ArticlePDF AvailableLiterature Review

Abstract and Figures

Neuroendocrine neoplasms (NENs) are a heterogeneous group of rare tumors with different types of physiology and prognosis. Therefore, prognostic information, including morphological differentiation, grade, tumor stage and primary location, are invaluable and contribute to the formulation of treatment decisions. Biomarkers that are currently used, including chromogranin A (CgA), serotonin and neuron-specific enolase, are singular parameters that cannot be used to accurately predict variables associated with tumor growth, including proliferation, metabolic rate and metastatic potential. In addition, site-specific biomarkers, such as insulin and gastrin, cannot be applied to all types of NENs. The clinical application of broad-spectrum markers, as it is the case for CgA, remains controversial despite being widely used. Due to limitations of the currently available mono-analyte biomarkers, recent studies were conducted to explore novel parameters for NEN diagnosis, prognosis, therapy stratification and evaluation of treatment response. Identification of prognostic factors for predicting NEN outcome is a critical requirement for the planning of adequate clinical management. Advances in 'liquid' biopsies and genomic analysis techniques, including microRNA, circulating tumor DNA or circulating tumor cells and sophisticated biomathematical analysis techniques, such as NETest or molecular image-based biomarkers, are currently under investigation as potentially novel tools for the management of NENs in the future. Despite these recent findings yielding promising observations, further research is necessary. The present review therefore summarizes the existing knowledge and recent advancements in the exploration of biochemical markers for NENs, with focus on gastroenteropancreatic-neuroendocrine tumors.
Content may be subject to copyright.
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021
Abstract. Neuroendocrine neoplasms (NENs) are a
heterogeneous group of rare tumors with different types of
physiology and prognosis. Therefore, prognostic informa
tion, including morphological differentiation, grade, tumor
stage and primary location, are invaluable and contribute to
the formulation of treatment decisions. Biomarkers that are
currently used, including chromogranin A (CgA), serotonin
and neuron‑specific enolase, are singular parameters that
cannot be used to accurately predict variables associated with
tumor growth, including proliferation, metabolic rate and
metastatic potential. In addition, site‑specic biomarkers, such
as insulin and gastrin, cannot be applied to all types of NENs.
The clinical application of broad‑spectrum markers, as it is the
case for CgA, remains controversial despite being widely used.
Due to limitations of the currently available mono‑analyte
biomarkers, recent studies were conducted to explore novel
parameters for NEN diagnosis, prognosis, therapy stratica
tion and evaluation of treatment response. Identication of
prognostic factors for predicting NEN outcome is a critical
requirement for the planning of adequate clinical manage
ment. Advances in ‘liquid’ biopsies and genomic analysis
techniques, including microRNA, circulating tumor DNA or
circulating tumor cells and sophisticated biomathematical
analysis techniques, such as NETest or molecular image‑based
biomarkers, are currently under investigation as potentially
novel tools for the management of NENs in the future. Despite
these recent ndings yielding promising observations, further
research is necessary. The present review therefore summa
rizes the existing knowledge and recent advancements in the
exploration of biochemical markers for NENs, with focus on
gastroenteropancreatic‑neuroendocrine tumors.
Contents
1. Introduction
2. Aim and search strategy
3. Currently available biomarkers
4. Potential novel biomarkers
5. Conclusions
1. Introduction
Neuroendocrine neoplasms (NENs) are a heterogenous group
of rare malignancies that arise from neuroendocrine cells
distributed throughout the body and produce peptide hormones
and/or biogenic amines (1). NENs can be dichotomised into
‘functional’ (F) and ‘non‑functional’ (NF) tumors (2). The
F‑NENs category represents 33% of all NENs and is mainly
characterized by well‑defined clinical symptoms that are
caused by the over secretion of their circulating products (2).
Examples of F tumors include NENs in the midgut (such as the
small intestine), which are classically associated with carci
noid syndrome (CS) due to the overproduction of serotonin (3).
Symptoms of CS have been previously reported to be asso
ciated with the overproduction of serotonin by F NENs. By
contrast, NF NENs are more common in terms of incidence
and can cause mechanical symptoms, including ischemia or
obstruction, the complications of which from local tumor
growth can ultimately result in mortality (2).
Due to the substantial heterogeneity in these NENs both
in terms of clinical aggressiveness and response to therapy,
management of patients with such diseases is a signicant
challenge (4). In total, 60‑80% of NENs have already metasta‑
sized on diagnosis (5). Prognostic and predictive markers have
been intensively investigated to explore the optimal clinical
management strategy for this category of neoplasms (6).
Investigation into prognostic markers contribute valuable
information in understanding the physiology of NENs and their
natural course. They reveal benecial mechanistic information
underlying the aggressive properties of the disease and the risk
of recurrence or death. By contrast, predictive markers can be
used to estimate the benet of a certain therapy compared with
their corresponding condition at baseline, which can assist
in implementing the optimal treatment strategy for the most
favorable outcome (7).
Perspectives on the diagnostic, predictive and prognostic
markers of neuroendocrine neoplasms (Review)
OANA ALEXANDRA CIOBANU1,2, SORINA MARTIN1,2 and SIMONA FICA1,2
1Department of Endocrinology and Diabetes, Elias Hospital, 011461 Bucharest; 2Department of Endocrinology,
Carol Davila University of Medicine and Pharmacy, 20021 Bucharest, Romania
Received July 27, 2021; Accepted September 23, 2021
DOI: 10.3892/etm.2 021.10914
Correspondence to: Mrs. Oana Alexandra Ciobanu, Department
of Endocrinology and Diabetes, Elias Hospital, 17 Marasti Avenue,
Sector 1, 011461 Bucharest, Romania
E‑mail: oana‑alexandra.ciobanu@drd.umfcd.ro
Key words: neuroendocrine neoplasms, biomarkers, prognostic
markers, liquid biopsies, molecular imaging
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
2
At present, the most important prognostic factor for NENs
is morphological differentiation characteristics, which can
be divided into the well‑ or poorly‑differentiated categories,
corresponding to neuroendocrine tumors (NETs) or neuroen
docrine carcinomas (NECs) (8). Furthermore, NENs display
a varying degree of proliferation known as grade (G), which
is approximated using the Ki‑67 proliferation index and
mitotic count (9). Specically, the latest 2019 World Health
Organization (WHO) classication of gastro‑entero‑pancre
atic (GEP)‑NENs distinguishes three grades of NENs that are
classied into the low (G1), intermediate‑(G2) and high‑grade
(G3) categories based on the degree of differentiation, with
the mention that poorly differentiated NECs are considered
denitively G3 (10,11). Although the WHO classication of
tumors is considered to be the gold standard tumor classica
tion, it differs depending on their primary site in the body. For
example, the 2015 WHO Classication of Tumors of the Lung,
Pleura, Thymus and Heart does not make use of the Ki‑67
proliferation index (11). However, the current consensus is to
improve the management of NENs by adopting an uniformized
nomenclature system towards different organs (12). It is hoped
that this new classication will serve as a novel grading tool
that can be introduced into common clinical practice to reveal
essential information regarding NENs (12).
In addition to adequate classication and grading, tumor
staging also carries prognostic signicance (13). European
Neuroendocrine Tumors Society (ENETS) and tumor, nodes
and metastases (TNM) classication of the American Joint
Committee on Cancer (AJCC) are currently used staging
systems that present the classification criteria but are not
identical and are site specic (14,15). A number of studies
have reported comparative data between the two systems
for the classification of pancreatic neuroendocrine tumors
(Pan‑NETs), suggesting a similar prognosis being found
by the two systems in ENETS and TNM of the AJCC, both
regarding progression free‑survival (PFS) and overall survival
(OS) (16,17). However, further studies are needed, as the
ENETS staging system appeared to be superior in stratifying
prognosis for each stage of pancreatic NENs according to a
previous report (18).
However, in terms of the formulation of concrete guide
lines for the clinical management of these tumors, numerous
issues persist due to the lack of comprehensive databases and
registries (9). In addition, an insufcient number of lesion
types have been studied despite wide variations in tumor
heterogeneity (9). The 5‑year survival rate ranges between 15
and 95%, depending on the location of the tumor primary site,
the level of metastatic spread at diagnosis, the available treat‑
ment options and the geographical site of care (19‑21).
Several methods have been reported to predict OS and
PFS, including nomograms taking into account the number
of liver metastases, tumor size and the Ki‑67 index (22), the
blood neutrophil‑lymphocyte ratio, Ki‑67 index and the lymph
node ratio (23) or phosphorylated histone H3 (24). However,
further research is required.
An important obstacle in clinical practice is the scarcity
of a set of sensitive and specic tumor biomarkers (25). The
‘perfect’ biomarker would ideally be characterized by high
sensitivity in diagnosing NENs, good prediction of disease
evolution and response to therapy (26). Currently available
NETs biomarkers belong to the mono‑analyte class and vary
in the rates of sensitivity and specicity for indicating the
biological characteristics, such as primary tumor site, or
functional or non‑functional type secretion. In addition,
these mono‑analyte measurements are unable to dene the
state of disease progression or efcacy of therapy (27), which
frequently do not correlated well with radiological evaluation
data (28). To overcome these shortcomings, interest in the
molecular profiling of NENs is increasing. Recent studies
have demonstrated the utility of molecular imaging as a
viable option for predicting the prognosis of NENs (29,30).
Since the density of somatostatin receptors (SSTRs) can also
be quantitively assessed using immunohistochemistry (IHC)
in the surgical specimens, this can be used to predict treat
ment response (31‑33). Furthermore complementary imaging
modalities or early radiological biomarkers, such as the tumor
grade rate (TGR), have been proposed to contribute valuable
prognostic information (34,35).
Molecular diagnostics have previously been performed
on biopsies of solid tumor tissues (13). As one branch of
this technique, ‘liquid’ biopsy is a non‑invasive strategy that
can provide the opportunity to investigate the molecular
genomic mechanism of tumor cells in circulating blood (the
‘tumor circulome’) to characterize the circulating molecular
proles, which may be useful for subsequent diagnosis and
monitoring (36,37). Emerging biochemical and therapeutic
markers have become increasingly popular since 2004 and
their relevance are expected to increase exponentially by
2050 (13,38,39). Identifying novel parameters is important for
optimally predicting the prognosis and treatment response for
each patient. Therefore, the present review summarizes some
of the current and future directions in this eld of research
with an emphasis on GEP‑NENs.
2. Aim and search strategy
The aim of the present review is to summarize the available
data on the diagnostic, prognostic and predictive biomarkers
for NENs. The review is structured into the following two
parts: i) Discussion of the markers most commonly used in
clinical practice at present; and ii) potential future diagnostic,
prognostic and predictive biomarkers.
Search strategy. Published data in the SCIENCEDIRECT
(https://www.sciencedirect.com/) and PUBMED
(https://pubmed.ncbi.nlm.nih.gov/) databases were collected
and analyzed. Publications from the past 5 years were
prioritized and selected using the following relevant key words
either alone or in combination: ‘Neuroendocrine neoplasms’,
‘neuroendocrine tumors’, ‘biomarkers’, ‘liquid biopsies’ and
‘molecular imaging’. Additional studies were identied by
reviewing the references of all selected articles, whereas publi‑
cations from major scientic meetings were searched manually.
After the exclusion of 52 duplicates, 353 articles were scanned,
following which 99 titles were removed because they didn't
satisfy the subject of the present review. We included A total
of 254 publications were selected for a full‑text evaluation.
The inclusion criteria were clinical and practical relevance in
the management of NENs. Full articles and English language
published original and review papers were included that
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 3
addressed the diagnosis and especially the prognostic and
predictive markers in NENs, with different degrees of statis
tical power due to the rarity of these neoplasms. Exclusion
criteria were case reports and publications with abstracts not
relevant for the present review were removed (case studies
removed, four; publications removed, 129), so that at the end of
the selection process, 121 publications were included (Fig. 1).
3. Currently available biomarkers
A number of biomarkers are currently applied for the diag
nosis of NENs. However, they are frequently not correlated
with diagnosis or clinical outcomes, mainly due to sensitivity
and specicity issues (Table I).
Chromogranin A (CgA). Human CgA is a glycoprotein that
belongs to the family of chromogranins and is localized
in the secretory granules alongside peptide hormones and
catecholamines throughout the neuroendocrine system (40).
Recognition of CgA as the default NET biomarker is widely
applied over the past decade due to its broad‑spectrum nature,
with high levels being found in both F and NF NENs (41‑43).
Yet, CgA detection and reliability are still based on different
non‑standardized assays (44). However, Cg A is highly
expressed in NETs tissue, but can also be measured in serum
or plasma as a widely used circulating tumor marker (40). High
levels of CgA are mainly observed in well‑differentiated NETs
and are associated with larger tumor burden, especially in gut
NETs (5,45). By contrast, CgA suffers from low sensitivity
for poorly‑differentiated tumors, where its production is less
pronounced (40,46).
As a result, enthusiasm for the application of CgA as a
marker is waning, which is compounded by the accumu
lating evidence of its low utility (43,47). The specic cut‑off
value (identified by the receiver operating characteristic
analysis between different assays) (44), primary location of the
NEN (48), endocrine‑associated syndrome (5), disease spread,
liver metastases (49), false‑positive elevations in CgA typi
cally caused by proton pump inhibitors, atrophic gastritis and
kidney failure (13), can all inuence the accuracy of this test.
However, a recent systematic review and meta‑analysis
evaluated the role of CgA in bronchopulmonary NENs because
of the scarcity of evidence in this eld (43). This previous
study reported the clinical utility of CgA for the diagnosis of
lung NENs, especially in small cell lung cancer (SCLC) with
a mean diagnostic specicity of 79.5±3.1 and sensitivity of
59.9±6.8%. Still, this nding require further validation (43).
Urinary 5‑hydroxyindoleacetic acid (5‑HIAA). 5‑HIAA is
a metabolite of serotonin, which is excessively produced by
serotonin‑secreting tumors and is excreted in the urine (13).
Prior to interpretating the 5‑HIAA test measurement in the
urine as a result, pharmacological and dietary artifacts must
rst be ruled out, typically by avoiding the intake of trypto
phan‑ and serotonin‑rich foods (50). Measurement of urinary
5‑HIAA excretion is more practical for patients with primary
midgut (jejunoileal, appendiceal and ascending colon) NETs,
which produces the highest levels of serotonin (51). For carci‑
noid syndrome (CS), this test has a reported sensitivity of >90%
and a specicity of 90% (52). Additionally, a level of 5‑HIAA
>300 mmol/24 h and three ushing episodes per day can be
considered to be a predictive factor of carcinoid heart disease
(CHD) (53). There is also evidence that 5‑HIAA in combination
with N‑terminal probrain natriuretic peptide (NT‑proBNP) can
be accurately used in screening for CHD (54). Nevertheless, its
prognostic role remains controversial.
N‑terminal Pro‑Brain Natriuretic Peptide (NT‑proBNP).
NT‑pro BNP is a peptide that is released by myocardial cells
as a result of an increase in heart volume and pressure and is
used especially for CHD prediction, which has been previously
correlated with patient survival (55,56). A high sensitivity
and specicity for CHD, 92 and 91%, respectively, has been
reported at the cut‑off value of 260 pg/ml (57). In addition,
measurement of the level of serum NT‑proBNP has been
recommended for intestinal NENs and CS for the diagnosis
and follow‑up of CHD (54).
Neuron specic enolase (NSE). NSE is the main enolase‑isoen‑
zyme present in neuronal and neuroendocrine tissues (58).
Although mainly expressed poorly‑differentiated NET cells,
measurement of serum NSE cannot be used to distinguish
among different subtypes of NENs (59). Consequently, although
elevated levels of NSE seems to have a degree of prognostic
value for poor outcome and correlate with tumor burden, NSE
is now rarely used in clinical practice since it is inferior to
CgA in terms of the information that can be extracted (59).
Figure 1. Flow chart for the selection of studies discussed in the present
re vie w.
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
4
However, NSE is currently applied as the default tumor marker
for SCLC diagnosis, prognosis and follow‑up, even though
elevated levels of NSE can also be found in non‑small cell lung
cancer (NSCLC) (58).
Pancreatic polypetide (PP). PP is serum marker produced by
the neuroendocrine cells of the colon and pancreas (60). PP
is considered to be a non‑specic marker for NENs, where
its levels has a modest degree of accuracy as a diagnostic
marker (13). However, it appears that an >50% increase in the
serum levels of PP in GEP‑NETs patients has a direct propor‑
tional relationship with the increase in tumor according to
RECIST 1.1 criteria on conventional imaging (CT scan ± MRI
scan) imaging (60).
Connective tissue growth factor (CTGF). CTGF is a modular
secreted protein that serves an important role in complex
biological mechanisms, including angiogenesis, tumorigen
esis and wound healing (61). In addition, CTGF can regulate
various types of brosis formation, such as cardiac brosis (62).
Therefore, CTGF has a reported sensitivity of 80% in detecting
heart disease (62). For NETs, previous studies have reported
that CTGF expression measured in plasma is more likely to
be found in small‑bowel ileal rather than in bronchial, pancre‑
atic or rectal NETs (63,64). However, a high level of CTGF
was found to be an independent predictive factor for right
ventricular dysfunction (61).
Ki‑67 proliferation index /mitotic count. Mitotic count,
Ki‑67 index and necrosis are typically used for the grading
of NENs (9). While mitotic count is recommended to be
reported as mitoses per mm2 area, in clinical practice this
process could be affected by limited areas available for
counting. On the other hand, Ki 67 is determined using IHC
measured in the most mitotically active areas of the patho
logical specimen, but due to the intratumoral heterogeneity
this process can be affected (9). However, this manner of
scoring can be time‑consuming, which is compounded by
the lack of consensus in the optimal method for determining
the proliferative rate (9,65). In clinical practice, when there
is a discordance between these two types of measurements
in assigning the grade, higher ‘grade counts’ are normally
negatively associated with poorer prognosis (65‑67). For
example, in ~33% of well‑differentiated Pan‑NETs, a G1
grade is determined based on the mitotic count, whereas
the G2 grade is determined using the Ki‑67 prolifera
tion index (66). In addition, the same percentage (33%) of
G2 Pan‑NETs are found for after mitotic count, but G3
according to the Ki‑67 proliferation index (64). Therefore,
the use of Ki‑67 IHC staining is essential for accurately
grading well‑differentiated NETs (65,66)
T h e lat e s t 2 019 W H O cl a ss i c a ti o n of GEP N E Ns (10,67 )
now recognizes the category of high‑grade tumors as those
with a Ki‑67 index of >20%, of which well‑differentiated
GEP‑NENs represent ≤7% of all NENs (68). In addition,
tumor grade based on the Ki‑67 index cut‑off values has been
demonstrated to correlate with patient survival independent
of tumor stage in primary and metastatic GEP‑NENs (69,70)
In particular, the Ki‑67 index has also been considered
to be a viable prognostic marker for recurrence after the
resection of PanNETs (71). In liver metastases, the Ki‑67
index has a predisposition to be higher compared with that
in primary GEP‑NETs, where a proportional relationship
between the size of the metastasis and the Ki‑67 index was
found (72‑74)
In clinical practice, detection of multiple primary
tumors, multiple lymph node metastases and/or multiple
distant metastases are recommended for evaluating tissue
blocks with the biggest focus of tumor due to the intratu
moral heterogeneity (75). Furthermore, the Ki‑67 index
may present a variability during the course of the disease or
between primary tumor and metastasis (76). In a previous
study, the Ki‑67 index was evaluated in 103 GEP‑NETs, of
which 24% presented with higher grades in the metastasis,
10% with higher grades in the primary tumor and 66%
with same grade between the metastasis and the primary
tumor (77). The PFS and OS were found to be identical for
both G1 primaries and G2 metastasis categories, and also
for G2 primaries and metastasis; however, these are worse
compared with the G1 primary tumors only (G1 stable
category). Therefore, any G2 tumors (in primary or in
metastases of NETs) inuences patient survival.
The Ki‑67 index also hold promising potential for
clinical application in NECs, since a Ki‑67 index with a 55%
cut‑off for response was found to be associated with poorer
prognosis, favorable response to platinum‑based chemo
therapy (CHT) (78) and adverse reactions to temozolomide
CHT (79).
Table I. Sensitivity and specicity of current biomarkers.
First author/s, year Tumor marker Primary tumor location Sensitivity % Specicity % (Refs.)
Oberg et al, 2015 Chromogranin A Nonspecic 60‑90 <50 (2)
Feldman, 1986 Urinary 5‑hydroxyindole Midgut 70 90 (51)
acetic acid
Bhattacharyya et al, 2008 N‑terminal probrain Midgut: carcinoid heart 92 91 (57)
natriuretic peptide disease
Baudin et al, 1998 Neuron specic enolase Nonspecic 33 68 (59)
Walter et al, 2012 Pancreatic polypeptide Pancreas No data 84 (60)
Laskaratos et al, 2017 Connective tissue Midgut: right ventricular 88 69 (61)
growth factor dysfunction
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 5
4. Potential novel biomarkers
The current demand in this eld is to improve the methodology
for the diagnosis, treatment and prognosis of NENs. Therefore,
novel markers have been evaluated over the past decade.
Genetic mutations. Recent studies have benefited from
advances in sequencing technology for characterizing the
complex molecular landscape of NENs (80,81). Regarding
PanNETs, the current trend is mainly focused on deter
mining the roles of multiple endocrine neoplasia 1 (MEN1),
death‑domain associated protein (DAX X), α‑thalassemia and
mental retardation syndrome X‑linked (ATR X) genes in the
alternative lengthening of telomeres (ALT) axis and compo
nents in the mTOR and DNA damage pathways (82,83). In
particular, molecular alterations have been consistently associ‑
ated with the following four events: DNA damage repair; cell
cycle regulation; PI3K/AKT/mTOR signaling; and telomere
maintenance (84).
DAXX and ATRX expression and ALT. Recent studies have
revealed increasingly consistent ndings despite the lower
mutation burden of PanNETs (82) as mutations in the MEN1,
ATRX and DA XX genes were frequently observed in this type
of tumors (85‑87). In total, up to 40% of well‑differentiated
NENs in the pancreas exhibit somatic mutations in the DAXX
and ATRX genes (82). These two genes encode proteins that
interact and have multiple cellular functions, including modu‑
lating telomeric chromatin. ATRX and DAX X interact to deposit
histone H3.3‑containing nucleosomes in the centromeric and
telomeric regions of the genome and may interact to suppress
the ALT pathway under normal circumstances (88,89).
ATRX/DAXX mutations result in loss of nuclear expression
of their proteins, as detected by IHC, in tumor tissue, which
correlates with the suppression of ALT (90). A negative
expression of ATRX/DAXX is typically associated with
well‑differentiated NENs, and correlate with worse survival
in Pan‑NETs (90,91). ALT is a telomerase‑independent
telomere maintenance mechanism that has been previously
studied using uorescence in situ hybridization (92). Altered
telomeres are a key process frequently found in PanNETs (93).
As such, positive ALT status in liver metastases of NENs was
found to associate with worse survival and increased risk of
recurrence (94,95). It has been previously shown that there is
100% concordance among the ALT phenotype, ATRX/DAXX
mutations and/or protein loss (92), where the presence of
ALT‑positive and ATRX/DAXX‑negative (inactivation muta
tion) in well‑differentiated PanNETs is associated with a
signicantly higher grade, size, grading, vascular/perineural
invasion, metastatic disease and with reduced relapse‑free
and tumor‑specic survival (96). Therefore, this prole can be
applied as a marker of more aggressive PanNET phenotypes
for patient stratication (97,98).
Application of fine‑needle aspiration (FNA) makes it
possible to detect the loss of ATRX/DAXX and the presence
of ALT as a non‑invasive method to sample tumors. As afore‑
mentioned, somatic mutations of ATRX/DAXX genes can be
detected using IHC to indicate a loss of nuclear expression of
their respective proteins (90). In addition, ALT can be assessed
using telomere‑specic uorescence in situ hybridization (92).
This procedure is becoming increasing important in clinical
practice, through which ALT or the loss of ATRX or DAX X
expression can be veried with higher degrees of condence
during the prognostic process (92).
mTOR signaling pathway. Somatic gene alterations are
currently studied in PanNETs (98,99), of which two processes
have been frequently observed to be affected chromatin
remodeling and activation of PI3K/Akt/mTOR signaling (100).
The mTOR pathway regulates cell proliferation, cell cycle
and apoptosis; however, in tumor cells, an abnormal activa
tion of mTOR pathway influences the tumor to grow and
metastasize (101). Since 14.7% of PanNET cases exhibit
mutations in the PI3K/Akt/mTOR pathway, this nding that
can be exploited to select patients for treatment with mTOR
inhibitors (100,102).
Mutations in the tuberous sclerosis complex 2 (TSC2) and
PTEN genes are suppressors of the Akt/mTOR pathway, which
are present in up to 11% of sporadic PanNETs (100). Reduced
expression of both PTEN and TSC2 was found to be associated
with more aggressive phenotypes, presence of liver metastases
and reduced disease‑free survival and OS in a cohort of 72
primary PanNETs according to microarray analysis (99).
However, further studies are warranted before mutations
of components in the PI3K pathway can be considered as a
biomarker of response.
Retinoblastoma protein 1 (RB1) and p53 in NECs but not in
NE Ts. The most recent 2019 WHO classication of GEP‑NENs
made a clear distinction between NET G3 and NEC (10). NET
G3 lacks consensus‑based recommendations is associated
with longer OS (98). In certain cases, histology is not suf
cient for differentiating between highly proliferative NETs
and NECs (103), where it is hoped that molecular proling can
amend this deciency. Abnormal immunolabeling of p53 and
RB1 pathways are currently being proposed for the distinction
of well‑differentiated NETs, especially NETs G3 from poorly
differentiated NECs during diagnosis (103). Although p53
and RB1 are considered to be two key drivers of NECs, but in
NETs these mutations are rarely observed (104‑107). Double
inactivation of the TP53 and RB1 genes is one of the genetic
signatures of SCLC (108). The presence of these inactivated
mutations is used as a predictive marker of the response to
platinum‑based CHT in lung and GEP‑NECs (104,105,108). In
addition, the loss of RB1 is associated with superior responses
to platinum salts in both lung and pancreas NECs (104,109),
whilst the presence of p53 staining in colorectal NECs is
associated with weaker responses to platinum‑based CHT and
worse prognosis (105). In GEP‑NECs, p53 IHC is currently
under examination as a possible diagnostic, prognostic and
predictive method for GEP‑NECs, which was reported in
several studies with frequencies of p53 immunoreactive cells
ranging from 20‑100% (103), where mutations in TP53 associ
ates poor survival, moreover TP53 being the most prevalent
mutation in NECs (105,110).
Expression markers
Insulinoma‑associated protein 1 (INSM1). INSM1 is a
zinc‑nger family of transcription factors that serves a role
in neurogenesis and neuroendocrine cell differentiation (111).
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
6
INSM1 confers certain advantages in NECs as it more sensitive
compared with traditional general neuroendocrine markers,
such as CgA or synaptophysin, since the clear nuclear expres‑
sion pattern of INSM1 facilitates its accurate interpretation
and quantication whilst reducing the incidence of staining
artifacts (65).
The Notch/hairy and enhancer of split‑1 (Hes‑1) signaling
pathway serves a key role in tumor growth and development,
which has also been reported to inhibit INSM1 (112). INSM1
has recently been reported to be an important biomarker in
the diagnosis of SCLC as it serves as an important factor in
ASCL1‑driven pathways. ASCL1 is needed for the protein
expression of NE molecules and in the development of lung
NE cells (113,114). Furthermore, functional interactions
between ASCL1 and the Notch1‑Hes1 pathway have been
reported (113). As aforementioned, the Notch1‑Hes1 pathway
is involved in the suppression of INSM1 expression (112).
Moreover, the implication of INSM1 suppression is being
studied as a modulator factor in PanNETs, which could be
connected to the Notch1‑Hes1 signaling pathway (112,115).
Tanigawa et al (112) revealed that INSM1 expression was
positive in all cases of PanNETs but negative in cases of pancre‑
atic ductal adenocarcinoma (PDAC), rendering it a potential
marker for distinguishing between PanNETs and PDAC. In
addition, INSM1 has also been found to be a potential marker
for SCLC, since a number of studies reported that INSM1
expression was present in 97‑100% SCLC cell lines, which
appeared to be more sensitive and specic compared with CgA
and l‑3,4‑dihydroxyphenylalanine decarboxylase (111,113).
Furthermore, recent studies reported the superior sensitivity of
INSM1 for the diagnosis of NENs regarding their neuroendo‑
crine origin (65,116). Bellizzi et al (65) studied a cohort of 93
NECs, where 95% sensitivity was found for INSM1 compared
with 83 and 82% for CgA and synaptophysin, respectively.
NETest‑a transcriptomic signature of NETs. The NETest uses
multianalyte assays with algorithmic analyses (MAAAs),
which is a novel method including procedures that incorpo
rate results derived from multiple assays and can increase
both the sensitivity and specicity (117). This assay involves
mRNA isolation, cDNA synthesis followed by the subse
quent quantitative‑PCR measurement of 51 circulating NET
marker genes (117). These genes were chosen based on the
analyzed microarray datasets containing the cellular proles
of fresh frozen tumor samples and whole blood samples from
patients diagnosed with NET to characterize the expression
patterns (117). Finally, a multi‑analyte liquid biopsy represen‑
tative for NETs was used to determine the biological activity
of the tumor and therefore clinical status of the patient (118).
Results are presented as the numeric score (NET score) that
include the following three categories: i) Low, ≤2140%;
ii) intermediate, 41‑79%; and iii) high (biologically aggres
sive), ≥80%. The PCR test is standardized and reproductible;
moreover, diet, proton pump inhibitors medication, age,
sex and ethnicity do not interfere with the accuracy of the
test (119,120). In addition, MAAA is reproducible at multiple
time points, which provides real‑time NET scores (118,120).
In a cohort of 100 patients with three different types of
tumors (GEP, lung bronchopulmonary NETs and unknown
origin tumors), the NETest test had a reported diagnostic
sensitivity of 96%, which was >90% effective in combination
with diagnostic imaging for guiding treatment decisions (121).
This previous study also demonstrated that a low NET score
(≤40%) was associated with superior outcomes, where the
PFS was not reached, whilst intermediate to high NETest
scores (41‑100%) were associated with signicantly shorter
PFS and treatment failures (121). In addition, the NETest has
been documented to be a useful tool for the detection of lung,
thymic, pancreatic and gastrointestinal tract NETs, as well as
paragangliomas and pheochromocytomas with ≥90% accu
racy (39). Moreover, the NETest can predict aggressive tumor
behavior in other NENs (122,123), or the outcome following
tumor resection and efcacy of medical treatments, such as
somatostatin analogues (SSAs) or peptide receptor radio‑
nuclide therapy (PRRT) (39).
A recent meta‑analysis was conducted to assess the
eligibility of NETest as a biomarker tool in the field of
oncology (124). The results supported the utility of this test
as a diagnostic tool for GEP and bronchopulmonary NETs,
which reported an accuracy of 85% in differentiating between
stable and progressive disease whereas a specicity of ~90%
was reported (124).
Aristaless related homeobox (ARX)/pancreatic and duodenal
homeobox 1 (PDX1) expression. ARX and PDX1 are regula
tory proteins involved as epigenetic modiers in pancreatic
development (86). The assessment of ARX/PDX1 expression
has been previously studied as a potential pre‑operative risk
stratication marker for PanNETs (125). Since >50% PanNETs
already have liver metastases at rst presentation, novel strate
gies are emerging for the other 50% with aims to reduce the
risk of metastasis (126). Although the choice of surgery for
the primary tumor can reduce the risk of metastasis, it is also
associated with increased risk of post‑surgery morbidity and
mortality. Therefore, apart from tumor size, transcription
factors ARX and PDX1 alongside ATRX/DAXX mutations
and the status of ALT were studied as prognostic markers
in resected NF PanNETs (96). Cytological specimens of
PanNETs obtained by endoscopic FNA was previously studied
using the IHC staining of ARX, PDX1 and telomere‑specic
uorescence in situ hybridization to detect ALT (125). Positive
ALT activity and ARX expression in the tumor coupled with
negative PDX1 staining was documented to predict metastatic
phenotype for the stratication of patients into the low‑ or
high‑risk groups preoperatively (125).
MicroRNA (miRNA or miR) markers. miRNAs are a class of
RNAs that do not encode protein and are ~22 nucleotides in
length (127). They typically regulate post‑transcriptional gene
expression by targeting mRNA molecules (127). Circulating
miRNAs can be used as minimally invasive biomarkers of
ovarian, cervical or breast cancer as they are readily detectable
in a wide variety of biouids, including plasma, serum and
saliva (128).
The global microRNA expression patterns were studied
in normal pancreas, PanNETs and acinar carcinomas in order
to assess the role of microRNAs in malignant transforma
tion and progression (129). miR‑103 and miR‑107 expression
levels were found to be higher whereas the level miR‑155
expression was lower in PanNETs compared with the normal
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 7
pancreas (129). Furthermore, miR‑21 expression level appears
to be higher in the plasma of patients with PanNETs compared
with that in patients with chronic pancreatitis (129). Therefore,
it can be applied both as a diagnostic and a prognostic tool for
PanNETs, since higher levels of miR‑21 expression and high
Ki‑67 proliferation index were associated with the presence of
metastases (129,130).
It was recently reported that in tumors without a known
primary site, miRNA expression can be used to facilitate
diagnosis (131). miRNA expression proles were previously
analyzed in four pathological types of GEP‑NETs, including
samples from pancreatic, ileal, appendiceal and rectal
NETs (131). The results were promising, as the midgut
NETs (ileum and appendix) could be discriminated from
non‑midgut NETs (rectum, pancreas) according to miR‑615
and miR‑92b expression (131). In addition, ileal NETs could be
discriminated from appendiceal NETs according to miR‑125b,
miR‑192 and miR‑149 expression, whilst rectal NETs could be
distinguished from pancreatic NETs based on miR‑429 and
miR‑487b expression (131).
Due to their stability in the circulation and abundance,
cell‑type and disease stage specificity and their reported
roles in a number of biological processes, miRNAs have been
investigated in various studies. However, due to the lack of
consistency in the reported signatures (between results that
used tissue and those that used circulating blood) for the same
disease and the lack of standardization methods with accurate
techniques, further research is required (132,133).
Methylation markers. Epigenetic events occur depending on
the subtype of NETs (134). DNA hypermethylation is an early
event that frequently occurs during cancer initiation and can
dictate the rate of disease development and progression (135).
The ‘Hypermethylation phenotype’ was associated with poorer
OS and with more progressive disease in PanNETs (135).
O‑6‑methylguanine‑DNA methyltransferase (MGMT) for
NETs but not for NECs. The role of the MGMT enzyme is
to repair DNA lesions as a result of alkylating agents usually
used in NENs. Loss of MGMT function occurs as a result of
epigenetic events, such as hypermethylation, of the MGMT
gene promoter (136). This in turn leads to the loss of MGMT
protein expression, which can be detected using IHC or
detected on the gene level by methylation analysis (for example
using methyl‑specic PCR or pyrosequencing). Analysis of
the MGMT status can be used to predict the prognosis and
response to alkylating agents that induce DNA damage in
well‑differentiated NENs (107). Previous studies showed
that reduced MGMT expression is associated with increased
rates of treatment response to temozolomide, dacarbazine and
streptozotocin CHT in digestive and lung NENs (136,137).
Circulating biomarkers
Cell‑free DNA (cfDNA). The impor tance of tumor‑specic alter
ations in cell‑free DNA (cfDNA) in liquid biopsies is becoming
increasingly recognized, which can either complement or
replace tissue biopsies for several types of cancer, including
NSCLC with mutations in the EGFR gene (138,139). cfDNA
consists of a proportion of circulating tumor DNA (ctDNA) in
the blood plasma, which originates from the tumor following
apoptosis, necrosis and active secretion (140,141). cfDNA can
be used as a biomarker for cancer, as patients with patients are
reported to have greater levels of plasma cfDNA compared with
tumor‑free controls (142); in addition, high levels of cfDNA are
also described in other diseases, such as autoimmune disor
ders (143). However, further studies in NENs are required to
verify the utility of cfDNA as a proling tool. Boons et al (141)
rst reported that the presence of ctDNA through the identi
cation of copy number variations and tumor specic point
mutations using shallow whole genome sequencing and
droplet digital PCR, respectively can be used to differentiate
between metastatic and localized PanNET. The results of
this study demonstrated that ctDNA is found in the plasma
samples of patients with metastatic disease. This was shown
by tumor‑specic variants that were obtained through whole
exome sequencing (WES) analysis of primary tumor tissue and
germline DNA, in comparison with localized PanNETs where
when genotyping variants in cfDNA, the variants could not
be detected. Therefore, cfDNA is a candidate as an alternative
biomarker to tissue biopsies for molecular proling.
Circulating tumor cells (CTCs). CTCs are typically released
into the blood of patients who have undergone epithe
lial and mesenchymal transition, which cause metastatic
disease (13,144). The presence of CTCs at the moment of
recruitment/initial evaluation in the blood samples of patients
with midgut, pancreas, bronchopulmonary and of unknown
primary metastatic NENs was previously associated with
worse PFS and OS (145). In addition, CTCs are associated
with higher tumor grades and burden, high levels of CgA and
higher Ki‑67 indices in G1 and G2 midgut and pancreatic
NETs (146). Their potential predictive role was previously
studied in a cohort of 138 patients with metastatic NENs,
where 41 (29.7%) received long‑term SSAs. Changes in repeat
CTC count at 3‑5 weeks after initiation of therapy were asso‑
ciated with both progressive disease and OS (147). Improved
survival was recorded in patients who did not have CTCs at
baseline and after therapy in addition to those who presented
with >50% CTC reduction after treatment (147). However,
CTC lacks sensitivity and specicity as a diagnostic tool for
different types of NENs (13). This limitation is currently under
investigation in a multicenter, exploratory CalmNET phase IV
study (ClinicalTrials.gov Identier, NCT02075606), which is
monitoring a relatively homogeneous group of patients with
F G1‑G2 midgut NETs treated with lanreotide autogel. This
study is attempting to evaluate the predictive power of CTC
count on clinical outcome, PFS and quality of life (148).
Molecular imaging as biomarkers
Somatostatin receptors agonists. Expression of SSTR in the
majority of NENs, particularly subtype 2, can be imaged by
labelling SSAs with a radionuclide, which is typically 68Ga,
using a chelate, such as 1,4,7,10‑tetraazacyclododecane‑1,4,7,1
0‑tetraacetic acid (DOTA) (149,150).
At present, two types of molecular imaging that can be
used to target SSTR in clinical practice: 111In‑pentetreotide
(OctreoScan) and 68Ga‑DOTA‑Phe1‑Ty r 3 O ctreot ide
(TOC), 68Ga‑DOTA‑NaI3‑Octreotide (NOC) or
68GaDOTA‑Tyr3‑Octreotate (TATE). 68Ga‑DOTA‑
TATE/TOC/NOC‑PET/CT confers higher scanner
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
8
sensitivity, superior spatial resolution and require lower radia‑
tion doses (151). Although all three radiotracers (DOTA‑TOC,
DOTA‑NOC and DOTA‑TATE) have good afnity for SSTR
types 2 and 5, 68Ga‑DOTA‑NOC exhibits higher afnity for
SSTR type 3 (152). However, 68Ga‑DOTA‑TATE confers
superior diagnostic precision for the nuclear imaging of NENs
and is currently used in USA, whilst 68Ga‑DOTATOC‑PET is
mainly used clinically in the European Union (153,154).
68Ga‑DOTA‑TATE‑PET has previously demonstrated
its superiority over Octreoscan for the location of primary
GEP‑NETs, with a 95.1% detection rate compared with
30.9 and 45.5% from other conventional imaging modali
ties, respectively (155). This outcome changed the medical
recommendation in 32.8% of the patients (155). Results from
68Ga‑DOTA‑TOC‑PET/CT imaging also demonstrated a high
afnity to SSTR2 expression derived from IHC and can serve
as a predictive marker for patient response to treatment with
PRRT (156,157). In addition, 68Ga‑DOTATOC‑PET/CT can be
used to select patients who may potentially benet from SSAs
and PRRT, as a high tumor uptake of 68Ga‑DOTA‑TOC‑PET/CT
can be useful in the treatment selection of the patients (158).
PRRT is a key second‑line treatment option for G1 or G2
midgut NETs with disease progression on SSA treatment (91).
PRRT can be used to identify radiation delivered by radionu‑
clides, such as lutetium‑177 (177Lu) or yttrium‑90 (90Y), to NET
cells following internalization after binding to SSTR (151).
However, the expression of SSTRs is currently being
studied as a predictive marker for treatment response (13).
In the NETTER‑1 prospective randomized phase 3
clinical trial (ClinicalTrials.gov Identifier, NCT01578239),
177Lu‑DOTA‑TATE demonstrated its superiority compared
with high‑dose octreotide i n prolonging the PFS in patients with
midgut NETs (PFS at month 20, 65.3% for 177Lu‑DOTATATE;
10.8% for high‑dose octreotide group) (159).
Somatostatin receptors antagonists. Higher tumor uptake of
radiolabeled somatostatin receptor antagonists their poten
tial role has been studied for diagnostic and therapeutic
approach in NETs (160). Previous studies reported higher
sensitivity and diagnostic accuracy with increased image
contrast for 68Ga‑NODAGA‑JR11 (68Ga‑OPS202), a SSTR‑2
antagonist, compared with agonists 68Ga‑DOTA‑TATE
and 68Ga‑DOTA‑TOC, for staging G1 and G2 GEP‑NETs
(ClinicalTrials.gov identier, NCT02162446) (160,161). A
‘theragnostic pair’‑68Ga/177 Lu‑DOTA‑JR11 combination was
also investigated in a single‑center study (ClinicalTrials.gov
identier, NCT02609737). Although there are indications
that it binds to more cell types compared with DOTATATE
or DOTA‑TOC in low‑grade NETs, this investigation
remain in progress at present. Similarly, a peptide ligand,
68Ga‑DOTA‑bombesin, which can bind to the gastrin
realizing peptide receptor in prostate cancer cells (162),
is another example of this receptor system that is under
evaluation for NENs. Its rst appl ication for PET imaging in
humans for prostate and breast cancer has been previously
reported (162).
18F‑uoro‑deoxyglucose (FDG)‑PET/CT (18F‑FDG‑PET/CT).
18F‑FDG‑PET/CT avidity, especially for detecting G3 NET, was
reported to be an indirect marker of proliferative activity in the
tumor, showing a higher sensitivity (87.5%) than somatostatin
receptor scintigraphy (87.6%) for detecting rapidly progressive
disease (163). In a retrospective study, 18F‑FDG avidity was
measured quantitatively as a potential prognostic marker in a
cohort of 89 patients with metastatic GEP‑NETs (164). These
patients were divided into three groups based on the ratio of
standardized uptake value (SUV) max of the lesion that had
the highest FDG uptake as compared to normal liver uptake of
FDG (tumor‑to‑liver T/L SUV ratio) (ratio ≤1, 1‑2.3 and >2.3).
These three categories associated positively with OS (median
OS not reached after 114 months for patients with T/L SUV
ratio ≤1 vs. 55 months for patients with T/L SUV ratio of 1‑2.3
vs. 13 months for patients with T/L SUV ratio >2.3) (164).
NET‑PET score. Somatostatin receptor imaging (SRI)
is currently considered the gold standard for detecting
well‑differentiated NETs. However, it has a number of limita
tions for detecting high grade NETs due to the possibility of
false‑negatives, since these types of tumors do not express
SSTRs (156). Although 18F‑FDG‑PET can be used for staging
G3 NECs, it is more suited for predicting the prognosis of
well‑differentiated NETs, where higher levels of uptake were
associated with an increased risk of early progression (163).
By contrast, lower levels of uptake is associated with a less
aggressive phenotype of the tumor (163). Chan et al (165)
therefore proposed a grading system combining these two
nuclear imaging techniques (SRI and 18F‑FDG‑PET) as a
single parameter, named ‘NET‑PET’, which was found to
associate with OS. This scoring system is designated into ve
risk category groups: i) Grade P0, negative uptake for both
scans; ii) grade P1, purely STTR‑positive lesions without
FDG uptake above background; iii) grades P2‑P4, interme
diate categories; and iv) grade P5, presence of signicant
FDG‑positive/STTR‑negative disease (166). The NET‑PET
score may influence the initial management method of
patients with well‑differentiated metastatic midgut NET,
since the ENETS guideline (167) recommends CHT or SSA
as the rst‑choice treatment option. During the initial phases
of SSA therapy, there is no consensus on the upper cut‑off
value of the Ki‑67 proliferation index (165). In this case, the
NET‑PET score would favor SSA treatment if there is high
SRI uptake and low FDG uptake, whilst the contrary would
favor CHT (168). The same line of reasoning can be made
during patient selection for PRRT, where high SRI uptake but
low FDG uptake would suggest PRRT as a viable treatment
option, whilst higher FDG uptake and low SRI uptake would
indicate likelihood of PPRT resistance (169). Since this was
a retrospective analysis, it remains to be elucidated how the
individual intermediate classications can inuence the prog
nosis and treatment decision in a prospective study.
Tumour growth rate as an early biological marker.
Accumulating data suggest that the response evaluation
criteria in solid tumors (RECIST 1.1) has several limitations
in predicting the response to different types of systemic treat‑
ments (ST) (34,170). According to RECIST 1.1, the majority
of patients with G1‑2 GEP‑NETs would be classied as having
‘stable disease’ (170). However, even non‑responders can
survive for a long period without disease progression, since it
is known that well‑differentiated GEP‑NETs have a relatively
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 9
slow growth rate (34). Therefore, is important to discover
a tumor marker that can identify patients who are at high
risk of disease progression at the early stage (171). Previous
studies reported TGR to be a dynamic marker, which analyzes
images from two examinations and the time between the
examinations and can reveal benecial quantitative informa
tion regarding the percentage of change in the tumor volume
each month (171,172). The GREPONET I study conrmed
that the TGR measured at 3 months (TGR3m) after starting ST
or watch and wait (WW) treatment with a cut‑off of 0.8%/m
(m, percentage of the change of the tumor size in one month),
could be used in clinical practice to monitor the treatment
response in NETs for the early prediction of PFS (34). In addi‑
tion, an increased TGR3m (≥0.8%/m) was found to associated
with shorter PFS, whilst a decreased TGR3m (<0.8%/m‑) was
found to associate with longer PFS (34). This nding suggests
that patients with high TGR3m should be followed up more regu‑
larly, whilst those with lower TGR3m can receive imaging less
regularly to avoid unnecessary radiation (34). Subsequently,
the GREPONET II study explored whether beginning any ST
including WW can induce any changes in TGR, which was
dened as TGR3m‑TGR0 (TGR0 was calculated by comparing
the baseline and imaging examination performed within
1 year before the baseline scan) (35). Since it was expected
that TGR can be used for monitoring treatment change with no
impact on PFS, further study is required.
5. Conclusions
Selection of the optimal treatment option for patients with
NEN is difficult due to the heterogeneity in the tumor
physiology and varying degrees of aggressiveness. There is a
demand for multidisciplinary tumor management guidelines
driven by data derived from modern radiology and molecular
proling techniques, to inform the optimal medical decision.
Overall, further studies integrating a combination of markers
based on tumor genomics and a large spectrum of radiological
techniques, such as molecular imaging, would be better placed
for shaping the future of clinical NEN research. The present
review highlights the importance of a second opinion for
improving the method of prognostic stratication and choice
of personalized treatment strategies.
Acknowledgements
Not applicable.
Funding
No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
OAC wrote the literature review. SF and SM critically
reviewed the manuscript. Data authentication is not applicable.
All authors read and approved the nal manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
1. Klöppel G: Neuroendocrine neoplasms: Dichotomy, origin and
classications. Visc Med 33: 324‑330, 2017.
2. Oberg K, Modlin IM, de Herder W, Pavel M, Klimstra D,
Frilling A, Metz DC, Heaney A, Kwekkeboom D,
Strosberg J, et al: Consensus on biomarkers for neuroendocrine
tumour disease. Lancet Oncol 16: e435‑e446, 2015.
3. Cheung VTF and Khan MS: A guide to midgut neuroendo
crine tumours (NETs) and carcinoid syndrome. Frontline
Gastroenterol 6: 264‑269, 2015.
4. Pedraza‑Arévalo S, Gahete MD, Alors‑Pérez E, Luque RM and
Castaño JP: Multilayered heterogeneity as an intrinsic hall
mark of neuroendocrine tumors. Rev Endocr Metab Disord 19:
179‑192, 2018.
5. Modlin IM, Gustafsson BI, Moss SF, Pavel M, Tsolakis AV
and Kidd M: Chromogranin A‑biological function and clinical
utility in neuro endocrine tumor disease. Ann Surg Oncol 17:
2427‑2443, 2010.
6. Zatelli MC, Grossrubatscher EM, Guadagno E, Sciammarella C,
Faggiano A and Colao A: Circulating tumor cells and mirnas as
prognostic markers in neuroendocrine neoplasms. Endocr Relat
Cancer 24: R223‑R237, 2017.
7. Sechidis K, Papangelou K, Metcalfe PD, Svensson D,
Weatherall J and Brown G: Distinguishing prognostic and
predictive biomarkers: An information theoretic approach.
Bioinformatics 34: 3365‑3376, 2018.
8. Lloyd RV, Osamura RY, Klöppel G and Rosai J (eds): WHO
Classification of Tumours of Endocrine Organs. 4th edition,
Volume 10. Lloyd RV, Osamura RY, IARC, 2017.
9. Rindi G, Klimstra DS, Abedi‑Ardekani B, Brambilla E,
Asa LS, Bosman TF, Busam JK, Dietel M, Fernandez‑Cuesta L,
Sasano H, et al: A common classication framework for neuro
endocrine neoplasms: An International Agency for Research on
Cancer (IARC) and World Health Organization (WHO) expert
consensus proposal. Mod Pathol 31: 1770‑1786, 2018.
10. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M,
Schirmacher P, Washington KM, Carneiro F and Cree IA; WHO
Classication of Tumours Editorial Board: The 2019 WHO clas
sication of tumours of the digestive system. Histopathology 76:
182‑188, 2020.
11. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM,
Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, et al:
The 2015 World Health Organization classification of lung
tumors: Impact of genetic, clinical and radiologic advances since
the 2004 classication. J Thorac Oncol 10: 1243‑1260, 2015
12. Rindi G and Inzani F: Neuroendocrine neoplasm update: Toward
universal nomenclature. Endocr Relat Cancer 27: R211‑R218,
2020.
13. Herrera‑Martínez AD, Hofland LJ, Gálvez Moreno MA,
Castaño JP, de Herder W W and Feelders RA: Neuroendocrine
neoplasms: Current and potential diagnostic, predictive and
prognostic markers. Endocr Relat Cancer 26: R157‑R179, 2019.
14. Rindi G, Klöpper G, Alhman H, Caplin M, Couvelard A,
de Herder WW, Eriksson B, Falchetti A, Falconi M,
Komminoth P, et al: TNM staging of foregut (neuro)endocrine
tumors: A consensus proposal including a grading system.
Virchows Arch 449: 395‑401, 2006.
15. Rindi G, Klöpper G, Couvelard A, Komminoth P, Körner M,
Lopes JM, McNicol AM, Nilsson O, Perren A, Scarpa A, et al:
TNM staging of midgut and hindgut (neuro) endocrine tumors:
A consensus proposal including a grading system. Virchows
Arch 451: 757‑762, 2007.
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
10
16. Cho JH, Ryu JK, Song SY, Hwang JH, Lee DK, Woo SM,
Joo YE, Jeong S, Lee SO, Park BK, et al: Prognostic validity
of the American joint committee on cancer and the European
neuroendocrine tumors staging classifications for pancreatic
neuroendocrine tumors: A retrospective nationwide multicenter
study in South Korea. Pancreas 45: 941‑946, 2016.
17. Strosberg JR, Cheema A, Weber J, Han G, Coppola D and
Kvols LK: Prognostic validity of a novel American Joint
Committee on Cancer Staging Classification for pancreatic
neuroendocrine tumors. J Clin Oncol 29: 3044‑3049, 2011.
18. Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L,
Buchler WM, Capella C, Caplin M, Couvelard A,
Doglioni C, et al: TNM staging of neoplasms of the endocrine
pancreas: Results from a large international cohort study. J Natl
Cancer Inst 104: 764‑777, 2012.
19. Gustafsson BI, Kidd M, Chan A, Malfertheiner MV and
Modlin IM: Bronchopulmonary neuroendocrine tumors.
Cancer 113: 5‑21, 2008.
20. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, Mares JE,
Abdalla EK, Flemming JB, Vauthey IN, Rashid A and Evans DB:
One hundred years after ‘carcinoid’: Epidemiology of and prog‑
nostic factors for neuroendocrine tumors in 35,825 cases in the
United States. J Clin Oncol 26: 3063‑3072, 2008.
21. Ga rcia‑Carbonero R, Capdevila J, Crespo‑Herrero G, Díaz Pérez JA,
Martínez Del Prado MP, Alonso Orduña V, Sevilla‑García I,
Villabona‑Art ero C, Beguir istain‑ Gómez A, Llanos‑Muñoz M, et al:
Incidence, patterns of care and prognostic factors for outcome
of gastroenteropancreatic neuroendocrine tumors (GEP‑NETs):
Results from the National cancer registry of Spain (RGETNE). Ann
Oncol 21: 1794‑1803, 2010.
22. Ruzzenente A, Bagante F, Bertuzzo F, Aldrighetti L, Ercolani G,
Giuliante F, Ferrero A, Torzilli G, Grazi GL, Ratti F, et al: A novel
nomogram to predict the prognosis of patients undergoing liver
resection for neuroendocrine liver metastasis: An analysis of the
Italian neuroendocrine liver metastasis database. J Gastrointest
Surg 21: 41‑48, 2017.
23. Cao LL, Lu J, Lin JX, Zheng CH, Li P, Xie JW, Wang JB,
Chen QY, Lin M, Tu RH and Huang CM: A novel predictive
model based on preoperative blood neutrophilto‑lymphocyte
ratio for survival prognosis in patients with gastric neuroendo
crine neoplasms. Oncotarget 7: 42045‑42058, 2016.
24. Villani V, Mahadevan KK, Ligorio M, Fernández‑Del Castillo C,
Ting DT, Sabbatino F, Zhang I, Vangel M, Ferrone S,
Warshaw AL, et al: Phosphorylated histone H3 (PHH3) is a
superior proliferation marker for prognosis of pancreatic neuro
endocrine tumors. Ann Surg Oncol 23 (Suppl 5): S609‑S617,
2016.
25. Modlin IM, Moss SF, Chung DC, Jensen RT and Snyderwine E:
Priorities for improving the management of gastroenteropancre‑
atic neuroendocrine tumors. J Natl Cancer Inst 100: 1282‑1289,
2008.
26. Turner GB, Johnston BT, McCance DR, McGinty A, Watson RGP,
Patterson CC and Ardill JE: Circulating markers of prognosis
and response to treatment in patients with midgut carcinoid
tumours. Gut 55: 1586‑1591, 2006.
27. Modlin IM, Oberg K, Taylor A, Drozdov I, Bodei L and Kidd M:
Neuroendocrine tumor biomarkers: Current status and perspec
tives. Neuroendocrinology 100: 265‑277, 2014.
28. Kulke MH, Siu LL, Tepper JE, Fisher G, Jaffe D, Haller DG,
Ellis LM, Benedetti JK, Bergsland EK, Hobday TJ, et al: Future
directions in the treatment of neuroendocrine tumors: Consensus
report of the National cancer institute neuroendocrine tumor
clinical trials planning meeting. J Clin Oncol 29: 934‑943, 2011.
29. Ambrosini V, Kunikowska J, Baudin E, Bodei L, Bouvier C,
Capdevila J, Cremonesi M, de Herder WW, Dromain C,
Falconi M, et al: Consensus on molecular imaging and theranos‑
tics in neuroendocrine neoplasms. Eur J Cancer 146: 56‑73, 2021.
30. Majala S, Seppänen H, Kemppainen J, Sundström J,
Shalin‑Jäntti C, Gullichsen R, Schildt J, Mustonen H,
Vesterinen T, Arola J and Kauhanen S: Prediction of the aggres‑
siveness of non‑functional pancreatic neuroendocrine tumors
based on the dual‑tracer PET/CT. EJNMMI Res 9: 116, 2019.
31. Bodei L, Sundin A, Kidd M, Prasad V and Modlin IM: The
status of neuroendocrine tumor imaging: From darkness to light?
Neuroendocrinology 101: 1‑17, 2015.
32. Righi L, Volante M, Tavaglione V, Billè A, Daniele L, Angusti T,
Inzani F, Pelosi G, Rindi G and Pappotti M: Somatostatin
receptor tissue distribution in lung neuroendocrine tumours: A
clinicopathologic and immunohistochemical study of 218 ‘clini‑
cally aggressive’ cases. Ann Oncol 21: 548‑555, 2010.
33. Reubi JC, Waser B, Cescato R, Gloor B, Stettler C and Christ E:
Internalized somatostatin receptor subtype 2 in neuroendocrine
tumors of octreotide‑treated patients. J Clin Endocrinol Metab 95:
2343 ‑2350, 2010.
34. Lamarca A, Crona J, Ronot M, Opalinska M, Lopez Lopez C,
Pezzutti D, Najran P, Carvhalo L, Franca Bezerra RO, Borg P, et al:
Value of tumor growth rate (TGR) as an early biomarker predictor
of patients' outcome in neuroendocrine tumors (NET)‑The
GREPONET Study. Oncologist 24: e1082‑e1090, 2019.
35. Lamarca A, Ronot M, Moalla S, Crona J, Opalinska M,
Lopez Lopez C, Pezzutti D, Najran P, Carvha lo L, Bezerra ROF, et al:
Tumor growth rate as a validated early radiological biomarker able
to reect treatment‑induced cha nges in neuroendoc rine tumors: The
GREPONET‑2 study. Clin Cancer Res 25: 6692‑6699, 2019.
36. Hanahan D and Weinberg RA: The hallmarks of cancer. Cell 100:
5770, 2000.
37. Hanahan D and Weinberg RA: Hallmarks of cancer: The next
generation. Cell 144: 646‑674, 2011.
38. De Rubis G, Rajeev Krishnan S and Bebawy M: Liquid biopsies
in cancer diagnosis, monitoring, and prognosis. Trends Pharmacol
Sci 40: 172‑186, 2019.
39. Malczewska A, Kos‑Kudła B, Kidd M, Drozdov I, Bodei L, Matar S,
Oberg K and Modlin IM: The clinical applications of a multigene
liquid biopsy (NETest) in neuroendocrine tumors. Adv Med Sci 65:
18‑29, 2020.
40. Nehar D, Lombard‑Bohas C, Olivieri S, Claustrat B, Chayvialle JA,
Penes MC, Sassolas G and Borson‑Chazot F: Interest of chromo
granin a for diagnosis and follow‑up of endocrine tumours. Clin
Endocrinol (Oxf) 60: 644‑652, 2004.
41. Baekdal J, Krogh J, Klose M, Holmager P, Langer SW, Oturai P,
Kjaer A, Federspiel B, Hilsted L, Rehfeld JF, et al: Limited
diagnostic utility of chromogranin A measurements in workup of
neuroendocrine tumors. Diagnostics (Basel) 10: 881, 2020.
42. Gkolnopoulos S, Tsapakidis K, Papadimitriou K, Papamichael D
and Kountourak is P: Chromogranin A as a val id marker in oncology:
Clinical application or false hopes? World J Methodol 7: 9‑15, 2017.
43. Malczewska A, Kidd M, Matar S, Kos‑Kudła B, Bodei L, Oberg K
and Modlin IM: An assessment of circulating chromogranin a as
a biomarker of bronchopulmonary neuroendocrine Neoplasia: A
systematic review and meta‑analysis. Neuroendocrinology 110:
198‑216, 2020.
44. Zatell i MC, Torta M, Leon A, Ambrosio MR, Gion M, Tomassetti P,
De Braud F, Delle Fave G, Dogliotti L and degli Uberti EC;
Italian CromaNet Working Group: Chromogranin A as a marker
of neuroendocrine neoplasia: An Italian multicenter study.
Endocr Relat Cancer 14: 473‑482, 2007.
45. Campana D, Nori F, Piscitelli L, Morselli‑Labate AM, Pezzilli R,
Corinaldesi R and Tomassetti P: Chromogranin A: Is it a useful
marker of neuroendocrine tumors? J Clin Oncol 25: 1967‑1973,
2007.
46. Stridsberg M, Oberg K, Li Q, Engstrom U and Lundqvist G:
Measurements of chromogranin A, chromogranin B (secreto
granin I), chromogranin C (secretogranin II) and pancreastatin
in plasma and urine from patients with carcinoid tumours and
endocrine pancreatic tumours. J Endocrinol 144: 49‑59, 1995.
47. Chan DL, Clarke SJ, Diakos CI, Roach PJ, Bailey DL, Singh S
and Pavlakis N: Prognostic and predictive biomarkers in neuro‑
endocrine tumours. Crit Rev Oncol Hematol 113: 268‑282, 2017.
48. Baudin E, Bidart JM, Bachelot A, Ducreux M, Elias D, Rufé P
and Schlumberger M: Impact of chromogranin A measurement
in the work‑up of neuroendocrine tumors.Ann Oncol 12 Suppl 2:
S79‑S82, 2001.
49. Nölting S, Kuttner A, Lauseker M, Vogeser M, Haug A,
Hermann KA, Hoffmann JN, Spitzweg C, Göke B and
Auernhammer CJ: Chromogranin a as serum marker for
gastroenteropancreatic neuroendocrine tumors: A single center
experience and literature review. Cancers (Basel) 4: 141‑155,
2012.
50. Mashige F, Matsushimal Y, Kanazawal H, Sakuma I, Takai N,
Besshof F and Ohkubo A: Acidic catecholamine metabolites and
5‑hydroxyindoleacetic acid in urine: The inuence of diet. Ann
Clin Biochem 33: 43‑49, 1996.
51. Feldman JM: Urinary serotonin in the diagnosis of carcinoid
tumors. Clin Chem 32: 840‑844, 1986.
52. Sjöblom SM: Clinical presentation and prognosis of gastrointes‑
tinal carcinoid tumours. Scand J Gastroenterol 23: 779‑787, 1988.
53. Bhattacharyya S, Raja SG, Toumpanakis C, Caplin ME,
Dreyfus GD and Davar J: Outcomes, risks and complications of
cardiac surgery for carcinoid heart disease. Eur J Cardiothorac
Surg 40: 168‑172, 2011.
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 11
54. Niederle B, Pape UF, Costa F, Gross D, Kelestimur F, Knigge U,
Öberg K, Pavel M, Perren A, Toumpanakis C, et al: ENETS
consensus guidelines update for neuroendocrine neoplasms
of the jejunum and ileum. Neuroendocrinology 103: 125‑138,
2016.
55. Korse CM, Taal BG, de Groot CA, Bakker RH and Bonfrer JM:
Chromogranin‑A and N‑terminal pro‑brain natriuretic peptide:
An excellent pair of biomarkers for diagnostics in patients with
neuroendocrine tumor. J Clin Oncol 27: 4293‑4299, 2009.
56. Modlin IM, Bodei L and Kidd M: Neuroendocrine tumor
biomarkers: From monoanalytes to transcripts and algorithms.
Best Pract Res Clin Endocrinol Metab 30: 59‑77, 2016.
57. Bhattacharyya S, Toumpanakis C, Caplin ME and Davar J:
Usefulness of N‑terminal pro‑brain natriuretic peptide as
a biomarker of the presence of carcinoid heart disease. Am
J Cardiol 102: 938‑942, 2008.
58. Isgrò MA, Bottoni P and Scatena R: Neuron‑speci c enolase
as a biomarker: Biochemical and clinical aspects. Adv Exp Med
Biol 867: 125‑143, 2015.
59. Baudin E, Gigliotti A, Ducreux M, Ropers J, Comoy E,
Sabourin JC, Bidart JM, Cailleux AF, Bonacci R, Rufé P and
Schlumberger M: Neuron‑specic enolase and chromogranin
A as markers of neuroendocrine tumours. Br J Cancer 78:
1102‑1107, 1998.
60. Walter T, Chardon L, Chopin‑Laly X, Raverot V, Cafn AG,
Chayvialle JA, Scoazec JY and Lombard‑Bohas C: Is the
combination of chromogranin A and pancreatic polypeptide
serum determinations of interest in the diagnosis and follow‑up
of gastro‑entero‑pancreatic neuroendocrine tumours? Eur
J Cancer 48: 1766‑1773, 2012.
61. Laskaratos FM, Rombouts K, Caplin M, Toumpanakis C,
Thirlwell C and Mandair D: Neuroendocrine tumors and brosis:
An unsolved mystery? Cancer 123: 4770‑4790, 2017.
62. Behnes M, Brueckmann M, Lang S, Weiß C, Ahmad‑Nejad P,
Neumaier M, Borggrefe M and Hoffmann U: Connective tissue
growth factor (CTGF/CCN2): Diagnostic and prognostic value in
acute heart failure. Clin Res Cardiol 103: 107‑116, 2014.
63. Cunningham JL, Tsolakis AV, Jacobson A and Janson ET:
Connective tissue growth factor expression in endocrine tumors
is associated with high stromal expression of alpha‑smooth
muscle actin. Eur J Endocrinol 163: 691‑697, 2010.
64. Kidd M, Modlin I, Shapiro M, Camp R, Mane S, Usinger W and
Murren J: CTGF, intestinal stellate cells and carcinoid brogen
esis. World J Gastroenterol 13: 5208‑5216, 2007.
65. Bellizzi AM: Immunohistochemistry in the diagnosis and clas
sication of neuroendocrine neoplasms: What can brown do for
you? Hum Pathol 96: 8‑33, 2020.
66. McCall CM, Shi C, Cornish TC, Klimstra DS, Tang LH,
Basturk O, Mun LJ, Ellison TA, Wolfgang CL, Choti MA, et al:
Grading of well‑differentiated pancreatic neuroendocrine tumors
is improved by the inclusion of both ki67 Proliferative index and
mitotic rate. Am J Surg Pathol 37: 1671‑1677, 2013.
67. Bellizzi AM: Pathologic considerations in gastroenteropancre
atic neuroendocrine tumors. Surg Oncol Clin N Am 29: 185‑208,
2020.
68. Coriat R: Aggressive gastro‑entero‑pancreatic neoplasms. Ann
Endocrinol (Paris) 80: 185‑186, 2019.
69. Pape UF, Jann H, Müller‑Nordhorn J, Bockelbrink A, Berndt U,
Willich SN, Koch M, Röcken C, Rindi G and Wiedenmann B:
Prognostic relevance of a novel TNM classification system
for upper gastroenteropancreatic neuroendocrine tumors.
Cancer 113: 256‑265, 2008.
70. Strosberg J, Nasir A, Coppola D, Wick M and Kvols L:
Correlation between grade and prognosis in metastatic gastro
enteropancreatic neuroendocrine tumors. Hum Pathol 40:
1262‑1268, 2009.
71. Lopez‑Aguiar AG, Ethun CG, Postlewait LM, Zhelnin K,
Krasinskas A, El‑Rayes BF, Russell MC, Sarmiento JM,
Kooby DA, Staley CA, et al: Redefining the Ki‑67 index
stratication for low‑grade pancreatic neuroendocrine tumors:
Improving its prognostic value for recurrence of disease. Ann
Surg Oncol 25: 290‑298, 2018.
72. Zen Y and Heaton N: Elevated Ki‑67 labeling index in ‘synchro‑
nous liver metastases’ of well differentiated enteropancreatic
neuroendocrine tumor. Pathol Int 63: 532‑538, 2013.
73. Grillo F, Albertelli M, Brisigotti MP, Borra T, Boschetti M,
Fiocca R, Ferone D and Mastracci L: Grade increases in
gastroenteropancreatic neuroendocrine tumor metastases
compared to the primary tumor. Neuroendocrinology 103:
452‑459, 2016.
74. Shi C, Gonzalez RS, Zhao Z, Koyama T, Cornish TC, Hande KR,
Walker R, Sandler M, Berlin J and Liu EH: Liver metastases of
small intestine neuroendocrine tumors: Ki‑67 heterogeneity
and World health organization grade discordance with primary
tumors. Am J Clin Pathol 143: 398‑404, 2015.
75. Cives M and Strosberg JR: Gastroenteropancreatic neuroendo
crine tumors. CA Cancer J Clin 68: 471‑487, 2018.
76. Singh S, Hallet J, Rowsell C and Law CH: Variability of Ki67
labeling index in multiple neuroendocrine tumors specimens
over the course of the disease. Eur J Surg Oncol 40: 1517‑1522,
2014.
77. Keck KJ, Choi A, Maxwell JE, Li G, O'Dorisio TM, Breheny P,
Bellizzi AM and Howe JR: Increased grade in neuroendo
crine tumor metastases negatively impacts survival. Ann Surg
Oncol 24: 2206‑2212, 2017.
78. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N,
Osterlund P, Dueland S, Hofsli E, Guren MG, Ohrling K, et al:
Predictive and prognostic factors for treatment and survival in
305 patients with advanced gastrointestinal neuroendocrine
carcinoma (WHO G3): The NORDIC NEC study. Ann Oncol 24:
152‑160, 2013.
79. Welin S, Sorbye H, Sebjornsen S, Knappskog S, Busch C and
Öberg K: Clinical effect of temozolomide‑based chemotherapy
in poorly differentiated endocrine carcinoma after progression
on rst‑line chemotherapy. Cancer 117: 4617‑4622, 2011.
80. Jiang R, Hong X, Zhao Y and Wu W: Application of multiomics
sequencing and advances in the molecular mechanisms of
pancreatic neuroendocrine neoplasms. Cancer Lett 499: 39‑48,
20 21.
81. van Riet J, van de Werken HJG, Cuppen E, Eskens FALM,
Tesselaar M, van Veenendaal LM, Klümpen HJ, Dercksen MW,
Valk GD, Lolkema MP, et al: The genomic landscape of 85
advanced neuroendoc rine neoplasms reveals subtype‑heterogeneity
and potential therapeutic targets. Nat Commun 12: 4612, 2021.
82. Scarpa A, Chang DK, Nones K, Corbo V, Patch AM,
Bailey P, Lawlor RT, Johns AL, Miller DK, Mafcini A, et al:
Whole‑genome landscape of pancreatic neuroendocrine tumours.
Nature 543: 65‑71, 2017.
83. Mafcini A and Scarpa A: Genomic landscape of pancreatic
neuroendocrine tumours: The International cancer genome
consortium. J Endocrinol 236: R161‑R167, 2018.
84. Scarpa A: The landscape of molecular alterations in pancreatic
and small intestinal neuroendocrine tumours. Ann Endocrinol
(Paris) 80: 153‑158, 2019.
85. Pea A, Yu J, Marchionni L, Noe M, Luchini C, Pulvirenti A,
de Wilde RF, Brosens LA, Rezaee N, Javed A, et al: Genetic
analysis of small well‑differentiated pancreatic neuroendocrine
tumors identies subgroups with differing risks of liver metas
tases. Ann Surg 271: 566‑573, 2020.
86. Cejas P, Drier Y, Dreijerink KMA, Brosens LAA, Deshpande V,
Epstein CB, Conemans EB, Morsink FHM, Graham MK,
Valk GD, et al: Enhancer signatures stratify and predict
outcomes of non‑functional pancreatic neuroendocrine tumors.
Nat Med 25: 1260‑1265, 2019.
87. Marinoni I: Prognostic value of DAXX/ATRX loss of expression
and ALT activation in PanNETs: Is it time for clinical implemen‑
tation? Gut gutjnl‑2021‑324664, 2021 (Epub ahead of print).
88. Lewis PW, Elsaesser SJ, Noh KM, Stadler SC and Allis CD:
Daxx is an H3.3‑specific histone chaperone and cooperates
with ATRX in replication‑independent chromatin assembly at
telomeres. Proc Natl Acad Sci USA 107: 14075‑14080, 2010.
89. Clynes D, Jelinska C, Xella B, Ayyub H, Scott C, Mitson M,
Taylor S, Higgs DR and Gibbons RJ: Suppression of the
alternative lengthening of telomere pathway by the chromatin
remodelling factor ATRX. Nat Commun 6: 7538, 2015.
90. Singhi AD, Liu TC, Roncaioli JL, Cao D, Zeh HJ, Zureikat AH,
Tsung A, Marsh JW, Lee KK, Hogg ME, et al: Alternative length‑
ening of telomeres and loss of DAXX/ATRX expression predicts
metastatic disease and poor survival in patients with pancreatic
neuroendocrine tumors. Clin Cancer Res 23: 600‑609, 2017.
91. Pavel M, Öberg K, Falconi M Krenning EP, Sundin A,
Perren A and Berruti A; ESMO Guidelines Committee:
Gastroenteropancreatic neuroendocrine neoplasms: ESMO clin
ical practice guidelines for diagnosis, treatment and follow‑up.
Ann Oncol 31: 844‑860, 2020.
92. VandenBussche CJ, Allison DB, Graham MK, Charu V,
Lennon AM, Wolfgang CL, Hruban RH and Heaphy CM:
Alternative lengthening of telomeres and ATRX/DAXX loss
can be reliably detected in FNAs of pancreatic neuroendocrine
tumors. Cancer Cytopathol 125: 544‑551, 2017.
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
12
93. Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C,
Bettegowda C, Rodriguez FJ, Eberhart CG, Hebbar S, et al:
Altered telomeres in tumors with ATRX and DAXX mutations.
Science 333: 425, 2011.
94. Luchini C, Lawlor RT, Bersani S, Vicentini C, Paolino G,
Mattiolo P, Pea A, Cingarlini S, Milella M and Scarpa A:
Alternative lengthening of telomeres (ALT) in pancreatic neuro‑
endocrine tumors: Ready for prime‑time in clinical practice?
Curr Oncol Rep 23: 106, 2021.
95. Dogeas E, Karagkounis G, Heaphy CM, Hirose K, Pawlik TM,
Wolfgang CL, Meeker A, Hruban RH, Cameron JL and Choti M:
Alternative lengthening of telomeres predicts site of origin in
neuroendocrine tumor liver metastases. J Am Coll Surg 218:
628‑ 635, 2 014.
96. Hackeng WM, Brosens LAA, Kim JY, O'Sullivan R, Sung YN,
Liu TC, Cao D, Heayn M, Brosnan‑Cashman J, An S, et al:
Non‑functional pancreatic neuroendocrine tumours:
ATRX/DAXX and alternative lengthening of telomeres (ALT)
are prognostically independent from ARX/PDX1 expression
and tumour size. Gut gutjnl‑2020‑322595, 2021 (Epub ahead of
print).
97. Kim JY, Brosnan‑Cashman JA, An S, Kim SJ, Song KB,
Kim MS, Kim MJ, Hwang DW, Meeker AK, Yu E, et al:
Alternative lengthening of telomeres in primary pancreatic
neuroendocrine tumors is associated with aggressive clinical
behavior and poor survival. Clin Cancer Res 23: 1598‑1606,
2017.
98. Marinoni I, Kurrer AS, Vassella E, Dettmer M, Rudolph T,
Banz V, Hunger F, Pasquinelli S, Speel EJ and Perren A: Loss of
DAXX and ATRX are associated with chromosome instability
and reduced survival of patients with pancreatic neuroendocrine
tumors. Gastroenterology 146: 453‑460.e5, 2014.
99. Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M,
della Peruta M, Piemonti L, Capurso G, Di Florio A,
delle Fave G, et al: Pancreatic endocrine tumors: Expression
profiling evidences a role for AKT‑mTOR pathway. J Clin
Oncol 28: 245‑255, 2010.
100. Stevenson M, Lines KE and Thakker RV: Molecular genetic
studies of pancreatic neuroendocrine tumors: New therapeutic
approaches. Endocrinol Metab Clin North Am 47: 525‑548, 2018.
101. Zou Z, Tao T, Li H and Zhu X: mTOR signaling pathway and
mTOR inhibitors in cancer: Progress and challenges. Cell
Biosci 10: 31, 2020.
102. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A,
Schulick RD, Tang LH, Wolfgang CL, Choti MA, et al:
DAXX/ATRX, MEN1, and mTOR pathway genes are frequently
altered in pancreatic neuroendocrine tumors. Science 331:
1199‑1203, 2011.
103. Coriat R, Walter T, Terris B, Couvelard A and Ruszniewski P:
Gastroenteropancreatic well‑differentiated grade 3 neuroendo
crine tumors: Review and position statement. Oncologist 21:
1191‑1199, 2016.
104. Hijioka S, Hosoda W, Matsuo K, Ueno M, Furukawa M,
Yoshitomi H, Kobayashi N, Ikeda M, Ito T, Nakamori S, et al:
Rb loss and KRAS mutation are predictors of the response to
platinum‑based chemotherapy in pancreatic neuroendocrine
neoplasm with grade 3: A Japanese multicenter pancreatic
NEN‑G3 study. Clin Cancer Res 23: 4625‑4632, 2017.
105. Ali AS, Grönberg M, Federspiel B, Scoazec JY,
Hjortland GO, Grønbæk H, Ladekarl M, Langer SW, Welin S,
Vestermark LW, et al: Expression of p53 protein in high‑grade
gastroenteropancreatic neuroendocrine carcinoma. PLoS
One 12: e0187667, 2017.
106. Basturk O, Tang L, Hruban RH, Adsay V, Yang Z, Krasinsk as AM,
Vakiani E, La Rosa S, Jang KT, Frankel WL, et al: Poorly
differentiated neuroendocrine carcinomas of the pancreas: A
clinicopathologic analysis of 44 cases. Am J Surg Pathol 38:
437‑447, 2014.
10 7. Scoazec JY: Lung and digestive neuroendocrine neoplasms.
From WHO classification to biomarker screening: Which
perspectives? Ann Endocrinol (Paris) 80: 163‑165, 2019.
108. George J, Lim JS, Jang SJ, Cun Y, Ozretić L, Kong G, Leenders F,
Lu X, Fernández‑Cuesta L, Bosco G, et al: Comprehensive
genomic proles of small cell lung cancer. Nature 524: 47‑53,
2015.
109. Derks J L, Leblay N, Thunnissen E, van Suylen RJ, den Bakker M,
Groen HJM, Smit EF, Damhuis R, van den Broek EC,
Chabrier A, et al: Molecular subtypes of pulmonary large‑cell
neuroendocrine carcinoma predict chemotherapy treatment
outcome. Clin Cancer Res 24: 33‑42, 2018.
110. Vijayvergia N, Boland PM, Handorf E, Gustafson KS, Gong Y,
Cooper HS, Sheriff F, Astsaturov I, Cohen SJ and Engstrom PF:
Molecular proling of neuroendocrine malignancies to identify
prognostic and therapeutic markers: A fox chase cancer center
pilot study. Br J Cancer 115: 564‑570, 2016.
111. Chen C, Notkins AL and Lan MS: Insulinoma‑associated‑1:
From neuroendocrine tumor marker to cancer therapeutics. Mol
Cancer Res 17: 1597‑1604, 2019.
112. Tanigawa M, Nakayama M, Taira T, Hattori S, Mihara Y,
Kondo R, Kusano H, Nakamura K, Abe Y, Ishida Y, et al:
Insulinoma‑associated protein 1 (INSM1) is a useful marker for
pancreatic neuroendocrine tumor. Med Mol Morphol 51: 32‑40,
2018.
113. Fujino K, Motooka Y, Hassan WA, Ali Abdalla MO, Sato Y,
Kudoh S, Hasegawa K, Niimori‑Kita K, Kobayashi H,
Kubota I, et al: Insulinoma‑associated protein 1 is a crucial
regulator of neuroendocrine differentiation in lung cancer. Am
J Pathol 185: 3164‑3177, 2015.
114. Augustyn A, Borromeo M, Wang T, Fujimoto J, Shao C,
Dospoy PD, Lee V, Tan C, Sullivan JP, Larsen JE, et al:
ASCL1 is a lineage oncogene providing therapeutic targets for
high‑grade neuroendocrine lung cancers. Proc Natl Acad Sci
USA 111: 14788‑14793, 2014.
115. Wang H, Chen Y, Fernandez‑Del Castillo C, Yilmaz O and
Deshpande V: Heterogeneity in signaling pathways of gastroen‑
teropancreatic neuroendocrine tumors: A critical look at notch
signaling pathway. Mod Pathol 26: 139‑147, 2013.
116. Rodriguez EF, Fite JJ, Chowsilpa S and Maleki Z:
Insulinoma‑associated protein 1 immunostaining on cytology
specimens: An institutional experience. Hum Pathol 85: 128‑135,
2019.
117. Modlin IM, Drozdov I and Kidd M: The identication of gut
neuroendocrine tumor disease by multiple synchronous tran
script analysis in blood. PLoS One 8: 63364, 2013.
118. Kidd M, Drozdov I and Modlin I: Blood and tissue neuroendo
crine tumor gene cluster analysis correlate, dene hallmarks and
predict disease status. Endocr Relat Cancer 22: 561‑575, 2015.
119. Genzen JR, Mohlman JS, Lynch JL, Squires MW and Weiss RL:
Laboratory‑developed tests: A legislative and regulatory review.
Clin Chem 63: 1575‑1584, 2017.
120. Modlin IM, Aslanian H, Bodei L, Drozdov I and Kidd M: A
PCR blood test outperforms chromogranin A in carcinoid
detection and is unaffected by proton pump inhibitors. Endocr
Connect 3: 215‑223, 2014.
121. Liu E, Paulson S, Gulati A, Freudman J, Grosh W, Kafer S,
Wickremesinghe PC, Salem RR and Bodei L: Assessment
of NETest clinical utility in a U.S. registry‑based study.
Oncologist 24: 783‑790, 2019.
122. Malczewska A, Bodei L, Kidd M and Modlin IM: Blood mRNA
measurement (NETest) for Neuroendocrine tumor diagnosis
of image‑negative liver metastatic disease. J Clin Endocrinol
Metab 104: 867‑872, 2019.
123. Modlin IM, Kidd M, Malczewska A, Drozdov I, Bodei L,
Matar S and Chung KM: The NETest: The clinical utility of
multigene blood analysis in the diagnosis and management of
neuroendocrine tumors. Endocrinol Metab Clin North Am 47:
485‑504, 2018.
124. Öberg K, Califano A, Strosberg JR, Ma S, Pape U, Bodei L,
Kaltsas G, Toumpanakis C, Goldenring JR, Frilling A and
Paulson S: A meta‑analysis of the accuracy of a neuroendocrine
tumor mRNA genomic biomarker (NETest) in blood. Ann
Oncol 31: 202‑212, 2020.
125. Hackeng WM, Morsink FHM, Moons LMG, Heaphy CM,
Offerhaus GJA, Dreijerink KM A and Brosens LAA: Assessment
of ARX expression, a novel biomarker for metastatic risk in
pancreatic neuroendocrine tumors, in endoscopic ultrasound
ne‑needle aspiration. Diagn Cytopathol 48: 308‑315, 2020.
126. Keutgen XM, Schadde E, Pommier RF, Halfdanarson TR,
Howe JR and Kebebew E: Metastatic neuroendocrine tumors
of the gastrointestinal tract and pancreas: A surgeon's plea
to centering attention on the liver. Semin Oncol 45: 232‑235,
2018.
12 7. Demes M, Aszyk C, Bartsch H, Schi rren J and Fisseler‑Eckhoff A:
Differential miRNA‑Expression as an adjunctive diagnostic tool
in neuroendocrine tumors of the lung. Cancers (Basel) 8: 38,
2016.
128. Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A,
Cretoiu D, Suciu N, Cretoiu SM and Voinea SC: MiRNAs as
biomarkers in disease: Latest ndings regarding their role in
diagnosis and prognosis. Cells 9: 276, 2020.
EXPERIMENTAL AND THERAPEUTIC MEDICINE 22: 1479, 2021 13
129. Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S,
Calin GA, Volinia S, Liu CG, Scarpa A and Croce CM:
MicroRNA expression abnormalities in pancreatic endocrine
and acinar tumors are associated with distinctive pathologic
features and clinical behavior. J Clin Oncol 24: 4677‑4684,
2006.
130. Vicentini C, Fassan M, D'Angelo E, Corbo V, Silvestris N,
Nuovo GJ and Scarpa A: Clinical application of microRNA
testing in neuroendocrine tumors of the gastrointestinal tract.
Molecules 19: 2458‑2468, 2014.
131. Panarelli N, Tyryshkin K, Wong JJM, Majewski A, Yang X,
Scognamiglio T, Kim MK, Bogardus K, Tuschl T, Chen YT
and Renwick N: Evaluating gastroenteropancreatic neuroen
docrine tumors through microRNA sequencing. Endocr Relat
Cancer 26: 47‑57, 2019.
132. Kulke MH, Anthony LB, Bushnell DL, de Herder WW,
Goldsmith SJ, Klimstra DS, Marx SJ, Pasieka JL,
Pommier RF, Yao JC, et al: NANETS treatment guidelines:
Well‑differentiated neuroendocrine tumors of the stomach and
pancreas. Pancreas 39: 735‑752, 2010.
133. Reddy KB: MicroRNA (miRNA) in cancer. Cancer Cell Int 15:
38, 2015.
134. Cives M, Simone V, Rizzo FM and Silvestris F: NETs:
organ‑related epigenetic derangements and potential clinical
applications. Oncotarget 7: 57414‑57429, 2016.
135. House MG, Herman JG, Guo MZ, Hooker CM, Schulick RD,
Lillemoe KD, Cameron JL, Hruban RH, Maitra A and Yeo CJ:
Aberrant hypermethylation tumor suppressor genes in pancre
atic endocrine neoplasms. Ann Surg 238: 423‑431; discussion
431‑2, 2 003.
136. Campana D, Walter T, Pusceddu S, Gelsomino F, Graillot E,
Prinzi N, Spallanzani A, Fiorentino M, Barritault M,
Dall'Olio F, et al: Correlation between MGMT promoter
methylation and response to temozolomide‑based therapy in
neuroendocrine neoplasms: An observational retrospective
multicenter study. Endocrine 60: 490‑498, 2018.
13 7. Walter T, van Brakel B, Vercherat C, Hervieu V, Forestier J,
Chayvialle JA, Molin Y, Lombard‑Bohas C, Joly MO and
Scoazec JY: O6‑Methylguanine‑DNA methyltransferase status
in neuroendocrine tumours: Prognostic relevance and asso
ciation with response to alkylating agents. Br J Cancer 112:
523 ‑531, 2015.
138. Marzese DM, Hirose H and Hoon DS: Diagnostic and prognostic
value of circulating tumor‑related DNA in cancer patients.
Expert Rev Mol Diagn 13: 827‑844, 2013.
139. Jänne PA, Yang JC, Kim DW, Planchard D, Ohe Y,
Ramalingam SS, Ahn MJ, Kim SW, Su WC, Horn L, et al:
AZD9291 in EGFR inhibitor‑resistant non‑small‑cell lung
cancer. N Engl J Med 372: 1689‑1699, 2015.
140. Schwarzenbach H, Hoon DS and Pantel K: Cell‑free nucleic
acids as biomarkers in cancer patients. Nat Rev Cancer 11:
426‑437, 2011.
141. Boons G, Vandamme T, Peeters M, Beyens M, Driessen A,
Janssens K, Zwaenepoel K, Roeyen G, Van Camp G and
Op de Beeck K: Cell‑free DNA from metastatic pancreatic
neuroendocrine tumor patients contains tumor‑specific
mutations and copy number variations. Front Oncol 8: 467,
2018.
142. Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C and
Beljanski M: Neoplastic characteristics of the DNA found
in the plasma of cancer patients. Oncology 46: 318‑322,
1989.
143. Raptis L and Menard HA: Quantitation and characterization
of plasma DNA in normals and patients with systemic lupus
erythematosus. J Clin Invest 66: 1391‑1399, 1980.
144. Kalluri R and Weinberg RA: The basics of epithelial‑mesen
chymal transition. J Clin Invest 119: 1420‑1428, 2009.
145. Khan MS, Tsigani T, Rashid M, Rabouhans JS, Yu D, Luong TV,
Caplin M and Meyer T: Circulating tumor cells and EpCAM
expression in neuroendocrine tumors. Clin Cancer Res 17:
337‑345, 2011.
146. Khan MS, Kirkwood A, Tsigani T, Garcia‑Hernandez J,
Hartley JA, Caplin ME and Meyer T: Circulating tumor cells as
prognostic markers in neuroendocrine tumors. J Clin Oncol 31:
365‑372, 2013.
147. Khan MS, Kirkwood AA, Tsigani T, Lowe H, Goldstein R,
Hartley JA, Caplin ME and Meyer T: Early changes in circu
lating tumor cells are associated with response and survival
following treatment of metastatic neuroendocrine neoplasms.
Clin Cancer Res 22: 79‑85, 2016.
148. Childs A, Vesely C, Ensell L, Lowe H, Luong TV, Caplin ME,
Toumpanakis C, Thirlwell C, Hartley JA and Meyer T:
Expression of somatostatin receptors 2 and 5 in circulating
tumour cells from patients with neuroendocrine tumours. Br
J Cancer 115: 1540‑1547, 2016.
149. Krenning EP, Kwekkeboom DJ, Bakker WH, Breeman WA,
Kooij PP, Oei HY, van Hagen M, Postema PT, de Jong M,
Reubi JC, et al: Somatostatin receptor scintigraphy with
[111In‑DTPA‑d‑Phe1]‑ and [123I‑Tyr3]‑octreotide: The
Rotterdam experience with more than 1000 patients. Eur J Nucl
Med 20: 716‑731, 1993.
150. Bombardieri E, Ambrosini V, Aktolun C, Baum RP,
Bishof‑Delaloye A, Del Vecchio S, Mafoli L, Mortelmans L,
Oyen W, Pepe G, et al: 111In‑pentetreotide scintigraphy:
Procedure guidelines for tumour imaging. Eur J Nucl Med Mol
Imaging 37: 1441‑1448, 2010.
151. Hope TA, Bergsland EK, Bozkurt MF, Graham M,
Heaney AP, Herrmann K, Howe JR, Kulke MH, Kunz PL,
Mailman J, et al: Appropriate use criteria for somatostatin
receptor PET imaging in neuroendocrine tumors. J Nucl
Med 59: 66‑74, 2018.
152. Eychenne R, Bouvry C, Bourgeois M, Loyer P, Benoist E and
Lepareur N: Overview of radiolabeled somatostatin analogs for
cancer imaging and therapy. Molecules 25: 4012, 2020.
153. Ivanidze J, Roytman M, Sasson A, Skada M, Fahey TJ III,
Osborne JR and Dutruel SP: Molecular imaging and therapy
of somatostatin receptor positive tumors. Clin Imaging 56:
146 ‑154, 2019.
154. Graham MM, Gu X, Ginader T, Breheny P and Sunderland JJ:
68Ga‑DOTATOC imaging of neuroendocrine tumors: A
systematic review and metaanalysis. J Nucl Med 58: 1452‑1458,
2017.
155. Sadowski SM, Neychev V, Millo C, Shih J, Nilubol N,
Herscovitch P, Pacak K, Marx SJ and Kebebew E:
Prospective study of 68Ga‑DOTATATE positron emis
sion tomography/computed tomography for detecting
gastro‑entero‑pancreatic neuroendocrine tumors and unknown
primary sites. J Clin Oncol 34: 588‑597, 2016.
156. Deroose CM, Hindié E, Kebebew E, Goichot B, Pacak K,
Taïeb D and Imperiale A: Molecular imaging of gastroentero
pancreatic neuroendocrine tumors: Current status and future
directions. J Nucl Med 57: 1949‑1956, 2016.
157. Miederer M, Seidl S, Buck A, Scheidhauer K, Wester HJ,
Schwaiger M and Perren A: Correlation of immunohistopatho‑
logical expression of somatostatin receptor 2 with standardised
uptake values in 68Ga‑DOTATOC PET/CT. Eur J Nucl Med
Mol Imaging 36: 48‑52, 2009.
158. Kratochwil C, Stefanova M, Mavriopoulou E, Holland‑Letz T,
Dimitrakopoulou‑Strauss A, Afshar‑Oromieh A, Mier W,
Haberkorn U and Giesel F L: SUV of [68Ga]DOTATOC‑PET/CT
Predicts Response Probability of PRRT in neuroendocrine
tumors. Mol Imaging Biol 17: 313‑318, 2015.
159. Strosberg J, El‑Haddad G, Wolin E, Hendifar A, Yao J, Chasen B,
Mittra E, Kunz PL, Kulke MH, Jacene H, et al: Phase 3 trial of
(177)Lu‑Dotatate for Midgut Neuroendocrine tumors. N Engl
J Med 376: 125‑135, 2017.
160. Nicolas G, Mansi R, Vomstein S, Kaufmann J, Bouterfa H,
Maecke H, Wild D and Fani M: Wider safety window with
radiolabeled somatostatin receptor antagonists over agonists.
J Nucl Med 56 (Suppl 3): S335, 2015.
161. Fani M, Nicolas GP and Wild D: Somatostatin receptor antago
nists for imaging and therapy. J Nucl Med 58 (Suppl 2): 61S‑66S,
2017.
162. Nock BA, Kaloudi A, Lymperis E, Giarika A, Kulkarni HR,
Klette I, Singh A, Krenning EP, de Jong M, Maina T and
Baum RP: Theranostic perspectives in prostate cancer with the
Gastrin‑Releasing peptide receptor antagonist NeoBOMB1:
Preclinical and rst clinical results. J Nucl Med 58: 75‑80,
2017.
163. Garin E, Le Jeune F, Devillers A, Cuggia M,
de LajarteThirouard AS, Bouriel C, Boucher E and Raoul JL:
Predictive value of 18F‑FDG PET and somatostatin receptor
scintigraphy in patients with metastatic endocrine tumors.
J Nucl Med 50: 858‑864, 2009.
164. Ezziddin S, Adler L, Sabet A, Pöppel TD, Grabellus F,
Yüce A, Fischer HP, Simon B, Höller T, Biersack HJ and
Nagarajah J: Prognostic stratification of metastatic gastroen
teropancreatic neuroendocrine neoplasms by 18F‑FDG PET:
Feasibility of a metabolic grading system. J Nucl Med 55:
1260‑1266, 2014.
CIOBANU et al: CURRENT AND POTENTIAL MARKERS IN NEUROENDOCRINE NEOPLASMS
14
165. Chan DL, Pavlakis N, Schembri GP, Bernard EJ, Hsiao E,
Hayes A, Barnes T, Diakos C, Khasraw M, Samra J, et al: Dual
somatostatin receptor/FDG PET/CT imaging in metastatic
neuroendocrine tumours: Proposal for a novel grading scheme
with prognostic signicance. Theranostics 7: 1149‑1158, 2017.
166. Hindié E: The netpet score: Combining FDG and soma
tostatin receptor imaging for optimal management of patients
with metastatic well‑differentiated neuroendocrine tumors.
Theranostics 7: 1159‑1163, 2017.
167. Pavel M, O'Toole D, Costa F, Cap devila J, Gross D, Kianma nesh R,
Krenning E, Knigge U, Salazar R, Pape UF, et al: ENETS
consensus guidelines update for the management of distant
metastatic disease of intestinal, pancreatic, bronchial neuroen
docrine neoplasms (NEN) and NEN of unknown primary site.
Neuroendocrinology 103: 172‑185, 2016.
168. Panagiotidis E, Alshammari A, Michopoulou S, Skoura E,
Naik K, Maragkoudakis E, Mohmaduvesh M, Al‑Harbi M,
Belda M, Caplin ME, et al: Comparison of the impact of
68Ga‑DOTATATE and 18F‑FDG PET/CT on clinical manage
ment in patients with neuroendocrine tumors. J Nucl Med 58:
91‑9 6, 2017.
169. Nilica B, Waitz D, Stevanovic V, Uprimny C, Kendler D,
Buxbaum S, Warwitz B, Gerardo L, Henninger B, Virgolini I
and Rodrigues M: Direct comparison of (68)Ga‑DOTA‑TOC
and (18)F‑FDG PET/CT in the follow‑up of patients with neuro‑
endocrine tumour treated with the rst full peptide receptor
radionuclide therapy cycle. Eur J Nucl Med Mol Imaging 43:
1585‑1592, 2016.
170. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH,
Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S,
Mooney M, et al: New response evaluation criteria in solid
tumours: Revised RECIST guideline (version 1.1). Eur
J Cancer 45: 228‑247, 2009.
171. Ferté C, Fernandez M, Hollebecque A, Koscielny S, Levy A,
Massard C, Balheda R, Bot B, Gomez‑Roca C, Dromain C, et al:
Tumor growth rate is an early indicator of antitumor drug
activity in phase I clinical trials. Clin Cancer Res 20: 246‑252,
2014.
172. Jain RK, Lee JJ, Ng C, Hong D, Gong J, Naing A, Wheler J
and Kurzrock R: Change in tumor size by RECIST correlates
linearly with overall survival in phase I oncology studies. J Clin
Oncol 30: 2684‑2690, 2012.
This work is licensed under a Creative Commons
Attribution-NonCommercia l-NoDerivatives 4.0
International (CC BY-NC-ND 4.0) License.
... Different biomarkers are used to more specifically diagnose subtypes of functioning pancreatic NENs in combination with other diagnostic (CgA) and imaging tools [69,80]. These markers include insulin, somatostatin, glucagon, gastrin, vasoactive intestinal peptide (VIP), serotonin, adrenocorticotropic hormone (ACTH), catecholamines, calcitonin, growth factor, insulin growth factor 1, and prolactin. ...
... mRNA is isolated from EDTA-collected whole blood samples and real-time PCR is performed to interrogate 51 genes with the aid of four different prediction algorithms [113]. The choice of these 51 genes was developed on tissue-based, blood-based, and literaturecurated panels of genes in order to define the expression profile of NENs [80,113]. In addition, these genes have been confirmed as bona fide neuroendocrine markers in a large dataset (11,232 samples) from The Cancer Genome Atlas (TCGA) [114]. ...
... These aspects can be very advantageous in the diagnosis of NENs, both to distinguish poorly differentiated NETs from non-neuroendocrine tumors and to identify different molecular subgroups [132][133][134]. However, little is known about circulating miRNAs in NENs (Table 3), due to the lack of standardized analysis methods and inconsistency between tissue and circulating signatures [21,59,80,135,136]. Moreover, necrosis in G1 and G2 NET is uncommon; thus, these tumors do not represent an adapted source of miRNAs [134,137]. ...
Article
Full-text available
Neuroendocrine neoplasms (NENs) are a heterogeneous group of diseases that are characterized by different behavior and clinical manifestations. The diagnosis and management of this group of tumors are challenging due to tumor complexity and lack of precise and widely validated biomarkers. Indeed, the current circulating mono-analyte biomarkers (such as chromogranin A) are ineffective in describing such complex tumors due to their poor sensitivity and specificity. In contrast, multi-analytical circulating biomarkers (including NETest) are emerging as more effective tools to determine the real-time profile of the disease, both in terms of accurate diagnosis and effective treatment. In this review, we will analyze the capabilities and limitations of different circulating biomarkers focusing on three relevant questions: (1) accurate and early diagnosis; (2) monitoring of disease progression and response to therapy; and (3) detection of early relapse.
... The presence of ctDNA in patients with NENs is indicative of several tumor characteristics, including primary tumor location, metastatic status, and higher tumor grades, and is associated with poorer prognosis, including lower OS and shorter PFS [112][113][114]. Elevated ctDNA levels generally correlate with advanced disease stages, higher tumor burden, and increased proliferation indices [113,115]. Quantitative analysis of ctDNA can aid in the assessment of tumor volume and could potentially support therapeutic decisions by providing a non-invasive method to monitor treatment efficacy and disease progression [116][117][118][119]. In addition, qualitative analysis of ctDNA, including the identification of specific mutations and hypomethylation patterns, helps to predict disease prognosis and response to therapies [24]. ...
... To date, numerous technological approaches have been used for the detection of miRs, highlighting the need for precise and reliable tests based on mathematical algorithms to improve their clinical viability. Future studies are needed to fully establish their role in tumor biology and their potential clinical applications [115,128,131]. ...
Article
Full-text available
Neuroendocrine neoplasms (NENs) are a heterogeneous group of neoplasms presenting unique challenges in diagnosis and management. Traditional markers such as chromogranin A (CgA), pancreatic polypeptide (PP), and neuron-specific enolase (NSE) have limitations in terms of specificity and sensitivity. Specific circulating markers such as serotonin and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) and various gastrointestinal hormones such as gastrin, glucagon, somatostatin, and vasoactive intestinal peptide (VIP) have a role in identifying functional NENs. Recent advances in molecular and biochemical markers, also accounting for novel genomic and proteomic markers, have significantly improved the landscape for the diagnosis and monitoring of NENs. This review discusses these developments, focusing on both traditional markers such as CgA and NSE, as well as specific hormones like gastrin, insulin, somatostatin, glucagon, and VIP. Additionally, it covers emerging genomic and proteomic markers that are shaping current research. The clinical applicability of these markers is highlighted, and their role in improving diagnostic accuracy, predicting surgical outcomes, and monitoring response to treatment is demonstrated. The review also highlights the need for further research, including validation of these markers in larger studies, development of standardized assays, and integration with imaging techniques. The evolving field of biochemical markers holds promise for improving patient outcomes in the treatment of NENs, although challenges in standardization and validation remain.
... Over the years, a multitude of NEN biomarkers have been investigated including immunohistochemical, radiological, and circulating markers, some of which are already implemented in daily clinical practice [7]. Despite the plethora of markers, it remains challenging in clinical practice to (i) accurately and timely diagnose patients, (ii) determine prognosis, and (iii) predict and monitor response to therapy, because of limitations in sensitivity, specificity, and accuracy indicating a great need for new markers [8,9]. Therefore, in recent years, more and more research is conducted into more complex, omics-based biomarkers in liquid biopsies that allow real-time monitoring of tumor evolution [10, 11, 12•]. ...
... Another general NEN marker is the neuron-specific enolase (NSE), a glycolytic enzyme produced in neurons and neuroendocrine cells of the central and peripheral nervous system [47]. Elevated NSE levels are most often found in poorly differentiated NECs and have a negative prognostic value [8,32,54]. The diagnostic potential of this marker is rather low since NENs and non-NENs can only be distinguished with a sensitivity of 39-43% and specificity of 65-73% [15][16][17]. ...
Article
Full-text available
Opinion statement Given the considerable heterogeneity in neuroendocrine neoplasms (NENs), it appears unlikely that a sole biomarker exists capable of fully capturing all useful clinical aspects of these tumors. This is reflected in the abundant number of biomarkers presently available for the diagnosis, prognosis, and monitoring of NEN patients. Although assessment of immunohistochemical and radiological markers remains paramount and often obligatory, there has been a notable surge of interest in circulating biomarkers over the years given the numerous benefits associated with liquid biopsies. Currently, the clinic primarily relies on single-analyte assays such as the chromogranin A assay, but these are far from ideal because of limitations such as compromised sensitivity and specificity as well as a lack of standardization. Consequently, the quest for NEN biomarkers continued with the exploration of multianalyte markers, exemplified by the development of the NETest and ctDNA-based analysis. Here, an extensive panel of markers is simultaneously evaluated to identify distinct signatures that could enhance the accuracy of patient diagnosis, prognosis determination, and response to therapy prediction and monitoring. Given the promising results, the development and implementation of these multianalyte markers are expected to usher in a new era of NEN biomarkers in the clinic. In this review, we will outline both clinically implemented and more experimental circulating markers to provide an update on developments in this rapidly evolving field.
... In cancer, miRNA promotes angiogenesis, cell metabolism and metastases. Although widely studied in other tumours, such as ovarian, cervical and breast cancers, there are very few data regarding GEP-NENs [10,16]. One study concluded that a combination of four types of miRNA (miR-125b-5p, miR-362-5p, miR-425-5p and miR-500a-5p) was able to distinguish SI-NETs from healthy individuals [17]. ...
Article
Full-text available
Neuroendocrine neoplasms incidence has been increasing, arising the need for precise and early diagnostic tools. Liquid biopsy (LB) offers a less invasive alternative to tissue biopsy, providing real-time molecular information from circulating tumour components in body fluids. The aim of this review is to analyse the current evidence concerning LB in NENs and its role in clinical practice. We conducted a systematic review in July 2024 focusing on LB applications in NENs, including circulating tumour cells (CTCs), circulating tumour DNA (ctDNA), micro RNA (miRNA), messenger RNA (mRNA) and extracellular vesicles. Sixty-five relevant articles were analysed. The LB showed potential in diagnosing and monitoring NENs. While CTCs face limitations due to low shedding, ctDNA provides valuable information on high-grade neoplasms. MiRNA and mRNA (e.g., the NETest) offer high sensitivity and specificity for diagnosis and prognosis, outperforming traditional markers like chromogranin A. The LB has significant potential for NEN diagnosis and monitoring but lacks widespread clinical integration due to limited prospective studies and guidelines, requiring further validation. Advances in sequencing technologies may enhance the clinical utility of LB in NENs. Future research should focus on refining LB methods, standardising protocols and exploring applications in high-grade NENs.
Article
Full-text available
Introduction: Clinical presentation and genetic profile of gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) are highly variable, hampering their management. Sequencing of circulating tumor DNA (ctDNA) from liquid biopsy (LB) has been proposed as a less invasive alternative to solid biopsy (SB). Our aim is to compare the mutational profile (MP) provided by LB with that deriving from SB in GEP-NETs. Methods: SB and LB derived simultaneously from 6 GEP-NETs patients. A comparative targeted Next Generation Sequencing (NGS) analysis was performed on DNA from SB and LB to evaluate the mutational status of 11 genes (MEN1, DAXX, ATRX, MUTYH, SETD2, DEPDC5, TSC2, ARID1A, CHECK2, MTOR, PTEN). Results: Patients (M:F =2:1; median age 64 yrs) included 3 with pancreatic and 3 with ileal NETs. NGS detected a median number of 55 variants/sample in SB and 66.5 variants/sample in LB specimens (mutational burden: 0.2-1.9 and 0.3-1.8 mut/Mb, respectively). Missense and nonsense mutations were prevalent in both, mainly represented by C>T transitions. ARID1A, MTOR, and ATRX were consistently mutated in SB and ARID1A, TSC2, MEN1, PTEN, SETD2, and MUTYH were consistently mutated in LB. DAXX mutations were absent in LB. 17 recurrent mutations were shared between SB and LB; in particular, MTOR single nucleotide variants (SNVs) c.G4731A and c.C2997T were shared by 5 out of 6 patients. Hierarchical clustering supported genetic similarity between SB and LB. Conclusions: This pilot study explores the applicability of LB in GEP-NETs MP evaluation. Further studies with larger cohorts are needed to validate LB and to define the clinical impact.
Article
Full-text available
Key Clinical Message Understand the importance of considering alternative diagnosis in patients presenting with atypical features, specially when they are not responding to the standard treatment. Understand the importance of considering common presentations of rare cases. Underscoring the critical importance of timely recognition and appropriate management of potentially life‐threatening conditions.
Preprint
Full-text available
Besides medullary thyroid cancer, a latent and relatively mild increase of serum procalcitonin (PCT) has been found in neuroendocrine neoplasms (NENs). Here we are aiming to supply more information about NENs related PCT elevation, in order to broaden the clinical experiences about diagnosis and treatment of shock in tumor patients. we reported an advanced pancreatic neuroendocrine carcinoma (pNEC) with liver and lung metastasis, in which a rare pseudo-sepsis shock with extremely high serum PCT level (exceeding 100 ng/ml) had been demonstrated. A series of screening tests to exclude bacterial infections, including blood culture, urine culture and even metagenomic NGS (mNGS), had been performed. Given negative evidence of bacterial infection and useless broad-spectrum antibiotics treatment, steroid was used to relieve the serious inflammation and its related shock. The patient’s condition was improved and discharged. In summary, despite the ubiquitous use of PCT used in bacterial infection and sepsis shock, pNEC could cause the high level of serum PCT and even result in severe inflammation accompanied by shock. As for diagnosis and treatment strategies, pNEC should be regarded as one of rare differential diagnosis when experimental antibiotics are not working. The potential mechanism of PCT elevation and its role in prognosis of pNEC still needed to be further studied.
Article
Carcinoid heart disease (CHD) is a serious cardiac condition which is caused by elevated serotonin in the systemic circulation, secreted by neuroendocrine tumours (NET). It mostly affects the right‐sided heart valves, where it causes fibrotic disturbances and is associated with worse survival. With this study, we describe a large cohort of patients with CHD and provide insight in the survival over the past decades. All consecutive patients with a serotonin producing NET and CHD referred to the Netherlands Cancer Institute that presented with CHD or developed CHD during their follow up time were included from 1984 until 2021. Patients were divided in three time periods: 1984‐2000, 2000‐2010 and 2010‐2018. Median N‐terminal pro B‐type natriuretic protein (NT‐proBNP) and serum serotonin levels were stratified according to tricuspid regurgitation severity. Kaplan‐Meier curves and logrank test were used for visualisation of survival. Cox regression was used for identification of characteristics associated with disease specific mortality (DSM). A total of 84 patients with CHD were included of whom 49 (58.3%) were male. Median age at NET diagnosis was 62.3 (range 23.9‐81.7) years, and median time to development of CHD was 1.1 (range 0‐24.2) years. NT‐proBNP was significantly higher when more severe TR was present (p=0.027). Median survival from CHD diagnosis for 1984‐2000, 200‐2010 and 2010‐2018 were 1.3 (confidence interval [CI] 0.9‐1.6), 1.9 (CI 1.2‐2.6) and 3.9 (CI 1.7‐6.2) years (p=0.025). Valve replacement surgery (VSR) occurred more frequent in later time periods. VSR (hazard ratio [HR] 0.33, p=0.005) and NT‐proBNP (HR 1.003, 1.00‐1.005, p=0.036) were significantly associated with DSM. The prognosis of patients with CHD has improved over the past decades, possibly caused by more VSR. NT‐proBNP is a valuable biomarker in patients with CHD. Clinical practice should be aimed at timely diagnosis and intervention of CHD.
Article
Full-text available
Background: Metastases in eye structures are rare (1-5% cases at systemic spread of different malignancies, mainly breast and lung cancers). The prognosis is poor. The overall survival usually does not exceed 12 or even 6 months. If metastases are found in the choroid membrane, the probability that the patient has multiple metastatic lesions of other organs increases significantly. Lung neuroendocrine neoplasms are rare (1-2% of all malignancies in adults), but mainly aggressive tumors. They are cha-racterized by "blurred", nonspecific clinical symptoms, the correct diagnosis is delayed seriously, and distant metastases are seen in more than 40% of patients (usually in chest structures, liver, bones, brain, and adrenal glands; metastasis to vascular membrane of the eye ranks the 6th place). Case report: Own clinical observation of a male patient with rare metastasis of lung neuroendocrine carcinoma to the choroid of the left eye is presented. The disease is manifested by an ocular metastasis, which was initially considered an embryonic tumor. Other metastatic lesions (hilar lymph nodes, liver, soft tissues) were detected on computed tomography a little bit later. The diagnostic algorithm using routine histological examination and immunohistochemistry, including detection of neuroendocrine markers (chromogranin A, synaptophysin), cytokeratin 7 and Ki-67 expression in primary and metastatic tumors is described.
Article
We studied the content of neuron-specific enolase (NSE) in 69 paired samples of blood serum and seminal plasma from men with azoospermia (n=11) and oligoastenozoospermia (n=10) and from men with fertile ejaculate (n=48). NSE concentration was determined by ELISA (Vector-Best kit). The median concentration and the interquartile range of the NSE content in seminal plasma were 65.7 (47.9; 83.4) ng/ml and 24.33 times (р<0.000001) exceeded those for blood serum 2.7 (1.45; 4.0) ng/ml. There were no differences in the content of NSE between the groups for both seminal plasma and blood serum. The content of NSE in seminal plasma did not correlate with the content of NSE in blood serum, and also did not depend on the content of spermatozoa. A weak negative correlation (r=-0.341; p=0.0057) was found between the age of the examinees and the level of NSE in seminal plasma, but not in blood serum.
Article
Full-text available
Metastatic and locally-advanced neuroendocrine neoplasms (aNEN) form clinically and genetically heterogeneous malignancies, characterized by distinct prognoses based upon primary tumor localization, functionality, grade, proliferation index and diverse outcomes to treatment. Here, we report the mutational landscape of 85 whole-genome sequenced aNEN. This landscape reveals distinct genomic subpopulations of aNEN based on primary locali-zation and differentiation grade; we observe relatively high tumor mutational burdens (TMB) in neuroendocrine carcinoma (average 5.45 somatic mutations per megabase) with TP53, KRAS, RB1, CSMD3, APC, CSMD1, LRATD2, TRRAP and MYC as major drivers versus an overall low TMB in neuroendocrine tumors (1.09). Furthermore, we observe distinct drivers which are enriched in somatic aberrations in pancreatic (MEN1, ATRX, DAXX, DMD and CREBBP) and midgut-derived neuroendocrine tumors (CDKN1B). Finally, 49% of aNEN patients reveal potential therapeutic targets based upon actionable (and responsive) somatic aberrations within their genome; potentially directing improvements in aNEN treatment strategies.
Article
Full-text available
Purpose of Review Alternative lengthening of telomeres (ALT) is a telomerase-independent mechanism used by some types of malignancies, including pancreatic neuroendocrine tumors, to overcome the issue of telomere shortening, thus supporting tumor growth and cell proliferation. This review is focused on the most important achievements and opportunities deriving from ALT assessment in PanNET onco-pathology, highlighting the most promising fields in which such biomarker could be implemented in clinical practice. Recent Findings In pancreatic neuroendocrine tumors (PanNET), ALT is strongly correlated with the mutational status of two chromatin remodeling genes, DAXX and ATRX . Recent advances in tumor biology permitted to uncover important roles of ALT in the landscape of PanNET, potentially relevant for introducing this biomarker into clinical practice. Indeed, ALT emerged as a reliable indicator of worse prognosis for PanNET, helping in clinical stratification and identification of “high-risk” patients. Furthermore, it is a very specific marker supporting the pancreatic origin of neuroendocrine neoplasms and can be used for improving the diagnostic workflow of patients presenting with neuroendocrine metastasis from unknown primary. The activation of this process can be determined by specific FISH analysis. Summary ALT should be introduced in clinical practice for identifying “high-risk” PanNET patients and improving their clinical management, and as a marker of pancreatic origin among neuroendocrine tumors.
Article
Full-text available
Objective Recent studies have found aristaless-related homeobox gene (ARX)/pancreatic and duodenal homeobox 1 (PDX1), alpha-thalassemia/mental retardation X-linked (ATRX)/death domain-associated protein (DAXX) and alternative lengthening of telomeres (ALT) to be promising prognostic biomarkers for non-functional pancreatic neuroendocrine tumours (NF-PanNETs). However, they have not been comprehensively evaluated, especially among small NF-PanNETs (≤2.0 cm). Moreover, their status in neuroendocrine tumours (NETs) from other sites remains unknown. Design An international cohort of 1322 NETs was evaluated by immunolabelling for ARX/PDX1 and ATRX/DAXX, and telomere-specific fluorescence in situ hybridisation for ALT. This cohort included 561 primary NF-PanNETs, 107 NF-PanNET metastases and 654 primary, non-pancreatic non-functional NETs and NET metastases. The results were correlated with numerous clinicopathological features including relapse-free survival (RFS). Results ATRX/DAXX loss and ALT were associated with several adverse prognostic findings and distant metastasis/recurrence (p<0.001). The 5-year RFS rates for patients with ATRX/DAXX-negative and ALT-positive NF-PanNETs were 40% and 42% as compared with 85% and 86% for wild-type NF-PanNETs (p<0.001 and p<0.001). Shorter 5-year RFS rates for ≤2.0 cm NF-PanNETs patients were also seen with ATRX/DAXX loss (65% vs 92%, p=0.003) and ALT (60% vs 93%, p<0.001). By multivariate analysis, ATRX/DAXX and ALT status were independent prognostic factors for RFS. Conversely, classifying NF-PanNETs by ARX/PDX1 expression did not independently correlate with RFS. Except for 4% of pulmonary carcinoids, ATRX/DAXX loss and ALT were only identified in primary (25% and 29%) and NF-PanNET metastases (62% and 71%). Conclusions ATRX/DAXX and ALT should be considered in the prognostic evaluation of NF-PanNETs including ≤2.0 cm tumours, and are highly specific for pancreatic origin among NET metastases of unknown primary.
Article
Full-text available
Background: Plasma chromogranin A (CgA) is related to tumor burden and recommended in the follow-up of patients diagnosed with neuroendocrine tumors (NETs). The use of CgA in the workup of a suspected NET is more questionable. Objective: To assess the positive predictive value (PPV) of CgA plasma concentrations above the upper reference limit (URL) in patients with suspected NET. Method: Patients referred to the NET Centre, Rigshospitalet, Copenhagen from 2015 to 2019 with clinically suspected NET were included if a CgA measurement was performed prior to referral. The utility of CgA was assessed by comparing pre-referral CgA concentrations to the outcome of a thorough workup. In 47 selected cases with continuously unexplained elevated CgA concentrations, a processing-independent analysis (PIA) for CgA was performed. Results: A total of 197 patients were included. NET was ultimately diagnosed in 25 patients. CgA plasma concentrations were above the URL (elevated) in 19/25 patients diagnosed with NET. In total, 167/197 had elevated CgA concentrations at referral. The positive predictive value (PPV) of elevated CgA concentration was 11% (19/167). Proton pump inhibitor (PPI) treatment was identified as the possible cause of CgA elevation in 55/148 patients with falsely elevated CgA. CgA concentration was normal in 28/47 patients when using PIA. Conclusion: Our data do not support using measurement of CgA for screening when NET is suspected since the PPV was rather low. PPI treatment is a common cause of increased CgA concentrations and should always be discontinued before CgA measurement. PIA of CgA could be a way of excluding NET when suspicion is based primarily on elevated CgA.
Article
Full-text available
Identified in 1973, somatostatin (SST) is a cyclic hormone peptide with a short biological half-life. Somatostatin receptors (SSTRs) are widely expressed in the whole body, with five subtypes described. The interaction between SST and its receptors leads to the internalization of the ligand-receptor complex and triggers different cellular signaling pathways. Interestingly, the expression of SSTRs is significantly enhanced in many solid tumors, especially gastro-entero-pancreatic neuroendocrine tumors (GEP-NET). Thus, somatostatin analogs (SSAs) have been developed to improve the stability of the endogenous ligand and so extend its half-life. Radiolabeled analogs have been developed with several radioelements such as indium-111, technetium-99 m, and recently gallium-68, fluorine-18, and copper-64, to visualize the distribution of receptor overexpression in tumors. Internal metabolic radiotherapy is also used as a therapeutic strategy (e.g., using yttrium-90, lutetium-177, and actinium-225). With some radiopharmaceuticals now used in clinical practice, somatostatin analogs developed for imaging and therapy are an example of the concept of personalized medicine with a theranostic approach. Here, we review the development of these analogs, from the well-established and authorized ones to the most recently developed radiotracers, which have better pharmacokinetic properties and demonstrate increased efficacy and safety, as well as the search for new clinical indications.
Article
Nuclear medicine plays an increasingly important role in the management neuroendocrine neoplasms (NEN). Somatostatin analogue (SSA)-based positron emission tomography/computed tomography (PET/CT) and peptide receptor radionuclide therapy (PRRT) have been used in clinical trials and approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA). European Association of Nuclear Medicine (EANM) Focus 3 performed a multidisciplinary Delphi process to deliver a balanced perspective on molecular imaging and radionuclide therapy in well-differentiated neuroendocrine tumours (NETs). NETs form in cells that interact with the nervous system or in glands that produce hormones. These cells, called neuroendocrine cells, can be found throughout the body, but NETs are most often found in the abdomen, especially in the gastrointestinal tract. These tumours may also be found in the lungs, pancreas and adrenal glands. In addition to being rare, NETs are also complex and may be difficult to diagnose. Most NETs are non-functioning; however, a minority present with symptoms related to hypersecretion of bioactive compounds. NETs often do not cause symptoms early in the disease process. When diagnosed, substantial number of patients are already found to have metastatic disease. Several societies' guidelines address Neuroendocrine neoplasms (NENs) management; however, many issues are still debated, due to both the difficulty in acquiring strong clinical evidence in a rare and heterogeneous disease and the different availability of diagnostic and therapeutic options across countries. EANM Focus 3 reached consensus on employing ⁶⁸gallium-labelled somatostatin analogue ([⁶⁸Ga]Ga-DOTA-SSA)-based PET/CT with diagnostic CT or magnetic resonance imaging (MRI) for unknown primary NET detection, metastatic NET, NET staging/restaging, suspected extra-adrenal pheochromocytoma/paraganglioma and suspected paraganglioma. Consensus was reached on employing ¹⁸fluorine-fluoro-2-deoxyglucose ([¹⁸F]FDG) PET/CT in neuroendocrine carcinoma, G3 NET and in G1-2 NET with mismatched lesions (CT-positive/[⁶⁸Ga]Ga-DOTA-SSA-negative). Peptide receptor radionuclide therapy (PRRT) was recommended for second line treatment for gastrointestinal NET with [⁶⁸Ga]Ga-DOTA-SSA uptake in all lesions, in G1/G2 NET at disease progression, and in a subset of G3 NET provided all lesions are positive at [¹⁸F]FDG and [⁶⁸Ga]Ga-DOTA-SSA. PRRT rechallenge may be used for in patients with stable disease for at least 1 year after therapy completion. An international consensus is not only a prelude to a more standardised management across countries but also serves as a guide for the direction to follow when designing new research studies.
Article
The incidence of pancreatic neuroendocrine neoplasms (PanNENs) has gradually increased. PanNENs comprise two subtypes with different clinical manifestations and molecular mechanisms: functional PanNENs and nonfunctional PanNENs. Excessive hormones and tumor progression severely affect the quality of life of patients or are even life threatening. However, the molecular mechanisms of hormone secretion and tumor progression in PanNENs have not yet been fully elucidated. At present, advancements in sequencing technologies have led to the exploration of new biological markers and an advanced understanding of molecular mechanisms in PanNENs. Multiomics sequencing could reveal differences and similarities in molecular features in different fields. However, sequencing studies of PanNENs are booming and should be summarized to integrate the current findings. In this review, we summarize the current status of multiomics sequencing in PanNENs to further guide its application. We explore mainly advancements in the genome, transcriptome, and DNA methylation fields. In addition, the cell origin of PanNENs, which has been a hot issue in sequencing research, is described in multiple fields.
Article
Neuroendocrine neoplasia is described in almost every tissue, either in the pure endocrine organs, the nerve structures or in the so-called diffuse neuroendocrine system. The current nomenclature contains time-honored widely accepted definitions; however, it is different according to anatomical sites. Diverse definitions may generate confusion and non-standard patient management. The International Agency for Research on Cancer – World Health Organization (IARC-WHO) proposed a framework for universal classification of neuroendocrine neoplasia. Evidence indicates that neuroendocrine cancer is composed by cells with a distinctive phenotype characterized by the expression of general and specific neuroendocrine markers. The neuroendocrine phenotype is indicated as descriptor of a unique cancer category, now recommended for all organs as Neuroendocrine Neoplasm. Evidence indicates that neuroendocrine neoplasia may be well or poorly differentiated, with diverse incidence and prevalence in different organs. It is proposed that the well differentiated neoplasm is universally defined neuroendocrine tumor (NET), and the poorly differentiated neoplasm neuroendocrine carcinoma (NEC). Evidence indicates that a cancer grading tool based on a proliferation measure by mitotic count, Ki67 % and/or necrosis assessment is useful to predict NET patient behavior. It is proposed to utilize this tool for grading NET universally, with site-specific cut-offs to be defined. It is also acknowledged that significant biological site-specific differences exist. It is recommended that current pathology reports contain this classification together with the current traditional classifiers. This IARC-WHO common classification framework for neuroendocrine neoplasm aims at uniformizing nomenclature toward different organs and at fostering the definition of a similar site-specific gene signature.