuroendocrine
neoplasm; neuroendocrine
tumor; pathogenetic
mechanism
1. Introduction
Neuroendocrine neoplasms (NENs) are a heterogeneous group of
malignancies arising from cells with a neuroendocrine phenotype,
scattered throughout various organs and systems in the body
[1,2]. Well-differentiated neuroendocrine tumors are characterized
by their unique ability to produce and release bioactive sub-
stances, including hormones and neurotransmitters [2].
Importantly, according to the SEER database their incidence is
increasing, partly due to improved diagnostic techniques and
awareness [3]. The complex interplay between the nervous and
endocrine systems gives rise to the intricate biology of these
neoplasms, making their diagnosis, management, and treatment
challenging yet fascinating [4–6]. NENs are mostly sporadic and
localized in the gastroenteropancreatic (GEP) tract (70%) and
bronchopulmonary system (25–30%), with a small percentage,
about 10%, occurring in association with hereditary syndromes
such as multiple endocrine neoplasia type 1 (MEN1), neurofibro-
matosis type 1 (NF1, von Recklinghausen’s disease), and Von
Hippel-Lindau (VHL) disease [2]. Understanding the etiology, clin-
ical presentation, and molecular mechanisms underlying neuroen-
docrine neoplasms is crucial for health-care professionals involved
in their care [4]. NENs are mainly nonfunctional diseases with an
indolent course in absence of specific clinical presentation or
symptoms, which leads to a delayed diagnosis. In view of their
heterogeneity, the World Health Organization (WHO) classifies
GEP-NENs into three major categories: well-differentiated neu-
roendocrine tumors (NETs), poorly differentiated neuroendocrine
carcinomas (NECs), and mixed neuroendocrine-non-
neuroendocrine neoplasms (MiNENs) [7,8]. GEP-NETs are further
classified into three grades based on the proliferative rate, includ-
ing grade 1 (G1), grade 2 (G2), and grade 3 (G3) [7]. The prolifera-
tive rate is assessed by the mitotic index and the Ki-67 labeling
index, which is a marker of cell proliferation [7]. G1 tumors have
a low proliferative rate, with a mitotic index of less than 2 and a Ki-
67 labeling index of less than 3% [7]. G2 tumors have a moderate
proliferative rate, with a mitotic index of 2 to 20 and a Ki-67
labeling index of 3% to 20% [7]. G3 tumors have a high prolifera-
tive rate, with a mitotic index of more than 20 and a Ki-67 labeling
index of more than 20% [7]. NECs are poorly differentiated NET
with a high proliferative rate and a Ki-67 labeling index of more
than 20% [7]. MiNENs are tumors composed of both neuroendo-
crine and non-neuroendocrine components, with each compo-
nent comprising at least 30% of the tumor [7]. Thoracic NENs
comprise typical carcinoids (TCs), atypical carcinoids (ACs), large-
cell neuroendocrine carcinoma (LCNEC), and small-cell lung carci-
noma (SCLC) divided according to mitotic rate and presence or
absence of necrosis [7,8]. Localized disease has a five-year survival
rate ranging from 78% to 93%, while in metastatic disease it
decreases to 19–38% with lymph node, liver, and bone being
the main sites of metastases [9,10]. Thus, morphological and
CONTACT Roberta Modica robertamodica@libero.it Endocrinology Unit, Department of Clinical Medicine and Surgery, Federico II University, Naples,
Napoli, Italy
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM
https://doi.org/10.1080/17446651.2023.2279540
© 2023 Informa UK Limited, trading as Taylor & Francis Group
functional examinations are requested to complete staging.
European Neuroendocrine Tumor Society (ENETS) guidelines
mainly suggest performing CT scan or MRI depending on site of
primary lesion, and 68 Ga-PET/CT as functional exam for low-grade
NENs, while FDG-PET is pivotal for G3 NENs (ENETS consensus,
Perren, 2017). Unfortunately, nowadays reliable diagnostic bio-
chemical markers are lacking and the role of chromogranin
A (CgA) and neuron-specific enolase (NSE) are debated, and no
routinary use is suggested [11].
Despite a growing attention regarding NENs, still little is
known about their physiopathology and underlying pathoge-
netic mechanisms. The pathogenesis of NENs is complex and
multifactorial, involving a combination of genetic, epigenetic,
and environmental factors [12–15]. Molecular studies have
shed light on the genetic alterations and signaling pathways
involved in the development and progression of NENs
[9,10,16]. In particular, the genetic mutations underlying
familiar syndrome associated with NENs are well recognized.
On the contrary, little is known about biology and early devel-
opment of these diseases regarding the possible evolution of
preneoplastic lesions into cancer both in sporadic and familial
NENs.
For the detection of molecular alterations, the most used
methods are monoclonal antibodies and genetic sequencing
of tumor cells with diagnostic and therapeutic purposes, and
this field is under development [17].
In this paper, we will provide a comprehensive overview of
our understanding of pathogenetic mechanisms in NENs, ana-
lyzing the current state of knowledge regarding their physio-
pathology, highlighting the most important biologic and
morphologic aspects according to anatomical site and their
relationship with classification, epidemiology, pathology, and
clinical manifestations.
2. Search methodology
Deepened research on ‘‘Pubmed’’ using the following terms:
‘neuroendocrine neoplasm,’’ ‘‘gastroenteropancreatic neu-
roendocrine tumor,’ ‘‘thymic neuroendocrine tumor,’’ ‘‘carci-
noid tumor,’’ ‘‘Merkel cell carcinoma,’’ ‘‘chronic atrophic
gastritis,’’ ‘‘MEN1,’’ ‘‘VHL,’’ ‘‘NF1,’’ “DIPNECH”,“medullary thyroid
carcinoma,” pathophysiology,’’ ‘‘preneoplastic lesion,’’ ‘‘envir-
onmental factor,’’ ‘‘neuroendocrine epithelial bodies,’’ and
pathogenetic mechanism” was carried out. No start date was
set and the last update was in April 2023. Also, manual search
was conducted within the reference lists of the identified
papers to add relevant studies. Among the reports identified
by the search strategy, all papers containing data about the
selected topic were considered. Abstracts were excluded. All
the identified references were assessed by title and abstract to
determine possible eligibility and papers were included if they
reported data about pathogenetic mechanisms in NENs. Only
articles published in the English language were considered.
3. GEP-NETs
The main site of NETs onset is the gastrointestinal tract, and
the vast diversity of its cells conveys GEP-NENs heterogeneity
[16]. GEP-NETs represent 1–4% of all gastrointestinal neo-
plasms and over the years, a steadily rising incidence of GEP-
NETs has been observed [3,16]. Consequently, a better under-
standing of their pathogenetic mechanisms could help to
identify the reason of these epidemiological rise and provide
an integration other identified predisposing and risk factors,
including environmental factors, sex, metabolic syndrome, and
nutrition, low vitamin D levels, microbiota, thus preventing
them and allowing clinicians to predispose prevention and
treatment strategies [10,12,18,19]. Different pathogenic
mechanisms have been proposed according to different pri-
mary. Sequential changes from hyperplasia to neoplasia have
been described in NETs [20]. Indeed, enterochromaffin-like
(ECL) cell hyperplasia is defined by at least two linear chains
per millimeter (linear) or at least five cells per gland (multi-
cellular), or one micronodule smaller than 150 m/mm (micro-
nodular) on bioptic sample, and the fusion of these elements
or the invasion of lamina propria could lead to neoplasm
Article highlights
Neuroendocrine neoplasms (NENs) may recognize different pathoge-
netic mechanisms according to different primaries and the onset of
precursor lesions, as well as genetic alterations seem to play a pivotal
role both in sporadic and familial neoplasms.
Among gastroenteropancreatic NENs, in small intestinal neuroendo-
crine tumors (SI-NETs) a carcinogenic process has been hypothesized
considering small intestinal enteroendocrine cells as a source of stem
cells. Genomic stability with no recurrent mutation has been reported
but according to epigenetic mechanisms specific miRNAs may play
an important role in SI-NETs development and prognosis: downregu-
lation of miR-133a, miR-145, miR-146, miR-222, and miR-10b and the
upregulation of miR-183, miR-488, and miR-19aCb has been found in
metastatic SI-NETs.
In Type 1 gastric NETs (G-NETs) associated with chronic atrophic
gastritis, a transition from atrophic gastritis to hyperplastic/dysplastic
neuroendocrine nodules and ultimately to neoplasia due to hyper-
gastrinemia stimulation has been proposed. In Type 2 G-NETs the
hypertrophic oxyntic mucosa seems to be stimulated by gastrinoma
in MEN1 patients and interestingly MEN1 mutation has been reported
in 30-40% of cases of Type 3 G-NETs, although not associated with
preneoplastic lesions. The chronic use of proton pump inhibitors (PPI)
causing hypergastrinemia and cell hyperplasia has been proposed in
the development of G-NENs, though genetic predisposition or envir-
onmental factors may also play a role.
MEN1-related pancreatic neuroendocrine tumors (pNETs) seem to
originate from pancreatic microadenomas and from non-islet tissue.
Main driver mutations in sporadic pNETs derive from MEN1, DAXX,
and ATRX alterations.
Increased prevalence of colorectal NETs has been observed in
patients with inflammatory bowel diseases, NETs did not originate
from areas with chronic inflammation leading to the hypothesis that
NETs development was not directly correlated to a proinflammatory
microenvironment and even the presence of precursor lesions is
debated.
Nicotine and hypoxia could be crucial in the pathogenetic mechan-
ism of lung carcinoids and carcinomas development, in particular
influence directly the signaling cascades Pi3K/AKT/mTOR and MAPK
both pivotal for cell proliferation, while VEGF secretion mediated by
the inflow of Ca2+ could lead to an increase of angiogenesis.
In medullary thyroid cancer (MTC) RET mutations are drivers for
cancer development in familial forms, but also RAS gene mutations
and epigenetic alteration seem to be pivotal, in particular in sporadic
MTC.
Data regarding pathogenetic mechanisms in NENs are currently
mainly available in gastro-entero-pancreatic (GEP) NENS, maybe due
to their relatively higher frequency, but better knowledge is still of
utmost importance in this field, to improve cancer prevention and
treatment strategies.
2R. MODICA ET AL.
development [20]. Molecular and genetic background that
may participate in the transition from naïve cell to hyperplastic
lesion and subsequently to malignancy is complex and not yet
completely recognized [21]. Apparently, these mechanisms are
different between pancreatic NEN (pNEN) and gastrointestinal
NET [21].
Small intestine NET s (SI-NET s) are the most common GEP-
NET s with an incidence of 1.05/100,000 per year [22]. No
precursor lesions have been observed for SI-NETs, on histolo-
gical exam of surgical specimen [23]. However, SI-NENs may
undergo a multiphase carcinogenic process due to the pre-
sence of small intestinal enteroendocrine cells, defined as an
active reserve of intestinal stem cells, taking part in the intest-
inal mucosa’s ongoing self-renewal and expressing a specific
molecular pattern (BMI1, HOPX, TERT, and Lrig1) [24]. Indeed it
has been proposed that these cells are able to dedifferentiate
leading to SI-NETs [24]. Moreover, SI-NETs seem to be less
associated with the specific molecular events seen in most
other NENs [25]. A genomic stability with no recurrent muta-
tions was observed in a study of 48 SI-NETs using a massively
parallel exome sequencing, on the other hand somatic copy
number alterations are common in with the loss of chromo-
some 18 found in over 60% SI-NETs [26,27]. Epigenetic
mechanisms are frequently involved in SI-NETs, indeed both
global hypomethylation and CpG island methylator pheno-
type have been described [28–30]. These DNA alterations
could lead to silent TCEB3C, implicated in tumor development,
and `to downregulate RASSF1A and CTNNB1 that are related
to a metastatic behavior [31,32]. Similarly, in metastatic SI-
NETs the downregulation of miR-133a, miR-145, miR-146,
miR-222, and miR-10b and the upregulation of miR-183, miR-
488, and miR-19aCb was observed and these alterations are
supposed to take part in pathogenetic mechanism [33]. Other
miRNA that could contribute to tumor dissemination are miR-
96, miR-182, miR-183, miR-196a, and miR-200a upregulated
and miR-31, miR-129-5p, miR-133a, and miR-215 downregu-
lated [34]. These data helped to classified SI-NETs in three
subgroups: one with chromosome 18 LOH-CDKN1B mutations,
found in older patients and with a better prognosis, another
with high degree of CpG island methylator phenotype but
lacking SCNAs with intermediate age at diagnosis and prog-
nosis and the last with a lot of copy number changes, in
younger patients and with a poor prognosis [35]. Lately, the
role of lipids has been evaluated as a risk factor in SI-NETs [13].
Indeed, lipids could support tumor development and progres-
sion, participating in the membrane phospholipids turnover.
Furthermore saturated fatty acids may protect the tumor cell
from the microenvironment oxidative stress [13]. Consistently,
a prospective study showed that meat and high fat intake are
associated with SI-NETs development [36]. Pancreatic NETs
(pNETs) have an incidence of 0.48/100.000 [22]. Several studies
investigated precursor lesions in pNETs, especially in heredi-
tary tumors, while in sporadic pNETs no precursor lesions have
yet been described [37]. In MEN1 patients pancreatic micro-
adenomas (<5 mm) are common findings and macroadenoma
(>5 mm) seem to originate from these lesions [38,39].
Furthermore, Vortmeyer et al. studied pancreas of MEN1
patients with no lesions and demonstrated a non-islet cell
origin of pNENs, in particular in the ductal/acinar system.
Indeed, these cells retain pluripotency and are able to form
islet-like structures and could be the real target of a ‘second
hit’ leading to pNENs [40]. The loss of heterozygosity has been
investigated as a marker for neoplastic growth in microadeno-
mas, macrotumors and monohormonal endocrine cells clus-
ters (MECCs) [41]. Loss of heterozygosity of MEN1 was
observed in 86% of macrotumors, in 100% of microadenomas
and, interestingly, in 95% of MECCs; these lesions could be
a microscopic form of microadenomas [41]. On the other
hand, morphologically normal islets and exocrine tissue
showed a retention of heterozygosity of MEN1 [41]. It is still
unclear if the mechanisms observed in MEN1 related pNENs
are similar to sporadic pNENs, because no precursor lesion has
been identified in these patients [41]. Molecular analysis of
pNENs has revealed several genetic alterations, such as inacti-
vating mutation of MEN1, DAXX (death-domain-associated
protein), and ATRX thalassemia/mental retardation syn-
drome X-linked) genes, involved in chromatin remodeling in
telomeric regions [42,43]. The MEN1 gene mutations can be
observed in hereditary tumors in patients with MEN1 syn-
drome, nevertheless in more than 35% of sporadic pNENs
this mutation occurs in a somatic way [42]. A mutation of
one of DAXX and ATRX is present in more than 45% of
sporadic pNETs, suggesting that they share the same pathway
[42]. DAXX/ATRX mutations lead to alternative lengthening of
the telomeres (ALT) phenotype and chromosomal instability
[44]. Less frequent mutations, observed in 15% of pNENs,
include PTEN, TSC2, TSC1, DEPDC5, and PIK3CA, all involved in
the mTOR pathway [45].
Gastric NETs (G-NETs) have an annual incidence of 0.4/
100,000 individuals [46]. These heterogeneous lesions have
been further divided into ECL cell NENs and antral NENs
based on the kind of neuroendocrine cell development [47].
Antral NENs represent the smaller part of G-NETs and are not
associated with precursor lesions, indeed neuroendocrine cell
dysplasia in the antrum has never been documented [47].
Three types of ECL NETs (Types 1, 2, and 3) are recognized
and precursor lesions have been correctly identified and
described in the context of Type 1 and Type 2 [20,46,48–50].
The most common G-NETs are Type 1 (80–90%), followed by
Type 3 (10–15%) and Type 2 (5–7%) [46]. Type 1 develops in
the presence of atrophic gastritis type A, a chronic inflamma-
tory gastric disease that selectively affects the fundus and
body of the stomach leading to hypo/achlorhydria that pro-
gresses to hypergastrinemia thanks to a compensatory hyper-
plasia of the antral gastrin cells [46,51–55]. Precursor lesions
that take place in a context of atrophic mucosa can be dys-
plastic (larger micronodules, joined micronodules, microno-
dules with new stroma, and microinfiltrative lesions) and
hyperplastic (diffuse, linear, micronodular, and adenomatoid
hyperplasia), this two conditions can evolve into a neoplasm,
this two conditions can evolve into a neoplasm [55,56]. TGF-α,
bFGF, or MEN1 gene alterations could represent the molecular
mechanisms underlying this transition [25]. Moreover,
a recently published whole-exome analysis on a familial clus-
ter has shown in 5 of 10 siblings with G-NETs Type 1 the
presence of a germline mutation in the ATP4A gene, which
codes for the subunit of the stomach proton pump [57]. On
the other hand, Type 2 originates from hypertrophic oxyntic
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 3
mucosa stimulated by gastrinoma, mostly duodenal, or pan-
creatic, in patients with MEN1 with Zollinger-Ellison syndrome
(ZES) [58–60]. Typically, there is a hypertrophic mucosa that
may cause a dysplastic ECL-cell proliferation observed only in
MEN1 patients. Hence, ECL cells appear to be more susceptible
to the mitogenic effect of hypergastrinemia due to menin
mutation [61]. Type 3 has no precursor lesion considering
that it originates from normal oxyntic mucosa [46]. These
aggressive G-NETs have demonstrated alterations of the
MEN1 locus in about 30–40% of cases [7]. Lately, other two
type of G-NETs have been hypothesized: Type 4, where hyper-
gastrinemia and hyperplasia of parietal cell lead to G-NET in
non MEN1 patient probably due to a defect in acid secretion;
Type 5 caused by the inappropriate chronic use of proton
pump inhibitor (PPI) [46,60,62]. Interestingly, a role of PPI has
been proposed in the pathogenetic mechanism of G-NETs.
A meta-analysis of six randomized controlled trials showed
that patients treated with PPI for more than 6 months were
significantly at risk of developing diffuse or linear/micronodu-
lar ECL cell hyperplasia than controls, although no NETs were
observed and only a mild hypergastrinemia was observed
(100–500 pg/mL) [63]. Several case reports have documented
the association between G-NETs and chronic PPI use with
a long follow-up time >10 years, speculating that much more
time is necessary to develop a G-NETs [64,65]. Alternatively,
since the majority of patients treated for a long time with PPI
have hypergastrinemia without developing G-NETs, it may be
suggested that hypergastrinemia alone is not sufficient and
another trigger is needed, such as genetic predisposition or
environmental factors [65–68]. Taken together, these data do
not clarify the role of PPI in the pathogenesis of G-NETs, which
remains still debated.
With regard to duodenal NETs (dNETs) representing up to
22% of NETs with an incidence of 0,34/100.000, different
entities have been recognized according to the cell pathology:
G-cell (>60%), D-cell (21%), gangliocytic paraganglioma (9%)
[23,69,70]. dNETs are mostly sporadic and nonfunctioning,
affecting patients of 50–70 years old and have a slight female
prevalence [71]. F Functioning dNENs are generally associated
with MEN1, showing a more aggressive manifestation in
younger patients with multiple primaries [72]. Interesting,
ampullary or periampullary NETs are more common in other
two genetic syndromes: NF1 and VHL [73]. Nevertheless, D-cell
dNENs have been associated more commonly with mutation
in NF1 gene than in MEN1 gene. In these tumors INFB1 muta-
tions and loss of chromosome 22 have been identified [25]. No
precursor lesion has been identified in sporadic dNENs and in
the context of NF1 or VHL. In case of MEN1, precursor lesion
has been linked to gastrin or somatostatin producing NETs
[74]. The causing mutation of the syndrome can influence the
type of lesion in the duodenum and allelic deletion of the
MEN1 gene is associated with microinvasive lesions, while
hyperplastic endocrine cells preserve heterozygosity [23,75].
Indeed, in case of MEN1 duodenal gastrinomas both diffuse
G-cell hyperplasia (simple, linear, micronodular or macronod-
ular neuroendocrine cell hyperplasia) and multicentric gastrin
producing microtumors (between 300 microns and 2 mm)
have been described, while no one of these lesions was
found in sporadic duodenal gastrinomas, thus suggesting dif-
ferent pathogenetic mechanisms [69]. In particular, in the
hyperplastic G-cell there was not loss of heterozygosity on
chromosome 11q13, even though they have germline MEN1
mutation, testifying that this event is crucial for neoplasm
development and maybe G-cell hyperplasia is secondary to
a greater susceptibility of MEN1 G-cell to an unknown growth
factor [69]. Moreover, a recent study focused on PPI showed
an association between G-cell hyperplasia in patients with
H. pylori gastritis treated with PPI and sporadic gastrinoma
[76]. However, data about duodenal precursor lesions are less
consistent and more studies about this topic should be
proposed.
The existence of preneoplastic lesions in esophageal NETs
(E-NETs) is debated, considering that E-NENs are extremely
rare. Only 0.04–1% of GEP-NETs are E-NETs, generally diag-
nosed during diagnostic examination when suspecting eso-
phageal adenocarcinomas or squamous-cell carcinomas,
developing in the lower third of the esophagus, as a single
lesion, but sometimes as an ulcerated or fungating mass [77].
Since E-NETs are typically a random finding on endoscopic
examination, early diagnosis is therefore difficult, but smoking
and alcohol abuse are acknowledged as major risk factors [78].
E-NENs can be found associated with adenocarcinoma and
Barrett mucosa or as a single large polypoid/nodular tumor
[79]. Mostly, E-NETs are carcinomas (90%) with focal or mild
expression of immunohistochemical markers, synaptophysin
and chromogranin A E-NETs and arise in the lower part of
the esophagus, probably because the bulk of neuroendocrine
cells is found in the lower esophageal submucosal [77,80]. To
date, no precursor lesions have been identified for E-NETs,
although the existence of endocrine cell hyperplasia also in
adenocarcinoma and Barrett mucosa supports the idea that
these lesions develop from a single stem cell who could
differentiate into goblet, Paneth, or endocrine cells [79].
Colon and rectal NETs account only for the 0.4% of all
malignancies of this district, their incidence is 1,04/100.000
individuals [22]. Areas of neuroendocrine differentiation of
less than 30% are often found in adenocarcinomas [81].
An increased colo-rectal NETs prevalence has been
described in patients with a chronic inflammatory bowel dis-
ease (IBD) undergoing surgery because of disease complica-
tion [82]. Albeit a standardized incidence ratio of colo-rectal
NETs of 2,5 for Crohn disease and 2,0 ulcerative colitis has
been identified, NETs did not originate from areas with chronic
inflammation leading to the hypothesis that NETs develop-
ment was not directly correlated to a proinflammatory micro-
environment and even the presence of precursor lesions is
debated [83,84]. Mucosal enterochromaffin cell (EC) hyperpla-
sia has been reported in the tissue surrounding NETs in these
patients, while EC dysplasia has never been described in these
districts [23,85]. Nevertheless, chronic inflammation could play
a role in the development of colon-rectal NENs, stimulating
endocrine cells proliferation [86].
NENs represent 50% to 70% of all appendiceal malignan-
cies, with an incidence of 0,15–06/100.000 people [87,88].
Appendiceal NENs (a-NENs) are generally single and mainly
on the tip of the organ (75% of cases) [89]. The age of onset is
4R. MODICA ET AL.
reduced compared to other sporadic NENs (30–40 years) [89].
Compared to colo-rectal NENs, there is no association with IBD
and mucosal EC hyperplasia [90]. In a-NENs an association with
Schwann cells has been observed. In particular, a subepithelial
aggregate composed of neurons, Schwann cells and neuroen-
docrine cells (subepithelial neuroendocrine complex, SNC) has
been recognized in the mucosa surrounding lesion [85,91].
The SNCs may sometimes be symptomatic by themselves,
representing the pathophysiologic substrate for the neuro-
genic appendicopathy, that leads to an increased release of
neuropeptides with the consequent onset of acute abdominal
pain mimicking acute appendicitis [92].
Several studies have observed that GI-NECs originate from
the same multipotent stem cells that may give rise to non-
NECs. NECs are very rare tumors with an incidence of 4,5/
1.000.000 people [22]. This suggestion derives from the fact
that often has been observed the presence of a well-
differentiated adenocarcinoma/adenoma overlying the
aggressive neuroendocrine component of MiNENs [93].
Moreover, GI-NECs share molecular alteration with non-NECs
lesions, such as the presence of a KRAS/BRAF, RB1, TP53,
CDKN2A, and ERBB2 mutation [94]. In gastric NEC, the RB1
mutation was the most distinctive, and these mutations have
not been observed in gastric neuroendocrine tumors [95]. In
a recent study, authors found that in tumor samples of colonic
NECs the most frequent mutations were TP53, BRAF, APC, and
KRAS, while in rectal tumors were TP53, APC, FBXW7, and
KRAS [96]. Some authors observed a BRAF mutation rate of
20% in a series of colorectal NECs while Idrees et al. reported
that all BRAF-mutated NECs were microsatellite stable [93,97].
Probably, NETs and NECs present different pathogenetic path-
ways and originate from different precursor cells, but because
of the rarity of these malignancies a clear pathway has not
been elucidated [98,99]. For example, RB1 mutations and
chromosomal abnormalities (extensive deletions and translo-
cations) were common in GI-NECs; TP53 and RB1 alteration are
prevalent in these neoplasms, and they contribute to neuroen-
docrine tumorigenesis, in which loss of TP53 and RB1 leads to
NECs, as confirmed by a murine model [100]. In precursor
lesions of GEP-NETs, such as pNETs, there are no mutations
or alterations of TP53, RB1, or other driver mutations common
in adenocarcinomas; instead, mutations of DAXX, MEN1, and
ATRX are observed [99]. Furthermore, NECs are anecdotally
associated with IBD thanks to some case series [101,102]. In
a case series of 14 patients with IBD and NENs, only in three
cases NEC was diagnosed [103].
3.1. LUNG-NENs
Pathological classification of lung NENs (L-NENs) has
evolved over years, reflecting the need of a better charac-
terization and recently a new histological diagnosis of car-
cinoid not-otherwise-specified (NOS) has been added to
TCs, ACs, SCLCs, and LCNECs (Figure 1) [104]. DIPNECH has
been classified as a premalignant lesion by the World
Health Organization, characterized as a histological entity
on incidental findings of lung nodule biopsies, more com-
mon in female rather than in male [105,106]. The preva-
lence of DIPNECH in female may support a role of gender
predisposition due to hormone profiling, but unfortunately
data are still lacking [97]. DIPNECH is characterized by
superficial and isolated lesions, or with basement mem-
brane invasion forming tumorlets (cell aggregates <5 mm)
or carcinoid tumors (if >5 mm) [105]. Clinically prognosis of
carcinoid is better than the one of carcinoma and patients
with TCs have got the best survival outcomes, but just few
data on pre-clinical differences between these two groups
are present in literature [107,108]. Patients with carcinoid
are usually younger than patients with carcinomas, as med-
ium age of onset in TCs is 45 years, while in SCLCs is 70
years [109,110]. This may imply that carcinoid’s cells may
have less time to accumulate genetic mutations in compar-
ison with that of carcinoma, as confirmed by microsatellite
marker analyses, and SCLCs have the highest mutational
rate [109]. There is not only a quantitative difference, but
also a qualitative one: loss of heterozygosity (LOH) for
Figure 1. Lung NEN. NEB: neuroendocrine body. α7nAChR: alpha-7 nicotinic acetylcholine receptors.
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 5
carcinoid often regards locus 11q, that includes MEN 1,
a tumor suppressor gene, while for carcinoma it often
regards locus 13q, that includes Rb gene, a proto-
oncogenic tumor suppressor gene (Table 1) [108,111].
Even p53, another important tumor suppressor gene, is
mutated in carcinomas, while it is normally expressed in
carcinoid [112]. Despite these differences, it has been
proved that for all lung NEN the activation of mammalian
target of rapamycin (mTOR) pathway is pivotal, something
of interest for therapeutic strategies [112]. Patients with
carcinomas are predominantly smokers, while smoking sta-
tus seems to be not relevant for carcinoid development
[109]. Lung NENs arise from pulmonary neuroendocrine
cells that are mostly organized in bodies (NEBs), well inner-
vated [113]. It has been proved that NEBs have got specific
receptors to both hypoxia and nicotine that cause not only
a secretion of serotonin, that could lead to pulmonary
fibrosis and to pulmonary hypertension, but also
a proliferative response [114,115]. Nicotine, inhaled through
cigarette and e-cigarette smoking, has a direct signaling
cascades that promote cancer development [116]. When
the homomeric ionic alpha-7 nicotinic acetylcholine recep-
tors (α7nAChR) are activated, different pathways start, in
particular Pi3K/AKT/mTOR and MAPK both pivotal for cell
proliferation, while VEGF secretion mediated by the inflow
of Ca2+ could lead to an increase of angiogenesis [116].
Moreover, a study on primate models proved that prenatal
nicotine exposure increases NEBs formation [114]. Thus,
nicotine could have a role in causing hyperplasia, while
dysplasia and carcinogenesis could occur after prolonged
exposition to smoke. Only 3% of SCLC is reported in not-
smoker population [117]. Interesting 25% of them have got
EGFR mutations, suggesting different mutation drivers lead-
ing to these cancers development and different therapeutic
strategies [118]. Usually, this pathway is not involved in
SCLC [119]. In fact, targeted therapies for EGFR-mutated
lung adenocarcinomas could lead to transformation to
SCLC or occasionally LCNEC [119]. This has been documen-
ted in 5–14% of patients receiving first (erlotinib), second
(afatinib), and third (osimertinib) generation EGFR inhibitors.
Lastly, SCLC in a not-smoker patient may represent an
unsuspected metastasis, such as from HPV-related sites
like cervix or oropharynx [120]. Recently, different sub-
types of both SCLC and LCNEC have been described, but
their physiopathology is still not clear [121]. Interestingly, it
has also been considered immuno-receptor expression in
these tumors that, even if not prognostic for immunother-
apy response, is the pathological landmark of an immune
cells involvement during cancer development [4,122].
3.2. RARE NENs
3.2.1. Thymic NENs
Thymic neuroendocrine neoplasms (T-NENs) are rare neo-
plasms representing approximately 5% of thymic and med-
iastinal neoplasms and 0.4% of all neuroendocrine neoplasms
(NENs) [123].
T-NENs, as well as L-NENs, are traditionally classified using
the same criteria into TCs and AC, LCNECs and SCCs. T-NENs
TCs and ACs are more frequent in males, whereas there is
a female preponderance in L-NENs [123]. This sex difference
has not been described for LCNEC and SCC however, it has not
yet been possible to find a pathophysiological mechanism
underlying this gender difference [123]. AC and LCNEC are
by far the most frequent subtypes in the thymus, while SCCs
and TCs prevail in the lung. Interestingly, unlike lung LCNECs
and SCCs, T-NENs are not associated with cigarette smoking
[124]. T-NENs also have a worse prognosis than L-NENs with
5-year survival rates varying between the sundry subtypes,
decreasing from 50–70% in TC and AC to 30–66% in LCNEC
and to 0% (median survival 13–26 months) in SCCs [125]. Only
a few data are available about pathogenic mechanisms in
T-NENs. A study using comparative whole-genome hybridiza-
tion (CGH) showed an increasing mutational profile among
the various subtypes of T-NENs [126]. In particular, three clus-
ters based on chromosomal instability were highlighted.
Cluster 1 characterized by a low instability index consisted
mainly of TC and AC but also some LCNECs, which sometimes
showed a profile overlapping with TC and AC [126].
Furthermore, sequencing of tissue from the primary and
metastases showed heterogeneity of the neoplasm with the
possibility of evolution from one subtype to another [125,126].
In addition, in thymic LCNEC this study showed amplification
of the myc oncogene, myc contributes to tumor development
is its ability to promote cell proliferation [125]. Myc regulates
the expression of genes involved in cell cycle progression and
DNA replication, leading to uncontrolled cell division [127].
Additionally, myc influences cellular metabolism, angiogen-
esis, and genomic instability [125,127]. Another frequently
encountered mutation is that of MEN 1, indeed approximately
25% of patients with thymic carcinoid tumors harbor a MEN1
germline mutation and 8% of MEN1 patients develop T-NENs
[128,129]. The molecular evaluation of T-NENs lead to identify
pathways involved in differentiated and poorly differentiated
forms [130]. The methyltransferase EZH2, part of PRC2 (poly-
comb repressive complex 2) one of the two classes of poly-
comb-group proteins, is positive in LCNECs and poorly
differentiated forms [130]. EZH2 through methylation is
involved in silencing genes involved in the regulation of cell
cycle. In particular, EZH2 is associated with over expression of
TP53 and is associated with increased proliferation and
decreased survival [124]. A gene that is overexpressed in well-
differentiated forms is ATRX, a transcriptional regulator
required for deposition of histone H3.3 at telomeres that
influence DNA tridimensional structure [131]. T-NENs over-
express PAK3 (p21-activated kinase 3) and its upstream
Table 1. Molecular features of lung neuroendocrine neoplasms (L-NEN).
L-NEN
Loss of
heterozygosity
Gene involved
Typical carcinoid 11q MEN-1
Atypical carcinoid
Large cell neuroendocrine
carcinoma
3p, 5q and 13q Rb
Small cell carcinoma 3p, 4q, 5q, 13q and
15q
Rb. Possible mutation
of p53
6R. MODICA ET AL.
regulator RAC, both of which are involved in the regulation of
cell migration, angiogenesis and invasion [132]. Particularly in
T-NENs ACTH-secreting PAK3 has an important action on
fibroblasts and epithelial cells. Indeed, its activation leads to
increased angiogenesis and invasion activity. In one study, the
presence of this mutation was associated with aggressive and
metastatic disease [132]. Approximately 40% of T-NENs exhibit
ACTH hypersecretion, elevated β-catenin, pro-
opiomelanocortin (POMC) and carboxypeptidase E (CPE) and
decreased NOTCH2 levels [133]. The Notch signaling pathway
plays a critical role in tumorigenesis, and its dysregulation has
been implicated in various cancers. Notch promotes tumori-
genesis through multiple mechanisms, including the regula-
tion of cell proliferation, survival, angiogenesis, and metastasis
[134]. Levels of POMC and CPE have been found increased by
3- and 4-fold respectively in ACTH-secreting TNRNs, however
their role is not yet well known [133]. It is important to note
that the exact mechanisms underlying T-NENs may vary
depending on the specific tumor subtype and individual
cases. Further research is needed to elucidate the precise
pathogenetic mechanisms involved in these tumors and to
identify potential therapeutic targets.
3.3. Merkel cell carcinoma
Merkel cell carcinoma (MCC) is a rare and aggressive neuroen-
docrine neoplasm [135]. MCC represents less than 1% of all
cutaneous carcinomas in Europe, but a 5.4-fold increase from
1986 to 2003 has been observed [136,137]. MCC can be
defined as immunogenic tumor, indeed the introduction of
immunotherapy improved its prognosis [138]. The pathoge-
netic mechanisms of MCC have been in depth analyzed. Two
forms of MCC characterized by two different pathogenetic
mechanisms have been identified: the first, which is found in
80% of cases, is due to the genetic integration of the poly-
omavirus; the second, less frequent, is due to exposure to
ultraviolet rays (UV) (Figure 2) [139–141]. Indeed, the two
main risk factors are immunosuppression (chronic lymphocytic
leukemia, HIV/AIDS, drugs, etc.) and chronic exposure to the
sun, respectively. Although the community has a significant
polyomavirus seropositivity, the incidence of MCC is very low
[139]. Indeed, two crucial events must occur at the same time
to determinate the MCC development: the clonal integration
of the viral small T antigen (ST) and large T antigen (LT) genes
in the cell genome and a mutation that lead to the loss of the
LT protein’s C-terminal expression [139,142]. These events
block viral replication, thereby LT and ST protein synthesis is
boosted, which encourages cell cycle advancement and survi-
val because mutated LT interacts with tumor suppressor pro-
tein retinoblastoma (pRB) involved in the cell cycle
progression. Concurrently, ST protein interacts with tumor
suppressor protein phosphatase 2A and mammalian target
of rapamycin (mTOR) pathway in mammalian cells [139]. UV-
induced MCC expresses a high tumor mutational burden char-
acterized by the ‘UV mutational signature’ that is the presence
of C-to-T pyrimidine dimers. Among these several mutations,
the ones involving RB and p53 are predominant [139].
3.4. Medullary thyroid cancer (MTC)
MTC is a distinct subtype of thyroid carcinoma originating
from the parafollicular C cells o, accounting for 3–5% of all
thyroid malignancies [143]. The development of MTC is pri-
marily driven by specific genetic mutations that lead to the
activation of critical signaling pathways and the dysregulation
of cellular processes [143]. Approximately 75–80% of MTCs are
Figure 2. Physiopathology of Merkel cell carcinoma. UV: ultraviolet radiation. DNA: desoxy-ribonucleic acid.
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 7
sporadic neoplasms, while a smaller proportion are hereditary
and associated with multiple endocrine neoplasia type 2
(MEN2) syndromes and familial medullary thyroid cancer
(FMTC) [144]. In both sporadic and hereditary MTC, activating
mutations in the RET proto-oncogene located on chromosome
10q11.21 play a central role in MTC pathogenesis [145,146].
The most common RET mutation in hereditary MTC is
a germline mutation in codon 634, while in sporadic MTC
cases various regions of the gene are involved [145,146].
These lead to activation of downstream signaling cascades,
primarily the mitogen-activated protein kinase (MAPK) and
phosphoinositide 3-kinase (PI3K)/Akt pathways, pivotal in pro-
liferation, survival, differentiation, and angiogenesis [147,148].
Moreover, it could have a role in increasing cell mobility.
Mutated RET can disrupt cell-cell and cell-matrix interactions
by altering the expression of adhesion molecules, such as
E-cadherin, and by inducing the secretion of matrix metallo-
proteinases (MMPs) [149]. These changes promote the invasive
behavior of MTC cells, facilitating their dissemination to regio-
nal lymph nodes and distant organs. In addition to the genetic
mutations driving MTC development, there is evidence to
suggest the presence of pre-neoplastic lesions that precede
the formation of fully malignant tumors [143]. These lesions,
known as C-cell hyperplasia, represent a precursor stage char-
acterized by the expansion and altered function of C cells in
the thyroid gland [143]. Genetic and molecular alterations
found in pre-neoplastic lesions, specifically C-cell hyperplasia,
provide insights into the molecular events leading to MTC.
Several genetic and molecular changes have been identified in
pre-neoplastic lesions, shedding light on the early stages of
MTC development [147]. For instance, mutations in the TP53
tumor suppressor gene have been observed in both pre-
neoplastic lesions and MTC, suggesting its involvement in
disease progression [147]. Activation of the RAS oncogene
pathway has also been implicated in the transformation of
pre-neoplastic lesions to MTC, with mutations in HRAS (17%)
and KRAS (7%) genes detected in these lesions [150]. Not only
genetic mutation could lead to the ‘second-hit,’ crucial to
carcinoma development but also epigenetic alteration.
Indeed, even if telomerase reverse transcriptase (TERT) promo-
ter is not mutated, his methylation has been reported in MTC,
increasing TERT expression and ending in telomerase activa-
tion [151]. This data has been associated with decreasing
disease-free and overall survival in MTC [151]. Moreover,
some microRNA (miRNA) could have a role in carcinogenesis.
In particular, miRNA-183 and 375 are overexpressed in spora-
dic versus hereditary MTC and were associated in lateral
lymph node metastases, residual disease, and distant metas-
tases, influencing mortality [152]. Interestingly, even without
clinical difference, global DNA methylation level has been
found to be higher in sporadic MTC rather than in hereditary
MTC [153].
3.5. Bone metastases
Bone metastases in NENs represent an advanced stage of
the disease and are associated with significant morbidity,
thus the understanding of their pathogenetic mechanism is
of great relevance [154]. The development of bone
metastases involves a complex interplay of cellular and
molecular mechanisms, which contribute to the infiltration,
growth, and survival of tumor cells within the bone micro-
environment [155]. The process of bone metastasis begins
with the invasion of tumor cells into the bloodstream or
lymphatic system. Once in circulation, tumor cells interact
with bone marrow-derived cells, such as osteoclasts, osteo-
blasts, and immune cells, through adhesion molecules and
chemotactic factors. This interaction facilitates the homing
of tumor cells to the bone microenvironment [155]. One of
the key factors contributing to bone metastases in NENs is
the secretion of various bioactive substances by tumor cells,
including growth factors, cytokines, and chemokines contri-
buting to the creation of a specific niche [156]. The creation
of this niche is crucial for the development of bone metas-
tasis, as neoplastic cells do not possess the characteristics
for the degradation of the extracellular matrix (ECM) and
require the activation of osteoclasts [155]. Cytokines pro-
duced by the tumor receptor led to increased expression of
nuclear factor-kappa B ligand (RANKL) and decreased osteo-
protegerin (OPG) values [10]. RANKL is a protein expressed
by osteoblasts and other stromal cells in the bone micro-
environment. It plays a central role in regulating bone
remodeling and osteoclast differentiation [157]. The
increased action of osteoclasts due to overexpression of
RANKL leads to increased resorption of bone ECM resulting
in the release of a number of growth factors that stimulate
homing, tumor cell proliferation, and bone destruction,
creating a vicious circle [10]. In addition, the ECM environ-
ment is characterized by hypoxia, acidic pH, and high levels
of extracellular calcium, all of which create a favorable
environment for tumor growth [10]. However, independent
of RANKL activation, tumor cytokines including cathepsin K,
IL 8, and VEGF alone are capable of stimulating bone
resorption through osteoclasts [158,159]. Studies have
shown the importance in the development of bone metas-
tases in NENs of the overexpression of CXCL12, together
with its C-X-C motif receptor 4 (CXCR4), which causes cell
hyperproliferation and angiogenesis [160]. Studies in other
neoplasms suggest a possible role of microRNAs as well,
however no evidence is currently available in NENs [10].
3.6. Conclusions
The heterogeneity of NENs biology and clinical course mir-
rors the heterogeneity in their pathogenetic mechanisms.
The identification of preneoplastic lesions as well as the
underlying mechanism leading to tumor onset may allow
to identify effective prevention strategies. Similar genetic
alterations seem to play a pivotal role both in sporadic
and familial neoplasms. Data regarding pathogenetic
mechanisms in NENs are currently mainly available in GEP
NENs, maybe due to their relatively higher frequency.
Enteroendocrine cells have been considered as a source of
stem cells in SI-NETs, while hypergastrinemia and chronic
use of PPI have been proposed as possible pathogenetic
mechanisms in G-NETs. The role of inflammation remains
debated mainly in colorectal NETs, while in lung carcinoids
nicotine and hypoxia could be crucial in tumor
8R. MODICA ET AL.
development. Available data remain heterogeneous and
better knowledge is still an unmet need in this field, though
essential to improve cancer prevention and treatment
strategies.
3.7. Expert opinion
Improved understanding of pathological mechanisms in NENs
remains a crucial issue, considering the steadily increasing
incidence and prevalence of these neoplasms. Furthermore,
the identification of the best treatment at the right time for
any individual patient is a compelling issue in NENs and
a deep knowledge of pathogenetic mechanisms will be essen-
tial to improve treatment strategies, as well as to identify new
therapeutic targets [4,161–163].
Genetic alterations both in familial and sporadic NENs, as
well as epigenetic modifications play a pivotal role in NEN
development [15]. Familial NENs, as pNENs in MEN1 genetic
syndrome, could represent a model to study early tumor
development, but these observations seem to be different in
the sporadic counterpart.
Other evidence, including the possible neoplastic suscept-
ibility suggested by a not negligible prevalence of second
primary malignancies in patients with NENs could find an
explanation with the identification of pathogenetic mechan-
isms [164]. Importantly, NENs and NECs may recognize dif-
ferent pathways of development rather than just
a progression from NEN to NEC, due to the different biolo-
gical characteristics reflecting different prognosis. Similarly,
the development of bone metastases could show peculiar
pathogenetic mechanisms, which differ from primary devel-
opment and consequently different treatment could be
required.
The role of inflammation in NENs is highly debated, despite
inflammation processes known to be involved in cancer onset
and progression. The tumor microenvironment including
growth factors, cytokines, and chemokines and other factors
including endocrine disrupting chemicals, as well as genetic
and epigenetic modification are known to create an interplay
with inflammation [12,15,164,165]. Environmental factors, gen-
der differences, metabolic syndrome and nutrition, lipid
homeostasis, low vitamin D levels, and microbiota have been
analyzed as risk factors in NENs, but their role in pathogenetic
mechanisms could not be negligible. One of the major chal-
lenges ahead is to integrate the available data because the
slight difference between risk factors and pathogenetic factors
may be due to difficulty in indentifying the underlying
mechanisms. The higher prevalence of DIPNECH in the female
sex, as well as other gender differences, should draw the
attention of clinicians to better evaluate the role of hormonal
status in NENs.
Substantial data have been currently obtained in MCC,
where the role of polyomavirus and UV ray exposure is clearly
involved in the pathogenetic mechanism. Nevertheless, MCC is
completely different from the majority of NENs in its biological
features, being extremely aggressive with high metastatic
potential, and treatment options, including immunotherapy
are not included in the therapeutic algorithm of other NENs.
Consequently, the effort to identify pathogenic mechanisms in
NENs may be even more difficult than in other cancer types.
The complex interplay among patient characteristics, environ-
ment, and tumor biology does not currently allow to depict
a reliable model of pathogenesis in NENs. This significant
clinical challenge warrants further investigation, mainly to
allow early diagnosis but also for the identification of potential
therapeutic targets and more effective and tailored treatment
options, leading to better survival outcomes and improved
quality of life [166].
Funding
This paper was not funded.
Declaration of interest
The authors have no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
Author contribution statement
Conception/Design: RoMo, AC.
Collection and/or assembly of data: RoMo, AL, RoMi, GC, EB.
Data analysis and interpretation: Ro Mo, AL, RoMi, GC, EB, AC.
Manuscript writing: Ro Mo, AL, RoMi, GC, EB.
Final approval of manuscript: RoMo, AL, RoMi, GC, EB, AC.
ORCID
Roberta Modica http://orcid.org/0000-0002-3768-5803
Alessia Liccardi http://orcid.org/0000-0003-1784-5491
Roberto Minotta http://orcid.org/0000-0002-8823-0702
Giuseppe Cannavale http://orcid.org/0000-0002-8524-1131
Elio Benevento http://orcid.org/0000-0002-4489-3651
Annamaria Colao http://orcid.org/0000-0003-4049-2559
References
Papers of special note have been highlighted as either of interest (•)
or of considerable interest (••) to readers.
1. Muscogiuri G, Altieri B, Albertelli M, et al. Epidemiology of pancrea-
tic neuroendocrine neoplasms: a gender perspective. Endocrine.
2020;69(2):441–450.
2. Ruggeri RM, Benevento E, De Cicco F, et al. Neuroendocrine neo-
plasms in the context of inherited tumor syndromes: a reappraisal
focused on targeted therapies. J Endocrinol Invest. 2023;46
(2):213–234.
3. Dasari A, Shen C, Halperin D, et al. Trends in the incidence, pre-
valence, and survival outcomes in patients with neuroendocrine
tumors in the United States. JAMA Oncol. 2017;3(10):1335–1342.
4. Modica R, Liccardi A, Minotta R, et al. Therapeutic strategies for
patients with neuroendocrine neoplasms: current perspectives.
Expert Rev Endocrinol Metab. 2022;17(5):389–403.
5. Faggiano A, Di Maio S, Mocerino C, et al. Therapeutic sequences in
patients with grade 1−2 neuroendocrine tumors (NET): an
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 9
observational multicenter study from the ELIOS group. Endocrine.
2019;66(2):417–424.
6. Faggiano A, Lo Calzo F, Pizza G, et al. The safety of available
treatments options for neuroendocrine tumors. Expert Opin Drug
Saf. 2017;16(10):1149–1161.
7. Rindi G, Mete O, Uccella S, et al. Overview of the 2022 WHO
classification of neuroendocrine neoplasms. Endocr Pathol.
2022;33(1):115–154. doi: 10.1007/s12022-022-09708-2
•• It is a very exhaustive and clear overview on new NENs classi-
fication necessary to understand and to manage these
neoplasms.
8. Zatelli MC, Guadagno E, Messina E, et al. Open issues on G3
neuroendocrine neoplasms: back to the future. Endocr Relat
Cancer. 2018;25(6):R375–R384.
9. Riihimäki M, Hemminki A, Sundquist K, et al. The epidemiology of
metastases in neuroendocrine tumors. Int J Cancer. 2016;139
(12):2679–2686.
10. Altieri B, Di Dato C, Martini C, et al. Bone Metastases in
Neuroendocrine Neoplasms: From Pathogenesis to Clinical
Management. Cancers (Basel). 2019;11(9):11.
11. Kečkéš Š, Palaj J, Waczulíková I, et al. Pretreatment levels of chro-
mogranin a and neuron-specific enolase in patients with gastro-
enteropancreatic neuroendocrine neoplasia. In Vivo. 2021;35
(5):2863–2868.
12. Modica R, Benevento E, Colao A. Endocrine-disrupting chemicals
(EDCs) and cancer: new perspectives on an old relationship.
J Endocrinol Invest. 2023;46(4):667–677. doi: 10.1007/s40618-022-
01983-4
13. Modica R, La Salvia A, Liccardi A, et al. Lipid metabolism and
homeostasis in patients with neuroendocrine neoplasms: from
risk factor to potential therapeutic target. Metabolites. 2022;12
(11):12.
14. Feola T, Puliani G, Sesti F, et al. Risk factors for gastroenteropan-
creatic neuroendocrine neoplasms (GEP-NENs): a three-centric
case–control study. J Endocrinol Invest. 2022;45(4):849–857.
15. Colao A, de Nigris F, Modica R, et al. Clinical epigenetics of neu-
roendocrine tumors: the road ahead. Front Endocrinol.
2020;11:604341. doi: 10.3389/fendo.2020.604341
16. Yao JC, Hassan M, Phan A, et al. One hundred years after “carci-
noid”: epidemiology of and prognostic factors for neuroendocrine
tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26
(18):3063–3072.
17. von Arx C, Capozzi M, López-Jiménez E, et al. Updates on the role
of molecular alterations and NOTCH signalling in the development
of neuroendocrine neoplasms. J Clin Med. 2019;8(9):1277.
18. Barrea L, Muscogiuri G, Modica R, et al. Cardio-metabolic indices
and metabolic syndrome as predictors of clinical severity of gastro-
enteropancreatic neuroendocrine tumors. Front Endocrinol.
2021;12:649496. doi: 10.3389/fendo.2021.649496
19. Altieri B, Di Dato C, Modica R, et al. Bone Metabolism and Vitamin
D Implication in Gastroenteropancreatic Neuroendocrine Tumors.
Nutrients. 2020;12(4):12.
20. Vanoli A, La Rosa S, Luinetti O, et al. Histologic changes in type
a chronic atrophic gastritis indicating increased risk of neuroendo-
crine tumor development: the predictive role of dysplastic and
severely hyperplastic enterochromaffin-like cell lesions. Hum
Pathol. 2013;44(9):1827–1837. doi: 10.1016/j.humpath.2013.02.005
Provides support for the transition of gastric preneoplastic
lesions.
21. Oberg K. Genetics and molecular pathology of neuroendocrine
gastrointestinal and pancreatic tumors (gastroenteropancreatic
neuroendocrine tumors). Curr Opin Endocrinol Diabetes Obes.
2009;16(1):72–78. doi: 10.1097/MED.0b013e328320d845
22. Das S, Dasari A. Epidemiology, incidence, and prevalence of
Neuroendocrine Neoplasms: are there global differences? Curr
Oncol Rep. 2021;23:43. doi: 10.1007/s11912-021-01029-7
23. Rindi G, Inzani F, Solcia E. Pathology of gastrointestinal disorders.
Endocrinol Metab Clin North Am. 2010;39(4):713–727. doi: 10.1016/
j.ecl.2010.08.009
24. Sei Y, Feng J, Samsel L, et al. Mature enteroendocrine cells con-
tribute to basal and pathological stem cell dynamics in the small
intestine. Am J Physiol Gastrointest Liver Physiol. 2018;315(4):
G495–G510. doi: 10.1152/ajpgi.00036.2018
Provides support for the hypothesis that stem cells may ded-
ifferentiate leading to SI-NENs.
25. Asa SL, La Rosa S, Basturk O, et al. Molecular pathology of
well-differentiated gastro-entero-pancreatic neuroendocrine
tumors. Endocr Pathol. 2021;32(1):169–191. doi: 10.1007/s12022-
021-09662-5
This paper focuses on the molecular pathways that can parti-
cipate in GEP-NENs development according to different
localizations.
26. Banck MS, Kanwar R, Kulkarni AA, et al. The genomic landscape of
small intestine neuroendocrine tumors. J Clin Invest. 2013;123
(6):2502–2508.
27. Cunningham JL, Díaz de Ståhl T, Sjöblom T, et al. Common patho-
genetic mechanism involving human chromosome 18 in familial
and sporadic ileal carcinoid tumors. Genes Chromosomes Cancer.
2011;50(2):82–94.
28. Choi I-S, Estecio MRH, Nagano Y, et al. Hypomethylation of LINE-1
and Alu in well-differentiated neuroendocrine tumors (pancreatic
endocrine tumors and carcinoid tumors). Mod Pathol. 2007;20
(7):802–810.
29. Stricker I, Tzivras D, Nambiar S, et al. Site- and grade-specific
diversity of LINE1 methylation pattern in gastroenteropancreatic
neuroendocrine tumours. Anticancer Res. 2012;32(9):3699–3706.
30. How-Kit A, Dejeux E, Dousset B, et al. DNA methylation profiles
distinguish different subtypes of gastroenteropancreatic neuroen-
docrine tumors. Epigenomics. 2015;7(8):1245–1258.
31. Edfeldt K, Ahmad T, Åkerström G, et al. TCEB3C a putative tumor
suppressor gene of small intestinal neuroendocrine tumors. Endocr
Relat Cancer. 2014;21(2):275–284.
32. Zhang H-Y, Rumilla KM, Jin L, et al. Association of DNA methylation
and epigenetic inactivation of RASSF1A and beta-catenin with
metastasis in small bowel carcinoid tumors. Endocrine. 2006;30
(3):299–306.
33. Ruebel K, Leontovich AA, Stilling GA, et al. MicroRNA expression in
ileal carcinoid tumors: downregulation of microRNA-133a with
tumor progression. Mod Pathol. 2010;23(3):367–375.
34. Li S-C, Essaghir A, Martijn C, et al. Global microRNA profiling of
well-differentiated small intestinal neuroendocrine tumors. Mod
Pathol. 2013;26(5):685–696.
35. Karpathakis A, Dibra H, Pipinikas C, et al. Prognostic impact of
novel molecular subtypes of small intestinal neuroendocrine
tumor. Clin Cancer Res. 2016;22(1):250–258.
36. Cross AJ, Leitzmann MF, Subar AF, et al. A prospective study of
meat and fat intake in relation to small intestinal cancer. Cancer
Res. 2008;68(22):9274–9279.
37. Klöppel G, Anlauf M, Perren A, et al. Hyperplasia to neoplasia
sequence of duodenal and pancreatic neuroendocrine diseases
and pseudohyperplasia of the PP-cells in the pancreas. Endocr
Pathol. 2014;25(2):181–185.
38. 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.
39. 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.
40. Vortmeyer AO, Huang S, Lubensky I, et al. Non-islet origin of
pancreatic islet cell tumors. J Clin Endocrinol Metab. 2004;89
(4):1934–1938. doi: 10.1210/jc.2003-031575
This paper focuses on the interesting non-islet oigin of pNENs.
41. Perren A, Anlauf M, Henopp T, et al. Multiple endocrine neoplasia
type 1 (MEN1): loss of one MEN1 allele in tumors and monohor-
monal endocrine cell clusters but not in islet hyperplasia of the
pancreas. J Clin Endocrinol Metab. 2007;92(3):1118–1128.
10 R. MODICA ET AL.
42. Pea A, Hruban RH, Wood LD. Genetics of pancreatic neuroendo-
crine tumors: implications for the clinic. Expert Rev Gastroenterol
Hepatol. 2015;9(11):1407–1419. doi: 10.1586/17474124.2015.
1092383
43. Ohmoto A, Rokutan H, Yachida S. Pancreatic neuroendocrine neo-
plasms: basic biology, Current treatment strategies and prospects
for the future. Int J Mol Sci. 2017;18(1):18. doi: 10.3390/
ijms18010143
44. de Wilde RF, Heaphy CM, Maitra A, et al. Loss of ATRX or DAXX
expression and concomitant acquisition of the alternative length-
ening of telomeres phenotype are late events in a small subset of
MEN-1 syndrome pancreatic neuroendocrine tumors. Mod Pathol.
2012;25(7):1033–1039.
45. Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors:
expression profiling evidences a role for AKT-mTOR pathway. J Clin
Oncol. 2010;28(2):245–255.
46. La Rosa S, Vanoli A. Gastric neuroendocrine neoplasms and related
precursor lesions. J Clin Pathol. 2014;67(11):938–948. doi: 10.1136/
jclinpath-2014-202515
47. La Rosa S, Inzani F, Vanoli A, et al. Histologic characterization and
improved prognostic evaluation of 209 gastric neuroendocrine
neoplasms. Hum Pathol. 2011;42(10):1373–1384.
48. La Rosa S, Solcia E. New insights into the classification of gastric
neuroendocrine tumours, expanding the spectrum of ECL-cell
tumours related to hypergastrinaemia. Histopathology. 2020;77
(6):862–864. doi: 10.1111/his.14226
49. Solcia E, Bordi C, Creutzfeldt W, et al. Histopathological classifica-
tion of nonantral gastric endocrine growths in man. Digestion.
1988;41(4):185–200.
50. Annibale B, Azzoni C, Corleto VD, et al. Atrophic body gastritis
patients with enterochromaffin-like cell dysplasia are at increased
risk for the development of type I gastric carcinoid. Eur
J Gastroenterol Hepatol. 2001;13(12):1449–1456.
51. Toh B-H, Chan J, Kyaw T, et al. Cutting edge issues in autoimmune
gastritis. Clin Rev Allergy Immunol. 2012;42(3):269–278.
52. Neumann WL, Coss E, Rugge M, et al. Autoimmune atrophic gas-
tritis—pathogenesis, pathology and management. Nat Rev
Gastroenterol Hepatol. 2013;10(9):529–541.
53. Whittingham S, Mackay IR. Autoimmune gastritis: historical ante-
cedents, outstanding discoveries, and unresolved problems. Int Rev
Immunol. 2005;24(1–2):1–29. doi: 10.1080/08830180590884413
54. Lahner E, Centanni M, Agnello G, et al. Occurrence and risk factors
for autoimmune thyroid disease in patients with atrophic body
gastritis. Am J Med. 2008;121(2):136–141.
55. Rugge M, Fassan M, Pizzi M, et al. Autoimmune gastritis: histology
phenotype and OLGA staging. Aliment Pharmacol Ther. 2012;35
(12):1460–1466.
56. Vannella L, Sbrozzi-Vanni A, Lahner E, et al. Development of type
I gastric carcinoid in patients with chronic atrophic gastritis.
Aliment Pharmacol Ther. 2011;33(12):1361–1369.
57. Calvete O, Reyes J, Zuñiga S, et al. Exome sequencing identifies
ATP4A gene as responsible of an atypical familial type I gastric
neuroendocrine tumour. Hum Mol Genet. 2015;24(10):2914–2922.
58. Solcia E, Capella C, Fiocca R, et al. Gastric argyrophil carcinoidosis in
patients with Zollinger-Ellison syndrome due to type 1 multiple
endocrine neoplasia. A newly recognized association. Am J Surg
Pathol. 1990;14(6):503–513.
59. Rindi G, Bordi C, Rappel S, et al. Gastric carcinoids and neuroendo-
crine carcinomas: pathogenesis, pathology, and behavior. World
J Surg. 1996;20(2):168–172.
60. Abraham SC, Carney JA, Ooi A, et al. Achlorhydria, parietal cell
hyperplasia, and multiple gastric carcinoids: a new disorder. Am
J Surg Pathol. 2005;29(7):969–975.
61. Berna MJ, Annibale B, Marignani M, et al. A prospective study of
gastric carcinoids and enterochromaffin-like cell changes in multi-
ple endocrine neoplasia type 1 and Zollinger-Ellison syndrome:
identification of risk factors. J Clin Endocrinol Metab. 2008;93
(5):1582–1591.
62. Trinh V-H, Shi C, Ma C. Gastric neuroendocrine tumours from
long-term proton pump inhibitor users are indolent tumours with
good prognosis. Histopathology. 2020;77(6):865–876. doi: 10.1111/
his.14220
63. Song H, Zhu J, Lu D. Long-term proton pump inhibitor (PPI) use
and the development of gastric pre-malignant lesions. Cochrane
Database Syst Rev. 2014; CD010623. doi: 10.1002/14651858.
CD010623.pub2
•• This meta analysis provides consistent data about PPI use and
pre-malignant lesions.
64. Jianu CS, Fossmark R, Viset T, et al. Gastric carcinoids after
long-term use of a proton pump inhibitor. Aliment Pharmacol
Ther. 2012;36(7):644–649.
65. Cavalcoli F, Zilli A, Conte D, et al. Gastric neuroendocrine neo-
plasms and proton pump inhibitors: fact or coincidence? Scand
J Gastroenterol. 2015;50(11):1397–1403.
66. Haga Y, Nakatsura T, Shibata Y, et al. Human gastric carcinoid
detected during long-term antiulcer therapy of H2 receptor
antagonist and proton pump inhibitor. Dig Dis Sci. 1998;43
(2):253–257.
67. Lahner E, Pilozzi E, Esposito G, et al. Gastric carcinoid in the
absence of atrophic body gastritis and with low Ki67 index:
a clinical challenge. Scand J Gastroenterol. 2014;49(4):506–510.
68. Nandy N, Hanson JA, Strickland RG, et al. Solitary gastric carcinoid
tumor associated with long-term use of omeprazole: a case report
and review of the literature. Dig Dis Sci. 2016;61(3):708–712.
69. Milione M, Parente P, Grillo F, et al. Neuroendocrine neoplasms of
the duodenum, ampullary region, jejunum and ileum. Pathologica.
2021;113(1):12–18.
70. Vinik AI, McLeod MK, Fig LM, et al. Clinical features, diagnosis, and
localization of carcinoid tumors and their management.
Gastroenterol Clin North Am. 1989;18(4):865–896.
71. Burke AP, Sobin LH, Federspiel BH, et al. Carcinoid tumors of the
duodenum. A clinicopathologic study of 99 cases. Arch Pathol Lab
Med. 1990;114(7):700–704.
72. Anlauf M, Enosawa T, Henopp T, et al. Primary lymph node gas-
trinoma or occult duodenal microgastrinoma with lymph node
metastases in a MEN1 patient: the need for a systematic search
for the primary tumor. Am J Surg Pathol. 2008;32(7):1101–1105.
73. Mete O, Asa SL. Precursor lesions of endocrine system neoplasms.
Pathology. 2013;45(3):316–330. doi: 10.1097/PAT.
0b013e32835f45c5
74. Anlauf M, Perren A, Klöppel G. Endocrine precursor lesions and
microadenomas of the duodenum and pancreas with and without
MEN1: criteria, molecular concepts and clinical significance.
Pathobiology. 2007;74(5):279–284. doi: 10.1159/000105810
75. Mpilla G, Aboukameel A, Muqbil I, et al. PAK4-NAMPT dual inhibi-
tion as a novel strategy for therapy resistant pancreatic neuroen-
docrine tumors. Cancers (Basel). 2019;11(12):11.
76. Holle GE, Spann W, Eisenmenger W, et al. Diffuse
somatostatin-immunoreactive D-cell hyperplasia in the stomach
and duodenum. Gastroenterology. 1986;91(3):733–739.
77. Mastracci L, Rindi G, Grillo F, et al. Neuroendocrine neoplasms of
the esophagus and stomach. Pathologica. 2021;113(1):5–11.
78. Giannetta E, Guarnotta V, Rota F, et al. A rare rarity: neuroendocrine
tumor of the esophagus. Crit Rev Oncol Hematol. 2019;137:92–107.
doi: 10.1016/j.critrevonc.2019.02.012
79. Hoang MP, Hobbs CM, Sobin LH, et al. Carcinoid tumor of the
esophagus: a clinicopathologic study of four cases. Am J Surg
Pathol. 2002;26(4):517–522.
80. Cai W, Ge W, Yuan Y, et al. A 10-year population-based study of the
differences between NECs and carcinomas of the esophagus in
terms of clinicopathology and survival. J Cancer. 2019;10
(6):1520–1527.
81. Konishi T, Watanabe T, Kishimoto J, et al. Prognosis and risk factors
of metastasis in colorectal carcinoids: results of a nationwide reg-
istry over 15 years. Gut. 2007;56(6):863–868.
82. Volante M, Grillo F, Massa F, et al. Neuroendocrine neoplasms of
the appendix, colon and rectum. Pathologica. 2021;113(1):19–27.
83. Yu J, Refsum E, Perrin V, et al. Inflammatory bowel disease and risk
of adenocarcinoma and neuroendocrine tumors in the small bowel.
Ann Oncol. 2022;33(6):649–656.
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 11
84. Derikx LAAP, Vierdag W-M, Kievit W, et al. Is the prevalence of
colonic neuroendocrine tumors increased in patients with inflam-
matory bowel disease? Int J Cancer. 2016;139(3):535–542.
85. Moyana TN, Satkunam N. A comparative immunohistochemical
study of jejunoileal and appendiceal carcinoids. Implications for
histogenesis and pathogenesis. Cancer. 1992;70(5):1081–1088. doi:
10.1002/1097-0142(19920901)70:5<1081:AID-CNCR2820700512>3.
0.CO;2-9
86. Nascimbeni R, Villanacci V, Di Fabio F, et al. Solitary microcarcinoid
of the rectal stump in ulcerative colitis. Neuroendocrinology.
2005;81(6):400–404.
87. Lyss AP. Appendiceal malignancies. Semin Oncol. 1988;15
(2):129–137.
88. Pape U-F, Niederle B, Costa F, et al. ENETS consensus guidelines for
neuroendocrine neoplasms of the appendix (excluding goblet cell
carcinomas). Neuroendocrinology. 2016;103(2):144–152.
89. Moertel CG, Dockerty MB, Judd ES. Carcinoid tumors of the vermi-
form appendix. Cancer. 1968;21(2):270–278. doi: 10.1002/1097-
0142(196802)21:2<270:AID-CNCR2820210217>3.0.CO;2-9
90. Auböck L, Höfler H. Extraepithelial intraneural endocrine cells as
starting-points for gastrointestinal carcinoids. Virchows Arch
A Pathol Anat Histopathol. Virchows Arch A Pathological
Anatomy Histopathol. 1983;401(1):17–33. doi: 10.1007/BF00644786
91. Papadaki L, Rode J, Dhillon AP, et al. Fine structure of
a neuroendocrine complex in the mucosa of the appendix.
Gastroenterology. 1983;84(3):490–497.
92. Sesia SB, Mayr J, Bruder E, et al. Neurogenic appendicopathy:
clinical, macroscopic, and histopathological presentation in pedia-
tric patients. Eur J Pediatr Surg. 2013;23(3):238–242.
93. Karkouche R, Bachet J-B, Sandrini J, et al. Colorectal neuroendo-
crine carcinomas and adenocarcinomas share oncogenic pathways.
A clinico-pathologic study of 12 cases. Eur J Gastroenterol Hepatol.
2012;24(12):1430–1437.
94. Sorbye H, Strosberg J, Baudin E, et al. Gastroenteropancreatic
high-grade neuroendocrine carcinoma. Cancer. 2014;120
(18):2814–2823.
95. Ikegame K, Hatakeyama K, Terashima M, et al. Molecular profiling
of gastric neuroendocrine carcinomas. Eur J Surg Oncol. 2023;49
(9):106987.
96. Venizelos A, Elvebakken H, Perren A, et al. The molecular charac-
teristics of high-grade gastroenteropancreatic neuroendocrine
neoplasms. Endocr Relat Cancer. 2021;29(1):1–14.
97. Idrees K, Padmanabhan C, Liu E, et al. Frequent BRAF mutations
suggest a novel oncogenic driver in colonic neuroendocrine
carcinoma. J Surg Oncol. 2018;117(2):284–289.
98. Yachida S, Vakiani E, White CM, et al. Small cell and large cell
neuroendocrine carcinomas of the pancreas are genetically similar
and distinct from well-differentiated pancreatic neuroendocrine
tumors. Am J Surg Pathol. 2012;36(2):173–184.
99. Scarpa A, Chang DK, Nones K, et al. Whole-genome landscape of
pancreatic neuroendocrine tumours. Nature. 2017;543(7643):65–71.
100. Ku SY, Rosario S, Wang Y, et al. Rb1 and Trp53 cooperate to
suppress prostate cancer lineage plasticity, metastasis, and antian-
drogen resistance. Science. 2017;355(6320):78–83.
101. Grassia R, Bodini P, Dizioli P, et al. Neuroendocrine carcinomas
arising in ulcerative colitis: coincidences or possible correlations?
World J Gastroenterol. 2009;15(33):4193–4195.
102. Bolzacchini E, Chini C, Cortelezzi CC, et al. Poorly differentiated
neuroendocrine carcinoma of the sigmoid tract in long-standing
ulcerative colitis: report of a case and review of the literature.
Int J Surg Pathol. 2018;26(5):479–483.
103. Sigel JE, Goldblum JR. Neuroendocrine neoplasms arising in inflam-
matory bowel disease: a report of 14 cases. Mod Pathol. 1998;11
(6):537–542.
104. Travis WD, Brambilla E, Burke AP, et al. Introduction to the 2015
World Health Organization classification of tumors of the lung,
Pleura, thymus, and heart. J Thorac Oncol. 2015;10(9):1240–1242.
105. McGuire AL, Maziak DE, Sekhon HS. Diffuse intrapulmonary neu-
roendocrine cell hyperplasia. Can Respir J. 2013;20(6):406–408. doi:
10.1155/2013/190409
This paper focuses on DIPNECH as an important entity to be
monitored.
106. Rossi G, Cavazza A, Spagnolo P, et al. Diffuse idiopathic pulmonary
neuroendocrine cell hyperplasia syndrome. Eur Respir J. 2016;47
(6):1829–1841.
107. Rea F, Rizzardi G, Zuin A, et al. Outcome and surgical strategy in
bronchial carcinoid tumors: single institution experience with 252
patients. Eur J Cardiothorac Surg. 2007;31(2):186–191.
108. Travis WD, Rush W, Flieder DB, et al. Survival analysis of 200
pulmonary neuroendocrine tumors with clarification of criteria for
atypical carcinoid and its separation from typical carcinoid. Am
J Surg Pathol. 1998;22(8):934–944.
109. Caplin ME, Pavel M, Ćwikła JB, et al. Anti-tumour effects of lanreo-
tide for pancreatic and intestinal neuroendocrine tumours: the
CLARINET open-label extension study. Endocr Relat Cancer.
2016;23(3):191–199.
110. Dingemans A-M, Früh M, Ardizzoni A, et al. Small-cell lung cancer:
ESMO Clinical Practice Guidelines for diagnosis, treatment and
follow-up. Ann Oncol. 2021;32(7):839–853.
111. Onuki N, Wistuba IIR, Travis WD, et al. Genetic changes in the
spectrum of neuroendocrine lung tumors. Cancer. 1999;85
(3):600–607.
112. Alì G, Boldrini L, Capodanno A, et al. Expression of p-AKT and
p-mTOR in a large series of bronchopulmonary neuroendocrine
tumors. Exp Ther Med. 2011;2(5):787–792.
113. Noguchi M, Furukawa KT, Morimoto M. Pulmonary neuroendocrine
cells: physiology, tissue homeostasis and disease. Dis Model Mech.
2020;13(12):13. doi: 10.1242/dmm.046920
114. Fu XW, Spindel ER. Recruitment of GABA(A) receptors in chemor-
eceptor pulmonary neuroepithelial bodies by prenatal nicotine
exposure in monkey lung. Adv Exp Med Biol. 2009;648:439–445.
115. Fu XW, Nurse CA, Wong V, et al. Hypoxia-induced secretion of
serotonin from intact pulmonary neuroepithelial bodies in neona-
tal rabbit. J Physiol. 2002;539(2):503–510.
116. Hajiasgharzadeh K, Sadigh-Eteghad S, Mansoori B, et al. Alpha7
nicotinic acetylcholine receptors in lung inflammation and carcino-
genesis: Friends or foes? J Cell Physiol. 2019;234(9):14666–14679.
117. Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smoking and lung
cancer—relative risk estimates for the major histological types
from a pooled analysis of case–control studies. Int J Cancer.
2012;131(5):1210–1219.
118. Varghese AM, Zakowski MF, Yu HA, et al. Small-cell lung cancers in
patients who never smoked cigarettes. J Thorac Oncol. 2014;9
(6):892–896.
119. Offin M, Chan JM, Tenet M, et al. Concurrent RB1 and TP53 altera-
tions define a subset of EGFR-Mutant lung Cancers at risk for
Histologic transformation and inferior clinical outcomes. J Thorac
Oncol. 2019;14(10):1784–1793.
120. Xiong W-M, Xu Q-P, Li X, et al. The association between human
papillomavirus infection and lung cancer: a system review and
meta-analysis. Oncotarget. 2017;8(56):96419–96432.
121. Rekhtman N. Lung neuroendocrine neoplasms: recent progress and
persistent challenges. Mod Pathol. 2022;35:36–50. doi: 10.1038/
s41379-021-00943-2
122. Borczuk AC. Pulmonary Neuroendocrine Tumors. Surg Pathol Clin.
2020;13(1):35–55. doi: 10.1016/j.path.2019.10.002
123. Modlin IM, Kidd M, Filosso P-L, et al. Molecular strategies in the
management of bronchopulmonary and thymic neuroendocrine
neoplasms. J Thorac Dis. 2017;9(S15):S1458–S1473.
124. Bohnenberger H, Ströbel P. Recent advances and conceptual
changes in the classification of neuroendocrine tumors of the
thymus. Virchows Arch. 2021;478(1):129–135. doi: 10.1007/s00428-
021-03037-1
125. Ströbel P, Zettl A, Shilo K, et al. Tumor genetics and survival of
thymic neuroendocrine neoplasms: a multi-institutional clinico-
pathologic study. Genes Chromosomes Cancer. 2014;53
(9):738–749.
126. Dinter H, Bohnenberger H, Beck J, et al. Molecular classification of
neuroendocrine tumors of the thymus. J Thorac Oncol. 2019;14
(8):1472–1483.
12 R. MODICA ET AL.
127. Dang CV. MYC on the path to cancer. Cell. 2012;149(1):22–35. doi:
10.1016/j.cell.2012.03.003
128. Teh BT, Zedenius J, Kytölä S, et al. Thymic carcinoids in multiple
endocrine neoplasia type 1. Ann Surg. 1998;228(1):99–105.
129. Gibril F, Chen Y-J, Schrump DS, et al. Prospective study of thymic
carcinoids in patients with multiple endocrine neoplasia type 1.
J Clin Endocrinol Metab. 2003;88(3):1066–1081.
130. Cowen T, Thrasivoulou C, Shaw SA, et al. Transplanted sweat
glands from mature and aged donors determine cholinergic phe-
notype and altered density of host sympathetic nerves. J Auton
Nerv Syst. 1996;58(3):153–162.
131. Wong LH, McGhie JD, Sim M, et al. ATRX interacts with H3.3 in
maintaining telomere structural integrity in pluripotent embryonic
stem cells. Genome Res. 2010;20(3):351–360.
132. Liu R, Wang W, Ye L, et al. p21-activated kinase 3 is overexpressed
in thymic neuroendocrine tumors (carcinoids) with ectopic ACTH
syndrome and participates in cell migration. Endocrine. 2010;38
(1):38–47.
133. Bi Y, Liu R, Ye L, et al. Gene expression profiles of thymic neuroen-
docrine tumors (carcinoids) with ectopic ACTH syndrome reveal
novel molecular mechanism. Endocr Relat Cancer. 2009;16
(4):1273–1282.
134. Artavanis-Tsakonas S, Matsuno K, Fortini ME. Notch signaling.
Science. 1995;268(5208):225–232. doi: 10.1126/science.7716513
135. Xavier-Junior JCC, Ocanha-Xavier JP, Xavier-Junior JCCO-X JP. WHO
(2018). Classification of skin tumors. Am J Dermatopathol. 2019;41
(9):699–700. doi: 10.1097/DAD.0000000000001446
136. Becker JC, Stang A, DeCaprio JA, et al. Merkel cell carcinoma. Nat
Rev Dis Primers. 2017;3(1):17077.
137. Schadendorf D, Lebbé C, Zur Hausen A, et al. Merkel cell carcinoma:
Epidemiology, prognosis, therapy and unmet medical needs. Eur
J Cancer. 2017;71:53–69. doi: 10.1016/j.ejca.2016.10.022
138. Park SY, Doolittle-Amieva C, Moshiri Y, et al. How we treat Merkel
cell carcinoma: within and beyond current guidelines. Future
Oncol. 2021;17(11):1363–1377.
139. DeCaprio JA. Molecular pathogenesis of Merkel cell carcinoma.
Annu Rev Pathol. 2021;16(1):69–91. doi: 10.1146/annurev-
pathmechdis-012419-032817
•• This paper gives a clear explanation regarding MCC
physiopathology.
140. Pietropaolo V, Prezioso C, Moens U. Merkel Cell Polyomavirus and
Merkel Cell Carcinoma. Cancers (Basel). 2020;12(7):12. doi: 10.3390/
cancers12071774
141. Wong SQ, Waldeck K, Vergara IA, et al. UV-Associated mutations
underlie the etiology of MCV-Negative Merkel cell carcinomas.
Cancer Res. 2015;75(24):5228–5234.
142. Cheng J, Park DE, Berrios C, et al. Merkel cell polyomavirus recruits
MYCL to the EP400 complex to promote oncogenesis. PLOS
Pathog. 2017;13(10):e1006668.
143. Wells SA, Asa SL, Dralle H, et al. Revised American thyroid associa-
tion guidelines for the management of medullary thyroid
carcinoma. Thyroid: Offic J Am Thyroid Association. 2015;25
(6):567–610.
144. Pavlidis E, Sapalidis K, Chatzinikolaou F, et al. Medullary thyroid
cancer: molecular factors, management and treatment. Rom
J Morphol Embryol. 2020;61(3):681–686.
145. Bongarzone I. RET genetic screening in patients with medullary
thyroid cancer: an update. Front Endocrinol (Lausanne).
2020;11:571205. & FL.
146. Frank-Raue K. Hereditary medullary thyroid carcinoma: current
state of knowledge and future perspectives. Eur J Endocrinol.
2021;185(3): R119–R132. BS, & RF.
147. Romei C, Ciampi R, Elisei R. A comprehensive overview of the role
of the RET proto-oncogene in thyroid carcinoma. Nat Rev
Endocrinol. 2016;12(4):192–202. doi: 10.1038/nrendo.2016.11
•• This paper focuses on the recognized RET role in MTC.
148. Kondo T. Aspects of the pathogenesis of medullary thyroid
carcinoma. Endocrinol Metab Clin North Am. 2018;47(3): 481–493.
& ES.
149. Machens A, Lorenz K, Sekulla C, et al. Molecular epidemiology of
multiple endocrine neoplasia 2: implications for RET screening in
the new millenium. Eur J Endocrinol. 2013;168(3):307–314.
150. Ciampi R, Romei C, Ramone T, et al. Genetic landscape of somatic
mutations in a large cohort of sporadic medullary thyroid carcino-
mas studied by next-generation targeted sequencing. iScience.
2019;20:324–336. doi: 10.1016/j.isci.2019.09.030
151. Wang N, Kjellin H, Sofiadis A, et al. Genetic and epigenetic back-
ground and protein expression profiles in relation to telomerase
activation in medullary thyroid carcinoma. Oncotarget. 2016;7
(16):21332–21346.
152. Abraham D, Jackson N, Gundara JS, et al. MicroRNA profiling of
sporadic and hereditary medullary thyroid cancer identifies predic-
tors of nodal metastasis, prognosis, and potential therapeutic
targets. Clin Cancer Res. 2011;17(14):4772–4781.
153. Ceolin L, Goularte APP, Ferreira CV, et al. Global DNA methylation
profile in medullary thyroid cancer patients. Exp Mol Pathol.
2018;105(1):110–114.
154. Coleman RE. Clinical features of metastatic bone disease and risk of
skeletal morbidity. Clin Cancer Res. 2006;12(20):6243s–6249s. doi:
10.1158/1078-0432.CCR-06-0931
155. Mundy GR. Mechanisms of bone metastasis. Cancer.
1997;80:1546–1556.
•• It is a very exhaustive paper on bone metastasis.
156. Kaplan RN, Rafii S, Lyden D. Preparing the “soil”: the premetastatic
niche. Cancer Res. 2006;66(23):11089–11093. doi: 10.1158/0008-
5472.CAN-06-2407
157. Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and
activation. Nature. 2003;423(6937):337–342. doi: 10.1038/
nature01658
158. Shupp AB, Kolb AD, Mukhopadhyay D, et al. Cancer metastases to
bone: concepts, mechanisms, and interactions with bone
osteoblasts. Cancers (Basel). 2018;10(6):10.
159. Bendre MS, Margulies AG, Walser B, et al. Tumor-derived interleu-
kin-8 stimulates osteolysis Independent of the receptor activator of
nuclear factor-κB ligand pathway. Cancer Res. 2005;65
(23):11001–11009.
160. Cives M, Quaresmini D, Rizzo FM, et al. Osteotropism of neuroen-
docrine tumors: role of the CXCL12/CXCR4 pathway in promoting
EMT in vitro. Oncotarget. 2017;8(14):22534–22549.
161. Pusceddu S, Prinzi N, Tafuto S, et al. Association of Upfront Peptide
receptor radionuclide therapy with progression-free survival
Among patients with enteropancreatic neuroendocrine tumors.
JAMA Netw Open. 2022;5(2):e220290.
162. Lania A, Ferraù F, Rubino M, et al. Neoadjuvant therapy for
Neuroendocrine Neoplasms: recent progresses and future
approaches. Front Endocrinol. 2021;12:651438. doi: 10.3389/fendo.
2021.651438
163. Albertelli M, Dotto A, Di Dato C, et al. PRRT: identikit of the perfect
patient. Rev Endocr Metab Disord. 2021;22(3):563–579.
164. Massironi S, Campana D, Pusceddu S, et al. Second primary neo-
plasms in patients with lung and gastroenteropancreatic neuroen-
docrine neoplasms: data from a retrospective multi-centric study.
Dig Liver Dis. 2021;53(3):367–374.
165. Ravindranathan D, Master VA, Bilen MA. Inflammatory markers in
Cancer immunotherapy. Biology. 2021;10(4):10. doi: 10.3390/
biology10040325
166. Scandurra C, Modica R, Maldonato NM, et al. Quality of life in
patients with neuroendocrine neoplasms: the role of severity, clin-
ical heterogeneity, and resilience. J Clin Endocrinol Metab.
2021;106(1):e316–e327.
EXPERT REVIEW OF ENDOCRINOLOGY & METABOLISM 13
... Secondarily, the co-presence of gastric, duodenal, and pancreatic neoplasia, in the majority of neuroendocrine tumors (NETs), represents a syndromic consequence via a common genetic background in multiple endocrine neoplasia (MEN) syndrome that comes with an increased burden of disease due to synchronous or asynchronous neoplasms requiring multidisciplinary management [11][12][13]. ...
Article
Full-text available
We aimed to provide an in-depth analysis with respect to three turning points in pancreas involvement in primary hyperparathyroidism (PHP): hypercalcemia-induced pancreatitis (HCa-P), MEN1 (multiple endocrine neoplasia)-related neuroendocrine tumors (NETs), and insulin resistance (IR). This was a comprehensive review conducted via a PubMed search between January 2020 and January 2024. HCa-P (n = 9 studies, N = 1375) involved as a starting point parathyroid NETs (n = 7) or pancreatitis (n = 2, N = 167). Case report-focused analysis (N = 27) showed five cases of pregnancy PHP-HCa-P and three reports of parathyroid carcinoma (female/male ratio of 2/1, ages of 34 in women, men of 56). MEN1-NET studies (n = 7) included MEN1-related insulinomas (n = 2) or MEN1-associated PHP (n = 2) or analyses of genetic profile (n = 3), for a total of 877 MEN1 subjects. In MEN1 insulinomas (N = 77), the rate of associated PHP was 78%. Recurrence after parathyroidectomy (N = 585 with PHP) was higher after less-than-subtotal versus subtotal parathyroidectomy (68% versus 45%, p < 0.001); re-do surgery was 26% depending on surgery for pancreatic NETs (found in 82% of PHP patients). MEN1 pathogenic variants in exon 10 represented an independent risk factor for PHP recurrence. A single pediatric study in MEN1 (N = 80) revealed the following: a PHP rate of 80% and pancreatic NET rate of 35% and 35 underlying germline MEN1 pathogenic variants (and 3/35 of them were newly detected). The co-occurrence of genetic anomalies included the following: CDC73 gene variant, glucokinase regulatory protein gene pathogenic variant (c.151C>T, p.Arg51*), and CAH-X syndrome. IR/metabolic feature-focused analysis identified (n = 10, N = 1010) a heterogeneous spectrum: approximately one-third of adults might have had prediabetes, almost half displayed some level of IR as reflected by HOMA-IR > 2.6, and serum calcium was positively correlated with HOMA-IR. Vitamin D deficiency was associated with a higher rate of metabolic syndrome (n = 1). Normocalcemic and mildly symptomatic hyperparathyroidism (n = 6, N = 193) was associated with a higher fasting glucose and some improvement after parathyroidectomy. This multilayer pancreas/parathyroid analysis highlighted a complex panel of connections from pathogenic factors, including biochemical, molecular, genetic, and metabolic factors, to a clinical multidisciplinary panel.
... The presence of chronic atrophic gastritis is pivotal in gastric NEN prognosis, whereas types 1 and 2 generally have a better prognosis than type 3 (which usually constitutes advanced disease at first presentation) [1,[18][19][20]. For example, Sheikh-Ahmad et al. [21] conducted a single-centric study on patients with type 1 gastric NENs who were followed up for 41 months (between 11 and 288 months). ...
Article
Full-text available
Neuroendocrine neoplasia (NEN) represents a sensational field of modern medicine; immense progress in emerging biochemical, molecular, endocrine, immunohistochemical, and serum tumour markers of disease, respectively, which are part of early diagnosis, genetic testing, and multidisciplinary approaches [...]
Chapter
Neuroendocrine neoplasms (NEN) represent a rare and heterogeneous group of malignancies, whose incidence is increasing trough the last decades, both because of better detection strategies and of a growing awareness. The aim of biochemical, radiological, and nuclear medicine improved techniques is to early identify and characterize these tumors, in order to offer new and tailored treatments. An updated overview of diagnosis, classification, and treatments in patients with NEN is provided, with the aim to summarize recent evidence in the field of NEN. Interesting novelties in pathological examination with new immunohistochemical panels, radiological and functional imaging techniques, as well as the latest published guidelines in NEN are discussed, with focus on therapeutic strategies according to primary. This concise but thorough examination of the current advancements in NEN aims to represent a useful tool for clinicians and researchers working in this field. NEN classification and characterization have changed trough years, aiming to obtain a precise tumor profile and the best therapeutic strategy. NEN diagnosis and treatment should be discussed by expert multidisciplinary group to improve management and survival rates.
Article
Full-text available
Lipid metabolism is known to be involved in tumorigenesis and disease progression in many common cancer types, including colon, lung, breast and prostate, through modifications of lipid synthesis, storage and catabolism. Furthermore, lipid alterations may arise as a consequence of cancer treatment and may have a role in treatment resistance. Neuroendocrine neoplasms (NENs) are a heterogeneous group of malignancies with increasing incidence, whose mechanisms of cancer initiation and progression are far from being fully understood. Alterations of lipid metabolism may be common across various cancer types, but data about NENs are scattered and heterogeneous. Herein, we provide an overview of the relevant literature on lipid metabolism and alterations in NENs. The available evidence both in basic and clinical research about lipid metabolism in NENs, including therapeutic effects on lipid homeostasis, are summarized. Additionally, the potential of targeting the lipid profile in NEN therapy is also discussed, and areas for further research are proposed.
Article
Full-text available
PurposeNeuroendocrine neoplasms can occur as part of inherited disorders, usually in the form of well-differentiated, slow-growing tumors (NET). The main predisposing syndromes include: multiple endocrine neoplasias type 1 (MEN1), associated with a large spectrum of gastroenteropancreatic and thoracic NETs, and type 4 (MEN4), associated with a wide tumour spectrum similar to that of MEN1; von Hippel-Lindau syndrome (VHL), tuberous sclerosis (TSC), and neurofibromatosis 1 (NF-1), associated with pancreatic NETs. In the present review, we propose a reappraisal of the genetic basis and clinical features of gastroenteropancreatic and thoracic NETs in the setting of inherited syndromes with a special focus on molecularly targeted therapies for these lesions. Methods Literature search was systematically performed through online databases, including MEDLINE (via PubMed), and Scopus using multiple keywords’ combinations up to June 2022.ResultsSomatostatin analogues (SSAs) remain the mainstay of systemic treatment for NETs, and radiolabelled SSAs can be used for peptide-receptor radionuclide therapy for somatostatin receptor (SSTR)-positive NETs. Apart of these SSTR-targeted therapies, other targeted agents have been approved for NETs: the mTOR inhibitor everolimus for lung, gastroenteropatic and unknown origin NET, and sunitinib, an antiangiogenic tyrosine kinase inhibitor, for pancreatic NET. Novel targeted therapies with other antiangiogenic agents and immunotherapies have been also under evaluation. Conclusions Major advances in the understanding of genetic and epigenetic mechanisms of NET development in the context of inherited endocrine disorders have led to the recognition of molecular targetable alterations, providing a rationale for the implementation of treatments and development of novel targeted therapies.
Article
Full-text available
In this review, we detail the changes and the relevant features that are applied to neuroendocrine neoplasms (NENs) in the 2022 WHO Classification of Endocrine and Neuroendocrine Tumors. Using a question-and-answer approach, we discuss the consolidation of the nomenclature that distinguishes neuronal paragangliomas from epithelial neoplasms, which are divided into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The criteria for these distinctions based on differentiation are outlined. NETs are generally (but not always) graded as G1, G2, and G3 based on proliferation, whereas NECs are by definition high grade; the importance of Ki67 as a tool for classification and grading is emphasized. The clinical relevance of proper classification is explained, and the importance of hormonal function is examined, including eutopic and ectopic hormone production. The tools available to pathologists for accurate classification include the conventional biomarkers of neuroendocrine lineage and differentiation, INSM1, synaptophysin, chromogranins, and somatostatin receptors (SSTRs), but also include transcription factors that can identify the site of origin of a metastatic lesion of unknown primary site, as well as hormones, enzymes, and keratins that play a role in functional and structural correlation. The recognition of highly proliferative, well-differentiated NETs has resulted in the need for biomarkers that can distinguish these G3 NETs from NECs, including stains to determine expression of SSTRs and those that can indicate the unique molecular pathogenetic alterations that underlie the distinction, for example, global loss of RB and aberrant p53 in pancreatic NECs compared with loss of ATRX, DAXX, and menin in pancreatic NETs. Other differential diagnoses are discussed with recommendations for biomarkers that can assist in correct classification, including the distinctions between epithelial and non-epithelial NENs that have allowed reclassification of epithelial NETs in the spine, in the duodenum, and in the middle ear; the first two may be composite tumors with neuronal and glial elements, and as this feature is integral to the duodenal lesion, it is now classified as composite gangliocytoma/neuroma and neuroendocrine tumor (CoGNET). The many other aspects of differential diagnosis are detailed with recommendations for biomarkers that can distinguish NENs from non-neuroendocrine lesions that can mimic their morphology. The concepts of mixed neuroendocrine and non-neuroendocrine (MiNEN) and amphicrine tumors are clarified with information about how to approach such lesions in routine practice. Theranostic biomarkers that assist patient management are reviewed. Given the significant proportion of NENs that are associated with germline mutations that predispose to this disease, we explain the role of the pathologist in identifying precursor lesions and applying molecular immunohistochemistry to guide genetic testing.
Article
Full-text available
Importance: Data about the optimal timing for the initiation of peptide receptor radionuclide therapy (PRRT) for advanced, well-differentiated enteropancreatic neuroendocrine tumors are lacking. Objective: To evaluate the association of upfront PRRT vs upfront chemotherapy or targeted therapy with progression-free survival (PFS) among patients with advanced enteropancreatic neuroendocrine tumors who experienced disease progression after treatment with somatostatin analogues (SSAs). Design, setting, and participants: This retrospective, multicenter cohort study analyzed the clinical records from 25 Italian oncology centers for patients aged 18 years or older who had unresectable, locally advanced or metastatic, well-differentiated, grades 1 to 3 enteropancreatic neuroendocrine tumors and received either PRRT or chemotherapy or targeted therapy after experiencing disease progression after treatment with SSAs between January 24, 2000, and July 1, 2020. Propensity score matching was done to minimize the selection bias. Exposures: Upfront PRRT or upfront chemotherapy or targeted therapy. Main outcomes and measures: The main outcome was the difference in PFS among patients who received upfront PRRT vs among those who received upfront chemotherapy or targeted therapy. A secondary outcome was the difference in overall survival between these groups. Hazard ratios (HRs) were fitted in a multivariable Cox proportional hazards regression model to adjust for relevant factors associated with PFS and were corrected for interaction with these factors. Results: Of 508 evaluated patients (mean ([SD] age, 55.7 [0.5] years; 278 [54.7%] were male), 329 (64.8%) received upfront PRRT and 179 (35.2%) received upfront chemotherapy or targeted therapy. The matched group included 222 patients (124 [55.9%] male; mean [SD] age, 56.1 [0.8] years), with 111 in each treatment group. Median PFS was longer in the PRRT group than in the chemotherapy or targeted therapy group in the unmatched (2.5 years [95% CI, 2.3-3.0 years] vs 0.7 years [95% CI, 0.5-1.0 years]; HR, 0.35 [95% CI, 0.28-0.44; P < .001]) and matched (2.2 years [95% CI, 1.8-2.8 years] vs 0.6 years [95% CI, 0.4-1.0 years]; HR, 0.37 [95% CI, 0.27-0.51; P < .001]) populations. No significant differences were shown in median overall survival between the PRRT and chemotherapy or targeted therapy groups in the unmatched (12.0 years [95% CI, 10.7-14.1 years] vs 11.6 years [95% CI, 9.1-13.4 years]; HR, 0.81 [95% CI, 0.62-1.06; P = .11]) and matched (12.2 years [95% CI, 9.1-14.2 years] vs 11.5 years [95% CI, 9.2-17.9 years]; HR, 0.83 [95% CI, 0.56-1.24; P = .36]) populations. The use of upfront PRRT was independently associated with improved PFS (HR, 0.37; 95% CI, 0.26-0.51; P < .001) in multivariable analysis. After adjustment of values for interaction, upfront PRRT was associated with longer PFS regardless of tumor functional status (functioning: adjusted HR [aHR], 0.39 [95% CI, 0.27-0.57]; nonfunctioning: aHR, 0.29 [95% CI, 0.16-0.56]), grade of 1 to 2 (grade 1: aHR, 0.21 [95% CI, 0.12-0.34]; grade 2: aHR, 0.52 [95% CI, 0.29-0.73]), and site of tumor origin (pancreatic: aHR, 0.41 [95% CI, 0.24-0.61]; intestinal: aHR, 0.19 [95% CI, 0.11-0.43]) (P < .001 for all). Conversely, the advantage was not retained in grade 3 tumors (aHR, 0.31; 95% CI, 0.12-1.37; P = .13) or in tumors with a Ki-67 proliferation index greater than 10% (aHR, 0.73; 95% CI, 0.29-1.43; P = .31). Conclusions and relevance: In this cohort study, treatment with upfront PRRT in patients with enteropancreatic neuroendocrine tumors who had experienced disease progression with SSA treatment was associated with significantly improved survival outcomes compared with upfront chemotherapy or targeted therapy. Further research is needed to investigate the correct strategy, timing, and optimal specific sequence of these therapeutic options.
Article
Gastric neuroendocrine carcinoma (G-NEC) usually has NEC and adenocarcinoma components and is considered to have a common origin in gastric adenocarcinoma because common pathogenic mutations are shared. However, G-NEC without adenocarcinoma also exists, and it may have a different mechanism of tumorigenesis. We aimed to elucidate the tumorigenesis of G-NEC by focusing on the proportion of NEC components. Thirteen patients with G-NEC were included in this study. Comprehensive genetic analysis using whole-exome sequencing was performed. G-NEC without an adenocarcinoma component was defined as pure NEC. TP53 was detected as the most frequent gene mutation (85% of the patients), independent of classification. RB1, KMT2C, LTBP1, and RYR2 mutations were identified in two of three pure NEC patients but were not detected in other G-NEC patients. Pure NEC has different somatic mutation profile than other NECs. This study provides insights into the mechanism of tumorigenesis in G-NEC.
Article
451 Background: Reports of comprehensive genetic analysis of gastric neuroendocrine carcinoma (G-NEC) are limited, and few have described the tumorigenesis of G-NEC. G-NEC usually has NEC and adenocarcinoma components and is considered to have a common origin in gastric adenocarcinoma because these two tumors share common pathogenic mutations and loss of heterozygosity. However, G-NEC without adenocarcinoma also exists, and it may have a different mechanism of tumorigenesis than that of G-NEC with adenocarcinoma. This study aimed to elucidate the tumorigenesis of G-NEC by focusing on the percentage of NEC component, using comprehensive genetic analysis. Methods: Of the 698 patients who had undergone gastrectomy for gastric cancer between January 2014 and March 2019, this study included 13 patients with G-NEC. Comprehensive genetic analysis using whole-exome sequencing, deep sequencing using a target gene panel, and microarray analysis were performed. NEC was classified according to the 2010 WHO classification. G-NEC without an adenocarcinoma component was defined as pure NEC. Results: There were six patients with mixed adeno-neuroendocrine carcinoma (MANEC), four patients with NEC, and three patients with pure NEC. TP53 was detected as the most frequent gene mutation, independent of classification (85%). RB1, ANKRD17, KMT2C, LTBP1, MAATS1, and RYR2 mutations were identified in two of three pure NEC patients but were not detected in other G-NEC patients. Gene expression analysis showed that six key transcripts of importance in NEC tumorigenesis were upregulated in two patients with pure NEC, while they were downregulated in all six MANEC patients. Conclusions: NEC and MANEC with adenocarcinoma components tend to share common pathogenic mutations, but Pure NEC has different genomic and transcriptomic characteristics than other NECs. This suggests that pure NEC has a different mechanism of tumorigenesis than other G-NECs with adenocarcinoma. This is the first study to present a comprehensive genetic analysis of G-NEC, classified by the percentage of NEC components.
Article
Purpose: Environmental endocrine-disrupting chemicals (EDCs) are a mixture of chemical compounds capable to interfere with endocrine axis at different levels and to which population is daily exposed. This paper aims to review the relationship between EDCs and breast, prostate, testicle, ovary, and thyroid cancer, discussing carcinogenic activity of known EDCs, while evaluating the impact on public health. Methods: A literature review regarding EDCs and cancer was carried out with particular interest on meta-analysis and human studies. Results: The definition of EDCs has been changed through years, and currently there are no common criteria to test new chemicals to clarify their possible carcinogenic activity. Moreover, it is difficult to assess the full impact of human exposure to EDCs because adverse effects develop latently and manifest at different ages, even if preclinical and clinical evidence suggest that developing fetus and neonates are most vulnerable to endocrine disruption. Conclusion: EDCs represent a major environmental and health issue that has a role in cancer development. There are currently some EDCs that can be considered as carcinogenic, like dioxin and cadmium for breast and thyroid cancer; arsenic, asbestos, and dioxin for prostate cancer; and organochlorines/organohalogens for testicular cancer. New evidence supports the role of other EDCs as possible carcinogenic and pregnant women should avoid risk area and exposure. The relationship between EDCs and cancer supports the need for effective prevention policies increasing public awareness.
Article
Introduction: Neuroendocrine neoplasms (NENs) are a heterogeneous group of malignancies mainly arising in the gastroenteropancreatic (GEP) and bronchopulmonary systems, with steadily increasing incidence. The therapeutic landscape has widened and the therapeutic strategy should be based on new sequences and combinations, still debated. Areas covered: Herein, we provide an overview of current approved pharmacological treatments in patients with NENs, with the aim to summarize evidence of efficacy of the main different options in GEP and pulmonary NENs, principally focusing on somatostatin analogs (SSAs), targeted therapy with everolimus and sunitinib, peptide receptor radionuclide therapy (PRRT) and chemotherapy. We discuss biological rationale and toxicities, including current indications according to differentiation and placement in the therapeutic algorithm, clinical trials, and combinations. Furthermore, we recommend areas for further research. Expert opinion: Therapeutic management of patients with NENs represents a challenge for clinicians and the identification of effective sequences and combinations is of utmost importance. Major efforts should be directed to early identify and overcome resistance and to limit toxicity. The progress in the therapeutic management of NENs grows faster and the choice of the best approach should be based on randomized clinical trials, as well as on long-term, real-world data.
Article
Background Uncertainty prevails about the magnitude of excess risk of small bowel cancer in patients with inflammatory bowel disease (IBD). Patients and Methods To quantify the risk of small bowel adenocarcinoma and neuroendocrine tumors in patients with ulcerative colitis (UC) and Crohn’s disease (CD), we undertook a population-based cohort study of all IBD-patients diagnosed in Norway and Sweden from 1987 through 2016. Patients were followed through linkage to national registers. We calculated standardized incidence ratios (SIRs) with 95% confidence intervals (CIs) of small bowel adenocarcinomas and neuroendocrine tumors for patients with CD and UC. We excluded the first year of follow-up to reduce reverse causality. Results Among 142.008 IBD-patients with a median follow-up of 10.0 years, we identified 66 adenocarcinomas and 57 neuroendocrine tumors in the small bowel. The SIR of small bowel adenocarcinoma was 8.3 (95% CI, 5.9-11.3) in CD and 2.0 (95% CI, 1.2-3.1) in UC. The incidence rates of adenocarcinomas were highest in CD with stricturing disease and extent limited to the small bowel, at 14.7 (95% CI, 8.2-24.2) and 15.8 (95% CI, 8.4-27.0) per 100,000 person-years, respectively. The SIR of neuroendocrine tumors was 2.5 (95% CI, 1.5-3.9) in CD and 2.0 (95% CI, 1.4-2.8) in UC. Conclusions Patients with CD experienced an 8-fold increased risk of small bowel adenocarcinomas, while both UC and CD patients experienced an about 2-fold increased risk of neuroendocrine tumors, and UC patients experienced 2-fold increased risk of small bowel adenocarcinoma. The small absolute excess cancer risk suggests that active surveillance to diagnose small intestinal cancer early is unlikely to be cost-effective.
Article
PurposeRisk factors for sporadic GEP-NENs are still not well defined. To identify the main clinical risk factors represents the aim of this study performed by three Italian referral centers for NENs.Methods We performed a retrospective case–control study including 148 consecutive sporadic GEP-NENs and 210 age- and sex-matched controls. We collected data on clinical features, cancer family history and other potential risk factors.ResultsMean age was 58.3 ± 15.8 years; 50% males, primary site was pancreas (50.7%), followed by ileum (22.3%). The 62.8% and 29.1% of cases were G1 and G2, respectively; the 40% had locally advanced or metastatic disease at diagnosis. Independent risk factors for GEP-NENs were: family history of non-neuroendocrine GEP cancer (OR 2.16, 95% CI 1.31–3.55, p = 0.003), type 2 diabetes mellitus (T2DM) (OR 2.5, 95% CI 1.39–4.51, p = 0.002) and obesity (OR 1.88, 95% CI 1.18–2.99, p = 0.007). In the T2DM subjects, metformin use was a protective factor (OR 0.28, 95% CI 0.08–0.93, p = 0.049). T2DM was also associated with a more advanced (OR 2.39, 95% CI 1.05–5.46, p = 0.035) and progressive disease (OR 2.47, 95% CI 1.08–5.34, p = 0.03). Stratifying cases by primary site, independent risk factors for pancreatic NENs were T2DM (OR 2.57, 95% CI 1.28–5.15, p = 0.008) and obesity (OR 1.98, 95% CI 1.11–3.52, p = 0.020), while for intestinal NENs family history of non-neuroendocrine GEP cancer (OR 2.46, 95% CI 1.38–4.38, p = 0.003) and obesity (OR 1.90, 95% CI 1.08–3.33, p = 0.026).Conclusion This study reinforces a role for family history of non-neuroendocrine GEP cancer, T2DM and obesity as independent risk factors for GEP-NENs and suggests a role of metformin as a protective factor in T2DM subjects. If confirmed, these findings could have a significant impact on prevention strategies for GEP-NENs.