ArticlePDF Available

JNETS clinical practice guidelines for gastroenteropancreatic neuroendocrine neoplasms: diagnosis, treatment, and follow-up: a synopsis

Authors:

Abstract and Figures

Neuroendocrine neoplasms (NENs) are rare neoplasms that occur in various organs and present with diverse clinical manifestations. Pathological classification is important in the diagnosis of NENs. Treatment strategies must be selected according to the status of differentiation and malignancy by accurately determining whether the neoplasm is functioning or nonfunctioning, degree of disease progression, and presence of metastasis. The newly revised Clinical Practice Guidelines for Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs) comprises 5 chapters—diagnosis, pathology, surgical treatment, medical and multidisciplinary treatment, and multiple endocrine neoplasia type 1 (MEN1)/von Hippel–Lindau (VHL) disease—and includes 51 clinical questions and 19 columns. These guidelines aim to provide direction and practical clinical content for the management of GEP-NEN preferentially based on clinically useful reports. These revised guidelines also refer to the new concept of “neuroendocrine tumor” (NET) grade 3, which is based on the 2017 and 2019 WHO criteria; this includes health insurance coverage of somatostatin receptor scintigraphy for NEN, everolimus for lung and gastrointestinal NET, and lanreotide for GEP-NET. The guidelines also newly refer to the diagnosis, treatment, and surveillance of NEN associated with VHL disease and MEN1. The accuracy of these guidelines has been improved by examining and adopting new evidence obtained after the first edition was published.
Content may be subject to copyright.
ORIGINAL ARTICLE—LIVER, PANCREAS, AND BILIARY TRACT
JNETS clinical practice guidelines for gastroenteropancreatic
neuroendocrine neoplasms: diagnosis, treatment, and follow-up:
a synopsis
Tetsuhide Ito
1,2
Toshihiko Masui
1,2
Izumi Komoto
1,2
Ryuichiro Doi
1,2
Robert Y. Osamura
1,2
Akihiro Sakurai
1,2
Masafumi Ikeda
1,2
Koji Takano
1,2
Hisato Igarashi
1,2
Akira Shimatsu
1,2
Kazuhiko Nakamura
1,2
Yuji Nakamoto
1,2
Susumu Hijioka
1,2
Koji Morita
1,2
Yuichi Ishikawa
1,2
Nobuyuki Ohike
1,2
Atsuko Kasajima
1,2
Ryoji Kushima
1,2
Motohiro Kojima
1,2
Hironobu Sasano
1,2
Satoshi Hirano
1,2
Nobumasa Mizuno
1,2
Taku Aoki
1,2
Takeshi Aoki
1,2
Takao Ohtsuka
1,2
Tomoyuki Okumura
1,2
Yasutoshi Kimura
1,2
Atsushi Kudo
1,2
Tsuyoshi Konishi
1,2
Ippei Matsumoto
1,2
Noritoshi Kobayashi
1,2
Nao Fujimori
1,2
Yoshitaka Honma
1,2
Chigusa Morizane
1,2
Shinya Uchino
1,2
Kiyomi Horiuchi
1,2
Masanori Yamasaki
1,2
Jun Matsubayashi
1,2
Yuichi Sato
1,2
Masau Sekiguchi
1,2
Shinichi Abe
1,2
Takuji Okusaka
1,2
Mitsuhiro Kida
1,2
Wataru Kimura
1,2
Masao Tanaka
1,2
Yoshiyuki Majima
1,2
Robert T. Jensen
1,2
Koichi Hirata
1,2
Masayuki Imamura
1,2
Shinji Uemoto
1,2
Received: 31 May 2021 / Accepted: 13 September 2021
ÓThe Author(s) 2021
Abstract Neuroendocrine neoplasms (NENs) are rare
neoplasms that occur in various organs and present with
diverse clinical manifestations. Pathological classification
is important in the diagnosis of NENs. Treatment strategies
must be selected according to the status of differentiation
and malignancy by accurately determining whether the
neoplasm is functioning or nonfunctioning, degree of dis-
ease progression, and presence of metastasis. The newly
revised Clinical Practice Guidelines for Gastroenteropan-
creatic Neuroendocrine Neoplasms (GEP-NENs) com-
prises 5 chapters—diagnosis, pathology, surgical
treatment, medical and multidisciplinary treatment, and
multiple endocrine neoplasia type 1 (MEN1)/von Hippel–
Lindau (VHL) disease—and includes 51 clinical questions
and 19 columns. These guidelines aim to provide direction
and practical clinical content for the management of GEP-
NEN preferentially based on clinically useful reports.
These revised guidelines also refer to the new concept of
‘neuroendocrine tumor’’ (NET) grade 3, which is based on
the 2017 and 2019 WHO criteria; this includes health
The original version of this article appeared in Japanese as ‘‘Sui-
Syoukakan Shinkeinaibunpitsushuyo Shinryo Guideline’’ from the
Japanese Neuroendocrine Tumor Society (JNETS), published by
Kanehara-Shuppan, Tokyo, 2019. Please see the article on the
standards, methods, and process of developing the guidelines
(ISBN978/4-307-20401-9).
Tetsuhide Ito, Toshihiko Masui, Izumi Komoto, Ryuichiro Doi,
Robert Y. Osamura, Akihiro Sakurai, Masafumi Ikeda, Koji Takano,
Hisato Igarashi, Akira Shimatsu, Kazuhiko Nakamura, Yuji
Nakamoto, Susumu Hijioka, Koji Morita, Yuichi Ishikawa, Nobuyuki
Ohike, Atsuko Kasajima, Ryoji Kushima, Motohiro Kojima,
Hironobu Sasano, Satoshi Hirano, Nobumasa Mizuno, Taku Aoki,
Takeshi Aoki, Takao Ohtsuka, Tomoyuki Okumura, Yasutoshi
Kimura, Atsushi Kudo, Tsuyoshi Konishi, Ippei Matsumoto,
Noritoshi Kobayashi, Nao Fujimori, Yoshitaka Honma, Chigusa
Morizane, Shinya Uchino, Kiyomi Horiuchi, Masanori Yamasaki,
Yuichi Sato, Masau Sekiguchi, Takuji Okusaka, Mitsuhiro Kida,
Wataru Kimura, Masao Tanaka, Yoshiyuki Majima, Robert T. Jensen,
Koichi Hirata, Masayuki Imamura and Shinji Uemoto: Guidelines
Committee for creating and evaluating the ‘‘Clinical practice
guidelines for Gastroenteropancreatic Neuroendocrine Neoplasms
(GEP-NEN)2019’’, The Japanese Neuroendocrine Tumor Society
(JNETS), 54 Kawaramachi, Shougoin, Sakyouku, Kyoto 104-0061,
Japan.
The members of the Guidelines Committee are listed in the Appendix
in the text.
&Tetsuhide Ito
itopapa@kouhoukai.or.jp
1
Neuroendocrine Tumor Centre, Fukuoka Sanno Hospital, 3-
6-45 Momochihama, Sawara-ku, Fukuoka 814-0001, Japan
2
Department of Gastroenterology, Graduate School of Medical
Sciences, Internal University of Health and Welfare, 3-6-45
Momochihama, Sawara-ku, Fukuoka 814-0001, Japan
123
J Gastroenterol
https://doi.org/10.1007/s00535-021-01827-7
insurance coverage of somatostatin receptor scintigraphy
for NEN, everolimus for lung and gastrointestinal NET,
and lanreotide for GEP-NET. The guidelines also newly
refer to the diagnosis, treatment, and surveillance of NEN
associated with VHL disease and MEN1. The accuracy of
these guidelines has been improved by examining and
adopting new evidence obtained after the first edition was
published.
Keywords Clinical practice guideline
Gastroenteropancreatic neuroendocrine neoplasm
Japanese Neuroendocrine Tumor Society
Introduction
To standardize the diagnosis and treatment of gastroen-
teropancreatic neuroendocrine neoplasms (GEP-NENs) in
Japan, the Japanese Neuroendocrine Tumor Society
(JNETS) published the first Clinical Practice Guidelines for
Gastroenteropancreatic Neuroendocrine Neoplasms in
Japan in 2015 [1]. However, several subsequent develop-
ments regarding neuroendocrine neoplasms (NENs)
necessitate the revision of clinical practice guidelines. The
Guidelines Revision Committee was established at JNETS
and began working on the revised guidelines in January
2018.
After making updates to reflect the assessment com-
mittee members’ suggestions, public hearings were held at
various academic societies starting April 2019, culminating
in the publication of the second edition in September 2019
[2]. This revised edition encompasses diagnosis, pathology,
surgical treatment, medical and multidisciplinary treat-
ment, and multiple endocrine neoplasia type 1 (MEN1)/von
Hippel–Lindau (VHL) disease and includes 51 clinical
questions and 19 columns. Topics under exploration are
introduced in the ‘‘Columns’’ in the guideline based on
expert consensus and evidence.
As a new development in the treatment of NEN in
Japan, somatostatin receptor scintigraphy (SRS) [3,4] was
approved for insurance coverage in 2015 for the general
diagnosis of NEN. In addition, the WHO classification for
GEP-NENs was revised in 2017 and 2019, adding the new
grade 3 (G3) well-differentiated neuroendocrine tumors
(NETs), which are characterized by well-differentiated
tissue and a Ki-67 index exceeding 20% [5,6]. Regarding
surgical treatment, the first edition of the guidelines did not
include explicit recommendations for nonfunctioning pan-
creatic NETs 1–2 cm in size. However, the revised edition
specifies indications for surgery and recommends surgical
approaches for well-differentiated nonfunctioning pancre-
atic NETs and specifically covers the management of small
pancreatic NETs from a broad perspective. The revised
guidelines also newly include indications for surgery for
poorly differentiated pancreatic neuroendocrine carcino-
mas (NECs). Regarding drug therapy, the molecular tar-
geted drug everolimus is now covered by insurance for the
treatment of well-differentiated NENs of the lungs and
gastrointestinal tract [7]. In addition, the somatostatin
analogue lanreotide is now covered by insurance for the
treatment of well-differentiated GEP-NENs [810], sub-
stantially broadening treatment options. Moreover, while
the first edition covered the diagnosis and treatment of
NENs associated with MEN1, the revised edition also
covers pancreatic NENs associated with VHL disease [11].
In this article, we explain the changes described above in
the sequential order of the 5 chapters of the guidelines.
Diagnosis
A GEP-NEN can be functioning or nonfunctioning. A Ja-
panese epidemiological study reports that approximately
35% of pancreatic NETs are functioning, indicating that
most pancreatic NETs are nonfunctioning [12]. Meanwhile,
approximately 1% of gastrointestinal NETs present with
carcinoid syndrome, which differs considerably from the
trends in Europe and the U.S. [12]. This is likely because
hindgut NENs are more prevalent in Japan, while midgut
NENs, which have higher rates of carcinoid syndrome
complication, are more prevalent in Europe and the U.S.
[12,13].
Functional NENs are often diagnosed on the basis of
endocrine symptoms due to hormonal hypersecretion.
Insulinoma primarily presents with fasting hypoglycemia
episodes and includes autonomic and neurologic symp-
toms. In cases in which hypoglycemic symptoms are
unrecognized, symptoms such as seizures and dementia
may be the earliest symptoms [14,15]. Testing such as a
72-h fasting test and a mixed-meal test are recommended
for definitive diagnosis [16], although there are recent
reports of a 48-h fasting test combined with a glucagon test
[17]. Symptoms of gastrinoma include peptic ulcer and
reflux esophagitis due to gastric hypersecretion and diar-
rhea due to pancreatic enzyme inactivation [18]. Mea-
surement of fasting serum gastrin level and gastric acid pH
are required for definitive diagnosis, while a calcium
infusion test is useful [19,20]. Determination of MEN1
complication is also recommended [21,22]. Symptoms and
recommended tests for functional NENs including other
relatively rare neoplasms are shown in Table 1.
Nonfunctional pancreatic NENs have no specific
symptoms and may present with jaundice, pancreatitis,
bloating, abdominal pain, or intestinal obstruction symp-
toms associated with tumor growth. Advanced cases are
often identified owing to the detection of distant metastases
123
J Gastroenterol
[13]. Pathological diagnosis such as histology and cytology
is recommended for differential diagnosis.
To determine the localization of pancreatic NENs, it is
recommended to consider and perform imaging such as
ultrasonography (US), computed tomography (CT), mag-
netic resonance imaging (MRI), endoscopic ultrasonogra-
phy (EUS), or SRS on a case-by-case basis. When
performing histology, endoscopic ultrasound-guided fine-
needle aspiration (EUS-FNA) is recommended [3,23]. If
microscopic insulinomas and gastrinomas cannot be
localized by imaging, then selective arterial secretagogue
injection (SASI) test is useful [24,25].
Endoscopic findings of gastrointestinal NETs are round,
submucosal, tumor-like protrusions, which when grown are
accompanied by central depression and ulceration [26].
Meanwhile, gastrointestinal NECs often appear as
advanced cancer. The next recommended tests are endo-
scopic biopsy, EUS-FNA, and imaging to rule out distant
metastases [27].
When testing for metastases, US, CT, MRI, positron
emission tomography (PET) with 18F-fluorodeoxyglucose
(FDG), or SRS should be performed as appropriate. For
liver metastases, the rate of detection by US can be
improved by the use of contrast media, and multiphasic
imaging using contrast media is recommended for CT [28].
Contrast-enhanced MRI using Gd-DOTA yields higher
detectability than CT or SRS [29]. Although only some
well-differentiated NETs are positive on FDG-PET, FDG-
PET is useful for finding metastases and recurrent lesions
of tumors with high proliferative potential, such as NECs;
this method is inversely correlated with and complemen-
tary to SRS. Although the sensitivity of SRS is not nec-
essarily high at 52%, it has a high specificity of 93% [30].
Histopathology
The WHO Classification of Endocrine Organs (2017) [5]
and Digestive System (2019) [6] categorizes NENs as well-
differentiated NENs (termed ‘‘NETs’’) or poorly differen-
tiated NECs. NENs are graded according to morphology
(i.e., well or poorly differentiated) and proliferative
Table 1 Symptoms and recommended tests for functional NENs
Functional
neuroendocrine
neoplasm
Symptoms and findings Differential diagnosis (presence diagnosis)
Insulinoma Central nervous system symptoms: impaired consciousness
(67–80%), abnormal vision (42–59%), amnesia (47%),
personality changes (16–38%), epilepsy (16–17%),
headache (7%)
Autonomic symptoms: sweating (30–69%), malaise
(28–56%), hyperphagia/obesity (14–50%), tremor
(12–14%), palpitation (5–12%), anxiety (12%)
Differentiating hypoglycemia: Whipple’s triad, exogenous
insulin, oral hypoglycemic agents, endogenous insulin
dyssecretion, insulin autoimmune syndrome
Definitive diagnosis: 72-h fasting test, mixed-meal test, 48-h
fasting test ?glucagon tolerance test
Gastrinoma Peptic ulcers: duodenal bulb (75%), distal duodenum (14%),
jejunum (11%)
Abdominal pain, steatorrhea
Fasting serum gastrin measurement, gastric pH measurement,
intravenous calcium injection test (MEN1 differential
diagnostics: blood calcium measurement, intact PTH
measurement)
Glucagonoma Glucose intolerance/diabetes (30–90%), weight loss
(60–90%), necrotizing erythema migrans (55–90%),
mucosal symptoms (30–40%), diarrhea (10–15%), anemia
(30–90%), hypoaminoacidemia (30–100%), venous
thrombosis, psychoneurotic symptoms
Plasma glucagon measurement, serum albumin
measurement, amino acid fraction measurement
VIPoma Profuse watery diarrhea, hypokalemia, fatigue, muscle
weakness, shortness of breath, muscle cramps
Diarrhea: dark brown, odorless, low osmotic gap and
secretory
Stool osmotic gap measurement
Blood VIP cannot be measured in Japan
Somatostatinoma Weight loss, abdominal pain, diabetes, cholelithiasis,
steatorrhea, diarrhea, hypoacidity, anemia (often
asymptomatic)
Blood somatostatin cannot be measured in Japan Diagnosis
by biopsy
Carcinoid
syndrome
Skin flushing (without sweating), diarrhea, pellagra
symptoms, psychiatric symptoms (i.e., confusion), heart
failure (especially right heart failure), bronchospasm,
intra-abdominal fibrosis
Urinary 5-HIAA excretion measurement, intake of serotonin-
containing foods and drugs
MEN1, multiple endocrine neoplasia type 1
123
J Gastroenterol
activities (i.e., mitotic rate measured as mitoses/2 mm
2
or
Ki-67 index by counting 500 tumor cells in hotspots).
Well-differentiated NETs with proliferative activi-
ties \3%, 3–20%, and [20% are graded as G1, G2, and
G3, respectively. In contrast, poorly differentiated NECs
usually exhibit a higher Ki-67 index (i.e., [20%) and
lower expression of somatostatin receptor 2 (Table 2).
It can be difficult to distinguish NET G3 from NECs.
However, the pathology of NET G3 is essentially similar to
that of NET G1 and G2 in that it forms well-demarcated,
medullary, expansive, solid masses; grows relatively
slowly; and has a compact organoid structure (e.g., funic-
ular, alveolar, pseudoglandular, etc.) with neuroendocrine
differentiation on histology (Figure S1). Cytologic atypia
remains mild to moderate, and components corresponding
to NET G1 and G2 coexist inside the tumor. On the other
hand, NECs form poorly demarcated medullary masses and
grow rapidly. Histologically, highly atypical cells exhibit
large alveolar to sheet-like and diffuse proliferation with an
ill-defined organoid structure. NET G3 and NECs both
have Ki-67 indices [20%, but NECs usually have a Ki-67
index [50% often with extensive necrotic foci; mean-
while, NET G3 rarely has a Ki-67 index [50% or necrotic
foci. Somatostatin receptor expression is often positive in
NET G3 but weakly positive or negative in NECs. As
NECs exhibit p53 overexpression and deletion of Rb pro-
tein, which are characteristic of extremely malignant
tumors, immunostaining for these markers is helpful for
differentiating NET G3 and NECs [3,32,34].
The WHO 2010 classification includes a category for
mixed adeno-neuroendocrine carcinoma (MANEC) as
combined adenocarcinoma and NET, although this did not
include combinations such as acinar cell carcinoma and
NET. In the new classification, such combinations are
instead described as tumors involving both neuroendocrine
and non-neuroendocrine cells, and are classified as mixed
neuroendocrine–non-neuroendocrine neoplasm (MiNEN)
[6].
Surgical treatment
The surgical treatment of pancreatic NENs varies
depending on the type of tumor. Pancreatectomy with
lymphadenectomy is generally recommended for non-
functioning pancreatic NENs [34,35]. Meanwhile, for
incidentally discovered asymptomatic tumors \10 mm
with no evidence of metastasis/invasion (e.g., hepatic or
lymphatic involvement, pancreatic duct stenosis, and bil-
iary stricture) on imaging, follow-up every 6–12 months
may be an option with the patient’s informed consent
(Fig. 1)[3639]. Surgery is generally recommended for
insulinomas; when indicated, a minimally invasive
approach is preferable [40,41]. Pancreatectomy with
lymphadenectomy is recommended for malignant insuli-
nomas [42]. For gastrinomas, the high malignant potential
is assumed, and resection with lymphadenectomy is rec-
ommended. Gastrinomas with MEN1 exhibit metachronous
recurrences, so care must be taken to avoid excessive
surgery [43]. As rare functional pancreatic NENs other
than insulinomas and gastrinomas (e.g., glucagonomas,
VIPomas, somatostatinomas, GRFomas, PPomas,
ACTHomas, and PTHomas) are highly malignant, pan-
createctomy with lymphadenectomy is recommended [44].
When feasible, macroscopic curative resection is rec-
ommended for pancreatic NET G3 as for NET G1 and G2,
whereas the indications for surgery are unclear for pan-
creatic NECs [45].
Surgical treatment of gastrointestinal NENs varies by
organ. More than 90% of esophageal NENs are NECs.
Endoscopic resection or surgical resection is indicated for
NETs according to stage, and drug therapy is indicated for
nonresectable cases. Esophageal NECs are frequently
accompanied by lymph node metastases (30%) and distant
metastases (50%), requiring more careful decisions on
surgery than that for esophageal cancers [46,47]. For
gastric NENs, the decision on surgical indications and
selection of a surgical procedure according to Rindi’s
classification [48] is recommended. For small intestinal
NENs, small intestine resection with lymphadenectomy is
Table 2 WHO grading criteria
for GEP-NENs (2017/2019) Classification Differentiation Grade Ki-67 index Mitotic index (/2mm
2
)
NET G1
NET G2
Well differentiated Low
Intermediate
\3%
3–20%
\2
2–20
NET G3 High [20% [20
NEC
Small-cell type
Large-cell type
Poorly differentiated High [20% [20
MiNEN Well or poorly differentiated Variable Variable Variable
GEP-NEN, gastroenteropancreatic neuroendocrine neoplasm; MiNEN, mixed neuroendocrine-–non-neu-
roendocrine neoplasm; NEC, neuroendocrine carcinoma; NET, neuroendocrine tumor
123
J Gastroenterol
recommended when curative resection is feasible [49].
Surgery is indicated for all appendiceal NENs, and the
surgical approach should be selected by taking tumor
localization, tumor size, and the presence of risk factors
into account [50,51]. Endoscopic resection is often indi-
cated for colonic NENs, but colectomy with lym-
phadenectomy is recommended for the following: tumor
size C1 cm or G2 or higher; muscularis propria invasion;
suspected lymph node metastasis; or endoscopic resection
specimens indicative of vascular invasion, muscularis
propria invasion, positive surgical margins, or G2 or
higher. Proctectomy with lymphadenectomy or rectal
amputation is recommended for rectal NENs for the fol-
lowing: tumor size C1 cm or G2 or higher; muscularis
propria invasion; suspected local lymph node involvement;
or endoscopic resection specimens indicative of the need
for additional treatment (Fig. 2)[5254].
Medical and multidisciplinary treatment (Fig. 3)
Treatment modalities for GEP-NENs differ for NETs and
NECs, and treatment approaches differ for NETs origi-
nating in the pancreas and gastrointestinal tract (Table 3).
Resection is indicated for NETs when feasible; endoscopic
treatment can also be considered for gastrointestinal NETs.
Moreover, radiofrequency ablation and transarterial
chemoembolization are used for liver metastases. How-
ever, adjuvant chemotherapy to prevent recurrence of
NETs has not been established [5557]. For functional
NETs with hormonal symptoms, somatostatin analogues
such as octreotide and lanreotide are used to control
symptoms [5558]. For tumor control, somatostatin ana-
logues, molecular targeted drugs, and cytotoxic anticancer
agents are indicated [5557]. Regarding somatostatin
analogues, insurance covers lanreotide [8] for pancreatic
NETs as well as octreotide [59] and lanreotide [8] for
gastrointestinal NETs. Regarding molecular targeted drugs,
insurance covers everolimus [60] and sunitinib [61] for
pancreatic NETs as well as everolimus [7] for gastroin-
testinal NETs. Regarding cytotoxic anticancer agents,
Fig. 1 Surgical approach for nonfunctioning pancreatic NETs.
(Superscript a) Check the swelling and firmness of the regional
lymph nodes and dissect if lymph node metastases are suspected; if
the tumor is discovered incidentally and there is no radiographic
evidence of metastasis or invasion, follow-up may be an option with
adequate explanation. PanNEC, pancreatic neuroendocrine carci-
noma; PanNEN, pancreatic neuroendocrine neoplasm; PanNET,
pancreatic neuroendocrine tumor
123
J Gastroenterol
insurance covers streptozocin [62,63] for both pancreatic
and gastrointestinal NETs; in addition, temozolomide [64]
is considered promising in Europe and the U.S. Temo-
zolomide combination therapy is one of the useful treat-
ments, but it is not approved for insurance in Japan.
Furthermore, radiation therapy may be used for palliative
purposes for bone and brain metastases. Moreover,
radionuclide-labeling peptide therapy (PRRT) [65] is often
used in Europe and the U.S. PRRT has recently been
covered by insurance in Japan, but at present, it should be
given priority to patients who are ineffective with other
drugs after the second treatment and need immediate PRRT
treatment. For that purpose, it is considered necessary to
build a network with feasible facilities.
While there is no established strategy for selecting
appropriate treatment modalities, Japanese experts have
proposed guidelines for NETs originating in the pancreas
[66], although future validation is required.
For NECs, resection is indicated when feasible, and
adjuvant chemotherapy can be used to prevent recurrence
after surgery. Resection is not recommended for liver
metastases in NECs. In nonresectable cases, platinum-
based chemotherapy is indicated, such as etoposide/cis-
platin, irinotecan/cisplatin, and etoposide/carboplatin
[13]. However, no effective drug therapy has been
established for cases refractory to these therapies [13].
Although GEP-NENs are rare, there are effective local
therapies such as resection, radiofrequency ablation, and
transarterial chemoembolization [5557]. Several ran-
domized controlled trials have demonstrated the usefulness
of various drugs—many of which have been approved for
use. In practice, the multidisciplinary treatment that takes
advantage of these therapies is offered.
MEN1/VHL disease
Some pancreatic NENs develop in settings of hereditary
neoplasms, specifically MEN1 and VHL disease. Epi-
demiological studies report the frequencies of these
Fig. 2 Surgical approach for NENs of the colon and rectum. NEC, neuroendocrine carcinoma; NEN, neuroendocrine neoplasm; NET,
neuroendocrine tumor
123
J Gastroenterol
hereditary neoplasms among all pancreatic NENs, and
whole-genomic sequencing revealed that approximately
6% and 1% of pancreatic NENs carry germline mutations
in MEN1 and VHL, respectively [67], suggesting that the
actual frequency is within this range.
Pancreatic NENs in settings of hereditary neoplasms
require different treatment approaches and surveillance
compared to nonhereditary cases and also lead to preclin-
ical diagnosis in family members. Therefore, it is important
to appropriately screen patients with suspected hereditary
neoplasia. In settings of MEN1, GEP-NEN is an important
prognostic factor along with thymic NEN [68,69].
Meanwhile, VHL disease rarely affects prognosis.
Regarding diagnosis, MEN1 or VHL disease is sus-
pected in cases of GEP-NENs that meet the criteria in
Table 4, thus requiring further assessment including
searching for associated pathologies and genetic testing.
MEN1 or VHL disease may also be associated with
multiple small pancreatic NENs, and EUS-FNA with CT or
MRI is recommended for localization. If a functional tumor
is suspected, other nonfunctioning tumors are also often
present; therefore, a SASI test is recommended [70].
The indications for surgery for GEP-NENs in settings of
MEN1 or VHL disease are essentially the same as those for
sporadic cases. However, because they involve multifocal
and recurrent tumors, follow-up is generally recommended
for nonfunctioning tumors \2 cm in a setting of MEN1
[71]. Surgery is considered for tumors that are C2cmor
have a high growth rate. If surgery is indicated, a procedure
that preserves as much pancreatic function as possible is
recommended. In a setting of VHL disease, surgery is
considered for tumors that are C2 cm and have a doubling
time \500 days [72].
Regarding surveillance, the growth rate of GEP-NETs
associated with MEN1 is slow (0.1–1.3 mm/year);
tumors \1 cm, in particular, show little growth [73]. In
patients with MEN1, annual follow-up including exami-
nation, imaging with CT or MRI, and biochemistry (i.e.,
fasting glucose, insulin, and gastrin) is recommended,
keeping functional NENs in mind [74]. In patients with
VHL disease, follow-up with dynamic CT every 2–3 years
is recommended for tumors that are \2 cm and have a
doubling time C500 days and every 6 months to 1 year
for tumors meeting only 1 of the 2 conditions.
Table 3 Treatment approaches for gastroenteropancreatic neuroendocrine neoplasms
NETs NECs
Pancreatic origin Gastrointestinal origin Pancreatic
origin
Gastrointestinal
origin
Local therapy Primary: resection Primary: resection, endoscopic treatment Resection ±adjuvant
chemotherapy
a
Metastasis: resection, RFA (for liver
metastasis), TACE (for liver
metastasis)
Metastasis: resection, RFA (for liver
metastasis), TACE (for liver
metastasis)
Symptom management:
somatostatin
analogues
Octreotide Octreotide
Lanreotide Lanreotide
Tumor control:
somatostatin
analogues
Lanreotide Octreotide – –
Lanreotide
Tumor control:
molecular targeted
drugs
Everolimus Everolimus –
Sunitinib
Tumor control:
cytotoxic anticancer
agents
Streptozocin Etoposide/cisplatin
Temozolomide
a
Irinotecan/cisplatin
– Etoposide/carboplatin
Tumor control: radiation Radiation (for bone metastases, brain metastases): PRRT* Radiation (for bone or brain
metastasis)
a
Off-label in Japan
In recent years, a high response rate of temozolomide therapy for pancreatic NET has been reported overseas [64]. Based on these results,
guidelines also recommend temozolomide combination therapy is recommend as options for patients with large tumors and symptomatic patients
in Europe and the U.S. Temozolomide combination therapy is one of the useful treatments, but it is not approved for insurance in Japan.
Furthermore, radionuclide-labeling peptide therapy (PRRT) [65] is often used in Europe and the U.S. as well. PRRT has recently been covered by
insurance in Japan, but at present, it should be given priority to patients who are ineffective with other drugs after the second treatment and need
immediate PRRT treatment.
123
J Gastroenterol
Conclusions
The revised clinical practice guidelines encompass the
revised WHO classification for new diagnostic modalities,
pathological diagnosis, surgical and
medical/multidisciplinary treatments for pancreatic NETs,
and the management of pancreatic NETs in a setting of
hereditary diseases by addressing issues encountered in
daily clinical practice.
Compared to countries other than Japan, the frequency
of rectal NETs among gastrointestinal NETs is high in
Japan, whereas the frequency of midgut NETs is high in
Europe and the U.S. In addition, the frequency of MEN1 in
cases of pancreatic NET is lower in Japan than that in
Europe and the U.S. Accordingly, diagnostic and treatment
approaches differ between patients from Japan and Europe
or the U.S., requiring specific guidelines for patients in
Japan. Thus, the revised guidelines contain specific
strategies for GEP-NEN care in Japan, emphasizing clini-
cal practicality.
Fig. 3 Treatment strategy for metastatic/recurrent GEP-NEN. GEP-
NEN: gastroenteropancreatic neuroendocrine neoplasm; NEC: neu-
roendocrine carcinoma; NET: neuroendocrine tumor; PPI: proton
pump inhibitor; RFA: radiofrequency ablation; SSA: somatostatin
analogue; TACE: transarterial chemoembolization; TAE: tran-
scatheter arterial embolization
Table 4 Criteria for suspected MEN1 or VHL disease in cases of
gastroenteropancreatic neuroendocrine neoplasms
1. Multiple pancreatic NETs
2. Recurrent pancreatic NETs
3. Gastrinomas (particularly of duodenal origin) (NEN-1)
4. Insulinomas in younger patients (MEN-1)
5. Complicated by hypercalcemia (MEN-1)
6. Presence and history of MEN1 or VHL-associated neoplasms
7. Familial history of MEN1 or VHL-associated neoplasms
MEN1, multiple endocrine neoplasia type 1; NET, neuroendocrine
tumor; VHL, von Hippel–Lindau
123
J Gastroenterol
Appendix
Members of the Guidelines Committee who created and
evaluated the JNETS ‘‘Clinical practice guidelines for
Gastroenteropancreatic Neuroendocrine Neoplasms’’ are
listed below.
Executive Committee: Chair: Tetsuhide ITO (Neuroen-
docrine Tumor Centre, Fukuoka Sanno Hospital.
Department of Gastroenterology, Graduate School of
Medical Sciences, Internal University of Health and
Welfare).
Vice-Chair: Toshihiko Masui (Department of Surgery,
Graduate School of Medicine, Kyoto University), and
Izumi Komoto (Neuroendorine Tumor Center, Kansai
Electric Power Hospital).
Members: Ryuichiro Doi (Department of Surgery, Otsu
Red Cross Shiga Hospital), Robert Y. Osamura (Depart-
ment of Diagnostic Pathology Nippon Koukan Hospital),
Akihiro Sakurai (Department of Medical Genetics and
Genomics, Sapporo Medical University School of Medi-
cine), Masafumi Ikeda (Department of Hepatobiliary and
Pancreatic Oncology, National Cancer Center Hospital
East), Koji Takano (Department of Endocrinology, Dia-
betes and Metabolism, Kitasato University School of
Medicine), Hisato Igarashi (Igarashi Medical Clinic), Akira
Shimatsu (Advanced Medical Care Center, Kusatsu Gen-
eral Hospital), Kazuhiko Nakamura (Department of Gas-
troenterology and Hepatology, National Hospital
Organization Fukuokahigashi Medical Center), Yuji
Nakamoto (Department of Diagnostic Imaging and Nuclear
Medicine, Graduate School of Medicine Kyoto University),
Susumu Hijioka (Department of Hepatobiliary and Pan-
creatic Oncology, National Cancer Center Hospital), Koji
Morita(Division of Endocrinology and Metabolism,
Department of Internal Medicine, Teikyo University
School of Medicine), Yuichi Ishikawa (Department of
Pathology, School of Medicine, International University of
Health and Welfare), Nobuyuki Ohike (Division of
Pathology, Shizuoka Cancer Center), Atsuko Kasajima
(Department of Pathology, Technical University Munich),
Ryoji Kushima (Department of Pathology, Shiga Univer-
sity of Medical Science), Motohiro Kojima (Division of
Pathology, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center), Hironobu Sasano
(Department of Pathology, Tohoku University School of
Medicine), Satoshi Hirano (Department of Gastroentero-
logical Surgery II, Hokkaido University Faculty of Medi-
cine), Nobumasa Mizuno (Department of
Gastroenterology, Aichi Cancer Center Hospital), Taku
Aoki (Second Department of Surgery, Dokkyo Medical
University), Takeshi Aoki (Department of Surgery, Tohoku
University Graduate School of Medicine), Takao Ohtsuka
(Department of Digestive Surgery, Breast and Thyroid
Surgery, Graduate School of Medical Sciences, Kagoshima
University), Tomoyuki Okumura (Department of Surgery
and Science, Faculty of Medicine, Academic Assembly,
University of Toyama), Yasutoshi Kimura (Department of
Surgery, Surgical Oncology and Science, Sapporo Medical
University), Atsushi Kudo (Department of Hepatobiliary
and Pancreatic Surgery, Graduate School of Medicine,
Tokyo Medical and Dental University), Tsuyoshi Konishi
(Cancer Institute Hospital of the Japanese Foundation for
Cancer Research), Ippei Matsumoto (Department of Sur-
gery, Kindai University Faculty of Medicine), Noritoshi
Kobayashi (Department of Oncology, Yokohama City
University Graduate School of Medicine), Nao Fujimori
(Department of Medicine and Bioregulatory Science,
Graduate School of Medical Sciences, Kyushu University),
Yoshitaka Honma (Department of Head and Neck, Eso-
phageal Medical Oncology, National Cancer Center
Hospital), Chigusa Morizane (Department of Hepatobiliary
and Pancreatic Oncology, National Cancer Center Hospi-
tal), Shinya Uchino (Noguchi Thyroid Clinic and Hospital
Foundation), Kiyomi Horiuchi (Department of Breast and
Endocrine Surgery, Tokyo Women’s Medical University),
Masanori Yamasaki (Division of Diabetes, Endocrinology
and Metabolism, Department of Internal Medicine, Shinshu
University School of Medicine).
Evaluation Committee: Chair: Masayuki Imamura
(Neuroendorine Tumor Center, Kansai Electric Power
Hospital).
Members: Koichi Hirata (Department of Surgery, JR
Sapporo Hospital), Takuji Okusaka (Department of Hepa-
tobiliary and Pancreatic Oncology, National Cancer Center
Hospital), Mitsuhiro Kida (Department of Gastroenterol-
ogy, Kitasato University School of Medicine), Wataru
Kimura (Toto Kasukabe Hospital), Masao Tanaka
(Department of Surgery, Shimonoseki City Hospital),
Yoshiyuki Majima (NPO PanCAN Japan).
Main Collaborators: Jun Matsubayashi (Department of
Surgery, Otsu Red Cross Shiga Hospital), Yuichi Sato
(Nagaoka central general Hospital), Masau Sekiguchi
(Cancer Screening Center/Endoscopy Division, National
Cancer Center Hospital), Shinichi Abe (Japan Medical
Library Association).
Observer: Robert T. Jensen (Cell Biology Section,
Digestive Diseases Branch, National Institutes of Diabetes,
Digestive and Kidney Diseases. National Institutes of
Health, USA.).
The Japanese Neuroendocrine Tumor Society: Presi-
dent, Shinji Uemoto (Department of Hepato-Biliary-Pan-
creatic Surgery and Transplantation, Graduate School of
Medicine, Kyoto University).
123
J Gastroenterol
Declarations
Conflict of interest Any financial relationship with enterprises,
businesses, or academic institutions in the subject matter or materials
discussed in the manuscript are listed as follows; Tetsuhide Ito
received speaker’s honoraria from Teijin Pharma Limited, Novartis
Pharma and Mylan EPD. Robert Y. Osamura received speaker’s
honoraria from Teijin Pharma Limited and Novartis Pharma. Akihiro
Sakurai received speaker’s honoraria from AstraZeneca. Masafumi
Ikeda received speaker’s honoraria from Novartis Pharma, Chugai
Pharmaceutical Co. Ltd. and Bayer Pharma. Susumu Hijioka received
speaker’s honoraria from Teijin Pharma Limited, Novartis Pharma
and Novel Pharma. Taku Aoki received speaker’s honoraria from
Novartis Pharma. Atsuko Kasajima received speaker’s honoraria from
Teijin Pharma Limited. Ryoji Kushima received speaker’s honoraria
from Takeda Pharmaceutical Co. Ltd. Hironobu Sasano received
speaker’s honoraria from Novartis Pharma. Tsuyoshi Konishi
received speaker’s honoraria from Olympus and Johnson & Johnson.
The authors, the spouse, partner or immediate relatives of the authors
report no other conflicts of interest in this work.
Supplementary InformationThe online version contains
supplementary material available at https://doi.org/10.1007/s00535-
021-01827-7.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons licence, and indicate
if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons licence, unless
indicated otherwise in a credit line to the material. If material is not
included in the article’s Creative Commons licence and your intended
use is not permitted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from the copyright
holder. To view a copy of this licence, visit http://creativecommons.
org/licenses/by/4.0/.
References
1. Imanura M, Ito T, Masui T, et al. Clinical practice guidelines for
gastroenteropancreatic neuroendocrine tumors (NET). In: Japa-
nese Neuroendocrine Tumor Society. 1st ed. Tokyo: Kanehara
syuppan; 2015 (in Japanese).
2. Ito T, Masui T, Komoto I, et al. Clinical practice guidelines for
gastroenteropancreatic neuroendocrine neoplasms (NEN) 2019.
In: Japanese Neuroendocrine Tumor Society. 2nd ed. Tokyo:
Kanehara syuppan; 2019 (in Japanese).
3. Ito T, Hijioka S, Masui T, et al. Advances in the diagnosis and
treatment of pancreatic neuroendocrine neoplasms in Japan.
J Gastroenterol. 2017;52:9–18.
4. Ito T, Jensen RT. Molecular imaging in neuroendocrine tumors:
recent advances, controversies, unresolved issues, and roles in
management. Curr Opin Endocrinol Diabetes Obes.
2017;24:15–24.
5. Lloyd RV, Osamura RY, Klo
¨ppel G, et al. WHO classification of
tumours of endocrine organs. In: Lloyd RV, editor. WHO clas-
sification of tumours, vol. 10. 4th ed. Lyon: International Agency
for Research on Cancer; 2017.
6. Gill A, Klimstra D, Lam A, et al. WHO classification of tumours:
digestive system tumours. 5th ed. Lyon: International Agency for
Research on Cancer; 2019.
7. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of
advanced, non-functional neuroendocrine tumours of the lung or
gastrointestinal tract (RADIANT-4): a randomised, placebo-
controlled, phase 3 study. Lancet. 2016;387:968–77.
8. Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic
enteropancreatic neuroendocrine tumors. N Engl J Med.
2014;371:224–33.
9. Ito T, Honma Y, Hijioka S, et al. Phase II study of lanreotide
autogel in Japanese patients with unresectable or metastatic well-
differentiated neuroendocrine tumors. Investig New Drugs.
2017;35(4):499–508.
10. Ito T, Fujimori N, Honma Y, et al. Long-term safety and efficacy
of lanreotide autogel in Japanese patients with neuroendocrine
tumors: final results of a phase II open-label extension study. Asia
Pac J Clin Oncol. 2020. https://doi.org/10.1111/ajco.13371
((online ahead of print)).
11. Igarashi H, Ito T, Nishimori I, et al. Pancreatic involvement in
Japanese patients with von Hippel–Lindau disease: results of a
nationwide survey. J Gastroenterol. 2014;49:511–6.
12. Ito T, Igarashi H, Nakamura K, et al. Epidemiological trends of
pancreatic and gastrointestinal neuroendocrine tumors in Japan: a
nationwide survey analysis. J Gastroenterol. 2015;50:58–64.
13. Ito T, Sasano H, Tanaka M, et al. Epidemiological study of
gastroenteropancreatic neuroendocrine tumors in Japan. J Gas-
troenterol. 2010;45:234–43.
14. Cryer PE, Axelrod L, Grossman AB, et al. Endocrine Society.
Evaluation and management of adult hypoglycemic disorders: an
endocrine society clinical practice guideline. J Clin Endocrinol
Metab. 2009;94:709–28.
15. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors:
pancreatic endocrine tumors. Gastroenterology.
2008;135:1469–92.
16. Cryer PE, et al. Glucose homeostasis and hypoglycemia. In:
Melmed S, Polonsky K, Larsen PR, et al., editors. Williams
textbook of endocrinology. 11th ed. Philadelphia: Saunders;
2008. p. 1503–33.
17. Ueda K, Kawabe K, Lee L, et al. Diagnostic performance of
48-hour fasting test and insulin surrogates in patients with sus-
pected insulinoma. Pancreas. 2017;46:476–81.
18. Jensen RT, Cadiot G, Brandi ML, et al. Barcelona consensus
conference participants. ENETS consensus guidelines for the
management of patients with digestive neuroendocrine neo-
plasms: functional pancreatic endocrine tumor syndromes. Neu-
roendocrinology. 2012;95:98–119.
19. Berna MJ, Hoffmann KM, Serrano J, et al. Serum gastrin in
Zollinger–Ellison syndrome: I. Prospective study of fasting serum
gastrin in 309 patients from the National Institutes of Health and
comparison with 2229 cases from the literature. Medicine (Bal-
timore). 2006;85:295–330.
20. Berna MJ, Hoffmann KM, Long SH, et al. Serum gastrin in
Zollinger–Ellison syndrome: II. Prospective study of gastrin
provocative testing in 293 patients from the National Institutes of
Health and comparison with 537 cases from the literature.
Evaluation of diagnostic criteria, proposal of new criteria, and
correlations with clinical and tumoral features. Medicine (Balti-
more). 2006;85:331–64.
21. Ito T, Igarashi H, Jensen RT. Zollinger–Ellison syndrome: recent
advances and controversies. Curr Opin Gastroenterol.
2013;29:650–61.
22. Gibril F, Schumann M, Pace A, et al. Multiple endocrine neo-
plasia type 1 and Zollinger–Ellison syndrome: a prospective
study of 107 cases and comparison with 1009 cases from the
literature. Medicine (Baltimore). 2004;83:43–83.
23. Zilli A, Arcidiacono PG, Conte D, et al. Clinical impact of
endoscopic ultrasonography on the management of neuroen-
docrine tumors: lights and shadows. Dig Liver Dis. 2018;5:6–14.
123
J Gastroenterol
24. Abboud B, Boujaoude J. Occult sporadic insulinoma: localization
and surgical strategy. World J Gastroenterol. 2008;14:657–65.
25. Imamura M, Takahashi K, Adachi H, et al. Usefulness of selec-
tive arterial secretin injection test for localization of gastrinoma
in the Zollinger–Ellison syndrome. Ann Surg. 1987;205:230–9.
26. Kim BN, Sohn DK, Hong CW, et al. Atypical endoscopic fea-
tures can be associated with metastasis in rectal carcinoid tumors.
Surg Endosc. 2008;22:1992–6.
27. Shim KN, Yang SK, Myung SJ, et al. Atypical endoscopic fea-
tures of rectal carcinoids. Endoscopy. 2004;36:313–6.
28. Ruf J, Schiefer J, Furth C, et al. 68Ga-DOTATOC PET/CT of
neuroendocrine tumors: spotlight on the CT phases of a triple-
phase protocol. J Nucl Med. 2011;52:697–704.
29. Dromain C, de Baere T, Lumbroso J, et al. Detection of liver
metastases from endocrine tumors: a prospective comparison of
somatostatin receptor scintigraphy, computed tomography, and
magnetic resonance imaging. J Clin Oncol. 2005;23:70–8.
30. Kubota K, Okasaki M, Minamimoto R, et al. Lesion-based
analysis of 18F-FDG uptake and 111In-Pentetreotide uptake by
neuroendocrine tumors. Ann Nucl Med. 2014;28:1004–10.
31. Konukiewitz B, Schlitter AM, Jesinghaus M, et al. Somatostatin
receptor expression related to TP53 and RB1 alterations in pan-
creatic and extrapancreatic neuroendocrine neoplasms with a
Ki67-index above 20. Mod Pathol. 2017;30:587–98.
32. Hijioka S, Hosoda W, Matsuo K, 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. 2017;23:4625–32.
33. Ohmoto A, Rokutan H, Yachida S. Pancreatic neuroendocrine
neoplasms: basic biology, current treatment strategies and pro-
spects for the future. Int J Mol Sci. 2017;18:143.
34. Gratian L, Pura J, Dinan M, et al. Impact of extent of surgery on
survival in patients with small nonfunctional pancreatic neu-
roendocrine tumors in the United States. Ann Surg Oncol.
2014;21:3515–21.
35. Hashim YM, Trinkaus KM, Linehan DC, et al. Regional lym-
phadenectomy is indicated in the surgical treatment of pancreatic
neuroendocrine tumors (PNETs). Ann Surg. 2014;259:197–203.
36. Sadot E, Reidy-Lagunes DL, Tang LH, et al. Observation versus
resection for small asymptomatic pancreatic neuroendocrine
tumors: a matched case-control study. Ann Surg Oncol.
2016;23:1361–70.
37. Lee LC, Grant CS, Salomao DR, et al. Small, nonfunctioning,
asymptomatic pancreatic neuroendocrine tumors (PNETs): role
for nonoperative management. Surgery. 2012;152:965–74.
38. Jung JG, Lee KT, Woo YS, et al. Behavior of small, asymp-
tomatic, nonfunctioning pancreatic neuroendocrine tumors (NF-
PNETs). Medicine (Baltimore). 2015;94:e983.
39. Rosenberg AM, Friedmann P, Del Rivero J, et al. Resection
versus expectant management of small incidentally discovered
nonfunctional pancreatic neuroendocrine tumors. Surgery.
2016;159:302–9.
40. Vanderveen K, Grant C. Insulinoma. Cancer Treat Res.
2010;153:235–52.
41. Espana-Gomez MN, Velazquez-Fernandez D, Bezaury P, et al.
Pancreatic insulinoma: a surgical experience. World J Surg.
2009;33:1966–70.
42. Akerstrom G, Hellman P. Surgery on neuroendocrine tumours.
Best Pract Res Clin Endocrinol Metab. 2007;21:87–109.
43. Albers MB, Manoharan J, Bollmann C, et al. Results of duo-
denopancreatic reoperations in multiple endocrine neoplasia type
1. World J Surg. 2019;43:552–8.
44. Doi R. Determinants of surgical resection for pancreatic neu-
roendocrine tumors. J Hepatobiliary Pancreat Sci.
2015;22:610–7.
45. Tang LH, Untch BR, Reidy DL, et al. Well-differentiated neu-
roendocrine tumors with a morphologically apparent high-grade
component: a pathway distinct from poorly differentiated neu-
roendocrine carcinomas. Clin Cancer Res. 2016;22:1011–7.
46. Wong AT, Shao M, Rineer J, et al. Treatment and survival out-
comes of small cell carcinoma of the esophagus: an analysis of
the National Cancer Data Base. Dis Esophagus. 2016;30:1–5.
47. Deng HY, Li G, Luo J, et al. The role of surgery in treating
resectable limited disease of esophageal neuroendocrine carci-
noma. World J Surg. 2018;42:2428–36.
48. Rindi G, Luinetti O, Cornaggia M, et al. Three subtypes of gastric
argyrophil carcinoid and the gastric neuroendocrine carcinoma: a
clinicopathologic study. Gastroenterology. 1993;104:994–1006.
49. Howe JR, Cardona K, Fraker DL, et al. The surgical management
of small bowel neuroendocrine tumors: consensus guidelines of
the North American Neuroendocrine Tumor Society. Pancreas.
2017;46:715–31.
50. Brighi N, La Rosa S, Rossi G, et al. Morphological factors related
to nodal metastases in neuroendocrine tumors of the appendix: a
multicentric retrospective study. Ann Surg. 2020;271:527–33.
51. Pawa N, Clift AK, Osmani H, et al. Surgical management of
patients with neuroendocrine neoplasms of the appendix:
appendectomy or more. Neuroendocrinology. 2018;106:242–51.
52. Sohn B, Kwon Y, Ryoo SB, et al. Predictive factors for lymph
node metastasis and prognostic factors for survival in rectal
neuroendocrine tumors. J Gastrointest Surg. 2017;21:2066–74.
53. Tsang ES, McConnell YJ, Schaeffer DF, et al. Prognostic factors
for locoregional recurrence in neuroendocrine tumors of the
rectum. Dis Colon Rectum. 2018;61:187–92.
54. Takatsu Y, Fukunaga Y, Nagasaki T, et al. Short- and long-term
outcomes of laparoscopic total mesenteric excision for neuroen-
docrine tumors of the rectum. Dis Colon Rectum. 2017;60:284–9.
55. National Comprehensive Cancer Network (NCCN) Clinical
practice guidelines in oncology for neuroendocrine and adrenal
tumors (Version 1.2019). https://www.nccn.org/professionals/
physician_gls/pdf/neuroendocrine.pdf. Accessed 3 July 2020.
56. Pavel M, O’Toole D, Costa F, et al. ENETS consensus guidelines
update for the management of distant metastatic disease of
intestinal, pancreatic, bronchial neuroendocrine neoplasms
(NEN) and NEN of unknown primary site. Neuroendocrinology.
2016;103:172–85.
57. Pavel M, O
¨berg K, Falconi M, et al. Gastroenteropancreatic
neuroendocrine neoplasms: ESMO clinical practice guidelines for
diagnosis, treatment and follow-up. Ann Oncol. 2020;31:844–60.
58. Kvols LK, Moertel CG, O’Connell MJ, et al. Treatment of the
malignant carcinoid syndrome. Evaluation of a long-acting
somatostatin analogue. N Engl J Med. 1986;315:663–6.
59. Rinke A, Mu
¨ller HH, Schade-Brittinger C, et al. Placebo-con-
trolled, double-blind, prospective, randomized study on the effect
of octreotide LAR in the control of tumor growth in patients with
metastatic neuroendocrine midgut tumors: a report from the
PROMID Study Group. J Clin Oncol. 2009;27:4656–63.
60. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pan-
creatic neuroendocrine tumors. N Engl J Med. 2011;364:514–23.
61. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the
treatment of pancreatic neuroendocrine tumors. N Engl J Med.
2011;364:501–13.
62. Moertel CG, Lefkopoulo M, Lipsitz S, et al. Streptozocin-dox-
orubicin, streptozocin-fluorouracil or chlorozotocin in the treat-
ment of advanced islet-cell carcinoma. N Engl J Med.
1992;326:519–23.
63. Sun W, Lipsitz S, Catalano P, et al. Phase II/III study of dox-
orubicin with fluorouracil compared with streptozocin with flu-
orouracil or dacarbazine in the treatment of advanced carcinoid
tumors: Eastern Cooperative Oncology Group Study E1281.
J Clin Oncol. 2005;23:4897–904.
123
J Gastroenterol
64. Pamela L. Kunz PJC, Halla N, et al. A randomized study of
temozolomide or temozolomide and capecitabine in patients with
advanced pancreatic neuroendocrine tumors: a trial of the ECOG-
ACRIN Cancer Research Group (E2211). J Clin Oncol.
2018;36(suppl):abstr 4004.
65. Strosberg JR, Haddad G, Wolin E, et al. Phase 3 trial of 177Lu-
dotatate for midgut neuroendocrine tumors. N Engl J Med.
2017;376:125–35.
66. Ikeda M, Morizane C, Hijioka S, et al. Optimal strategy of sys-
temic treatment for unresectable pancreatic neuroendocrine
tumors based upon opinion of Japanese experts. Pancreatology.
2020;20:944–50.
67. Scarpa A, Chang DK, Nones K, et al. Whole-genome landscape
of pancreatic neuroendocrine tumours. Nature. 2017;543:65–71.
68. Goudet P, Murat A, Binquet C, et al. Risk factors and causes of
death in MEN1 disease. A GTE (Groupe d’Etude des Tumeurs
Endocrines) cohort study among 758 patients. World J Surg.
2010;34:249–55.
69. Ito T, Igarashi H, Uehara H, et al. Causes of death and prognostic
factors in multiple endocrine neoplasia type 1: a prospective
study: comparison of 106 MEN1/Zollinger–Ellison syndrome
patients with 1613 literature MEN1 patients with or without
pancreatic endocrine tumors. Medicine (Baltimore).
2013;92:135–81.
70. Imamura M, Nakamoto Y, Uose S, et al. Diagnosis of functioning
pancreaticoduodenal neuroendocrine tumors. J Hepatobiliary
Pancreat Sci. 2015;22:602–9.
71. Sadowski SM, Pieterman CRC, Perrier ND, et al. Prognostic
factors for the outcome of nonfunctioning pancreatic neuroen-
docrine tumors in MEN1: a systematic review of literature.
Endocr Relat Cancer. 2020;27:R145–61.
72. Blansfield JA, Choyke L, Morita SY, et al. Clinical, genetic and
radiographic analysis of 108 patients with von Hippel-Lindau
disease (VHL) manifested by pancreatic neuroendocrine neo-
plasms (PNETs). Surgery. 2007;142:814–8.
73. D’Souza SL, Elmunzer BJ, Scheiman JM. Long-term follow-up
of asymptomatic pancreatic neuroendocrine tumors in multiple
endocrine neoplasia type I syndrome. J Clin Gastroenterol.
2014;48:458–61.
74. Niederle B, Selberherr A, Bartsch D, et al. Multiple endocrine
neoplasia type 1 (MEN1) and the pancreas—diagnosis and
treatment of functioning and non-functioning pancreatic and
duodenal neuroendocrine neoplasia within the MEN1 syn-
drome—an international consensus statement. Neuroendocrinol-
ogy. 2021;111:609–30.
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
123
J Gastroenterol
... Neuroendocrine neoplasms (NEN) are derived from neuroendocrine cells and can occur in a variety of organs, although those primary to the biliary tract are rare [1][2][3]. Mixed neuroendocrine non-neuroendocrine neoplasms (MiNEN) are defined as mixed neoplasms with both neuroendocrine and non-neuroendocrine components, with either component representing at least 30% [1,2]. A diagnosis of MiNEN of the biliary tract is difficult, and its prognosis is extremely poor. ...
... Neuroendocrine neoplasms (NEN) are derived from neuroendocrine cells and can occur in a variety of organs, although those primary to the biliary tract are rare [1][2][3]. Mixed neuroendocrine non-neuroendocrine neoplasms (MiNEN) are defined as mixed neoplasms with both neuroendocrine and non-neuroendocrine components, with either component representing at least 30% [1,2]. A diagnosis of MiNEN of the biliary tract is difficult, and its prognosis is extremely poor. ...
... The surgical method was pancreaticoduodenectomy, which was highly invasive, and it was suggested that R0 resection might not be possible due to tumor invasion, so we decided to start preoperative chemotherapy. Chemotherapy for NEC recommends combination therapy of cisplatin (CDDP) and ETP, and as an alternative therapy is recommended to use CBDCA instead of CDDP [1,13]. We chose treatment with CBDCA because the patient was relatively elderly and we wanted to reduce the occurrence of adverse events. ...
Article
Full-text available
A 74-year-old man with obstructive jaundice presented with a thickened distal bile duct wall. A transpapillary forceps biopsy revealed an adenocarcinoma; however, because the tumor image was different from that of a typical cholangiocarcinoma, endoscopic ultrasound-guided fine-needle aspiration was performed on the tumor and enlarged lymph nodes. The tumor cells were positive for synaptophysin and CD56 with a Ki67 labeling index of 95%, and he was diagnosed with small cell neuroendocrine carcinoma. We diagnosed a bile duct tumor with neuroendocrine carcinoma component with lymph node metastasis. Preoperative chemotherapy for neuroendocrine carcinoma was administered because R0 resection was difficult and the risk of postoperative recurrence was high. Three courses of chemotherapy with carboplatin and etoposide resulted in marked tumor shrinkage, and radical resection was performed 3 months after diagnosis. Postoperative pathology revealed adenocarcinoma in the mucosal epithelium and small cell neuroendocrine carcinoma in the submucosa, most of which resolved with chemotherapy. Carboplatin and etoposide were resumed as adjuvant chemotherapy, and 67 months of recurrence-free survival were achieved after surgery.
... Tumors recommends surgical resection of non-functioning tumors < 2 m [10] . Recent studies have also demonstrated that aggressive surgical resection prolongs patient survival, and in the study of Toshitaka Sugawara et al., a total of 4,641 patients were enrolled and were divided into group 1a (≤ 1 cm) according to tumor size and group 1b (1.1-2.0 cm) and found that surgical resection was not associated with survival in group 1a, but was positively correlated with prolonged survival in patients in group 1b [11] . ...
Preprint
Full-text available
Objective To explore the effectiveness and safety of fluorescence imaging technology in the intraoperative localization of small neuroendocrine tumors of the pancreas. Methods Indocyanine green fluorescence imaging technology was used in operation, and the specific process of indocyanine green administration was as follows: 25 mg of indocyanine green was dissolved in 10 ml of sterilized water for injection, and 1 ml of indocyanine green was injected rapidly through the peripheral vein during the laparoscopic exploration of the tumor, and the tumor was observed to show green fluorescence about 1 min later, and the tumor showed green fluorescence, which was regarded as the success of development, and laparoscopic pancreatic tumor local excision surgery was carried out under the guidance of the real-time fluorescence imaging, and postoperative monitoring of the amylase index of drainage fluid to observe whether there was a pancreatic fistula. Under the real-time guidance of fluorescence imaging, laparoscopic pancreatic tumor local resection was performed, and the amylase index of the drainage fluid was monitored after surgery to observe whether pancreatic fistula occurred or not and to record the tumor grade, surgical margins, and other pathological conditions. RESULTS The fluorescence imaging effect of the two patients was good, the lesion boundary was clear, and both of them completed the local resection of the tumor laparoscopically without pancreatic fistula, and the pathology suggested that the pancreatic neuroendocrine tumors were grade G1, and the margins of the surgery were negative. CONCLUSION Fluorescence imaging technology helps to localize small pancreatic neuroendocrine tumors intraoperatively with good safety.
... In the assessment of rectal operations, a positive margin assumes critical importance indicative of worse prognosis and regarded as a sign of the need for salvage treatment 8 . Consequently, relevant guidelines advocate for salvage radical surgery following endoscopic resection in cases where RNETs exhibit a positive resection margin 9 . Recently, several retrospective studies have witnessed a low recurrence rate among RNETs patients with positive resection margin following endoscopic resection 10,11 , which resulted in the question about the clinical signi cance of positive resection and a lack of consensus regarding the management strategy after the endoscopic resection of RNETs patients revealing a positive resection margin. ...
Preprint
Full-text available
Background The incidence of rectal neuroendocrine tumors (RNETs) has witnessed a significant surge, with a notable proportion being amenable to endoscopic removal. However, the clinical significance of positive resection margin for RNETs patients following endoscopic resection remain unknown, resulting in a lack of consensus regarding the appropriateness of implementing salvage treatment. Methods In this large, multicenter, retrospective cohort study, we analyzed the medical records of individuals who underwent endoscopic resection for RNETs and classified them into two groups: the positive resection margin and the negative resection margin group. The overall survival (OS) and disease-free survival (DFS) were compared among two group. The independent variables were identified using univariate and multivariate logistic regression analyses to predict positive resection margin. Then, the model was established to predict the patients with positive resection margin using multivariate logistic regression. Results 181 RNETs patients (34.3%) represented positive margin after endoscopic resection. Following a median follow-up period of 72 months, tumor recurrence manifested in 12 out of 527 patients (2.2%) and the presence of positive resection margin was associated with worse DFS. Independent factors correlating with positive resection margin included endoscopic resection method choice, RNETs located in the low rectum, NLR > 4.44 and tumor size exceeding 14.89 mm. A prediction model was therefore established with high predictive accuracy and excellent clinical applicability determined by calibration curves and DCA curve. Among RNETs patients with positive margin following endoscopic resection, implementing salvage treatment was beneficial for improving DFS and salvage endoscopic resection offer equal efficiency compared with salvage radical resection . Conclusions Positive resection margin following endoscopic resection may indicate negative prognosis. Salvage treatment can improve the prognosis of RNETs patients with positive resection margin. Notably, salvage local resection exhibited similar efficacy compared with radical surgery in term of survival benefit.
... PanNEN is expected to have a relatively good prognosis, but even in Grade 1, the recurrence rate is by no means low, and since it often occurs in young people, long-term follow-up is necessary. In recent years, the number of therapeutic agents that have been used (such as sustained somatostatin analog preparations (octreotide), antineoplastic agents (streptozocin, everolimus, and sunitinib), and diabetic drugs) has increased, but treatment duration is lengthy, even in unresectable/recurrent cases [4][5][6]. It is difficult to continue ordinary chemotherapy alone, and effective drug selection that can be used without adverse events in the long term is required. ...
Article
Full-text available
Although pancreatic neuroendocrine neoplasms (panNENs) are much less common and have a better prognosis than exocrine pancreatic cancers, their recurrence rate is not low, even in Grade 1 (World Health Organization classification) panNEN. Recently, there have been several reports that the progression-free survival in patients with unresectable panNEN could be improved by an antidiabetic drug, metformin, with the co-treatment of everolimus or a somatostatin analog. In this study, we aimed to evaluate the effects of metformin on cell metabolism and viability using the panNEN cell line, QGP-1, and RIN-m in culture. We observed an inhibitory effect of metformin on QGP-1 cell proliferation in a dose-dependent manner. Metformin was found to decrease the oxygen consumption rate in QGP-1 and RIN-m cells after metformin 48 h treatment and immediately after exposure. Cell proliferation was suppressed after metformin treatment. Phosphorylated adenosine monophosphate-activated protein kinase (AMPK) expression was increased, and cyclin D1 expression was decreased in RIN-m cells 24 h after metformin treatment by Western blotting in a dose-dependent manner. In conclusion, suppressive mitochondrial respiration and AMPK activation by metformin are, thus, suggested to inhibit panNEN cell viability and cell survival.
... In Japan, endoscopic treatment is indicated for rectal NETs that are less than 10 mm wide, designated a World Health Organization (WHO) classification of no higher than grade 1, and remain in the submucosa without metastasis [4]. Endoscopic treatment for rectal NETs includes conventional endoscopic mucosal resection (EMR), cap-assisted EMR, endoscopic submucosal resection with a ligation device, and endoscopic submucosal dissection (ESD) [5][6][7]. ...
Article
Background Controversy surrounds the indications for endoscopic treatment (ET) versus surgery in addressing gastrointestinal neuroendocrine neoplasms (GI-NENs). This paper aims to compare the long-term survival prognosis between ET and surgery for patients with GI-NENs. Methods A retrospective analysis of GI-NEN patients diagnosed between 2000 and 2020 was conducted using the SEER database. Overall survival (OS) and cancer-specific survival (CSS) were estimated using the Kaplan-Meier method. Prognostic factors were assessed through univariate and multivariate Cox regression analyses. Propensity score matching (PSM) was employed to adjust for clinical variables. Results This study encompassed 12,016 patients with GI-NENs, with 3732 patients (31.1%) undergoing ET and 8284 patients (68.9%) opting for surgery. The rectum was the most frequent location for ET, while the small bowel was the predominant site for surgery. Both the ET and surgical groups exhibited similar overall survival risk and cancer-specific survival risk before and after matching. No significant differences in the 1-year, 3-year, 5-year, and 10-year OS and CSS rates were observed between the ET and surgery groups after PSM. Nevertheless, subgroup analysis revealed a significantly better CSS in the ET group than the surgery group, particularly in stage I and tumors sized <10mm ( P <0.01). In the colon subgroup, the OS and CSS of the ET group were superior to those of the surgery group ( P <0.05). Conclusion Endoscopic treatment and surgery demonstrate comparable long-term survival prognoses for treating GI-NENs. ET emerges as a viable option for patients averse to surgical interventions.
Article
Objective We aimed to compare different segmentation methods used to calculate prognostically valuable volumetric parameters, somatostatin receptor expressing tumor volume (SRETV), and total lesion somatostatin receptor expression (TLSRE), measured by ⁶⁸ Ga-DOTATATE PET/CT and to find the optimal segmentation method to predict prognosis. Patients and methods Images of 34 patients diagnosed with gastroenteropancreatic neuroendocrine tumor (GEPNET) who underwent ⁶⁸ Ga-DOTATATE PET/CT imaging were reanalyzed. Four different threshold-based methods (fixed relative threshold method, normal liver background threshold method, fixed absolute standardized uptake value (SUV) threshold method, and adaptive threshold method) were used to calculate SRETV and TLSRE values. SRETV of all lesions of a patient was summarized as whole body SRETV (WB-SRETV) and TLSRE of all lesions of a patient was computed as whole body TLSRE (WB-TLSRE). Results WB-SRETVs calculated with all segmentation methods were statistically significantly associated with progression-free survival except WB-SRETV at which was calculated using adaptive threshold method. The fixed relative threshold methods calculated by using 45% (WB-SRETV 45% ) and 60% (WB-SRETV 60% ) of the SUV value as threshold respectively, were found to have statistically significant highest prognostic value (C-index = 0.704, CI = 0.622–0.786, P = 0.007). Among WB-TLSRE parameters, WB-TLSRE 35% , WB-TLSRE 40% , and WB-TLSRE 50% had the highest prognostic value (C-index = 0.689, CI = 0.604–0.774, P = 0.008). Conclusion The fixed relative threshold method was found to be the most effective and easily applicable method to measure SRETV on pretreatment ⁶⁸ Ga-DOTATATE PET/CT to predict prognosis in GEPNET patients. WB-SRETV 45% (cutoff value of 11.8 cm ³ ) and WB-SRETV 60% (cutoff value of 6.3 cm ³ ) were found to be the strongest predictors of prognosis in GEPNET patients.
Article
Background: High-grade neuroendocrine neoplasms (NENs) comprise both well-differentiated grade 3 neuroendocrine tumors (G3 NETs) and poorly differentiated neuroendocrine carcinomas (NECs). Mixed neuroendocrine–non-neuroendocrine neoplasms (MiNENs) nearly always include poorly differentiated NEC as the neuroendocrine component. The efficacy and safety of frontline mFOLFIRINOX chemotherapy has never been investigated in patients with high-grade NENs. Patients and Methods: We conducted a multi-institutional retrospective analysis of patients with advanced high-grade NEN of the gastroenteropancreatic tract or of unknown origin seen between February 2016 and April 2023 who received treatment with frontline mFOLFIRINOX. Results: A total of 35 patients were included (G3 NETs: n=2; NECs: n=25; MiNENs: n=8; stage III: n=5; stage IV: n=30). The objective response rate was 77% (complete response: 3%; partial response: 74%). Median progression-free survival was 12 months (95% CI, 9.2–16.2 months) and median overall survival was 20.6 months (95% CI, 17.2–30.6 months). No significant differences in efficacy were seen according to primary site, histopathology, and Ki-67 proliferative index. All 5 patients with stage III disease who received mFOLFIRINOX obtained an objective response and underwent radical surgery or definitive radiotherapy with curative intent, with a recurrence rate of 40%. Grade 3 or 4 adverse events were observed in 43% of patients (mainly neutropenia and diarrhea). Females were at significantly increased risk of developing severe toxicities. Conclusions: mFOLFIRINOX shows antitumor activity against high-grade NENs. Well-designed, prospective clinical trials are needed to assess the efficacy of mFOLFIRINOX in both the neoadjuvant and metastatic settings.
Article
Introduction: Well-calibrated models for personalized prognostication of patients with gastrointestinal neuroendocrine neoplasms (GINENs) are limited. This study aimed to develop and validate a machine-learning model to predict the survival of patients with GINENs. Methods: Oblique random survival forest (ORSF) model, Cox proportional hazard risk model, Cox model with least absolute shrinkage and selection operator penalization, CoxBoost, Survival Gradient Boosting Machine, Extreme Gradient Boosting survival regression, DeepHit, DeepSurv, DNNSurv, logistic-hazard model, and PC-hazard model were compared. We further tuned hyperparameters and selected variables for the best-performing ORSF. Then, the final ORSF model was validated. Results: A total of 43,444 patients with GINENs were included. The median (interquartile range) survival time was 53 (19-102) months. The ORSF model performed best, in which age, histology, M stage, tumor size, primary tumor site, sex, tumor number, surgery, lymph nodes removed, N stage, race, and grade were ranked as important variables. However, chemotherapy and radiotherapy were not necessary for the ORSF model. The ORSF model had an overall C index of 0.86 (95% confidence interval, 0.85-0.87). The area under the receiver operation curves at 1, 3, 5, and 10 years were 0.91, 0.89, 0.87, and 0.80, respectively. The decision curve analysis showed superior clinical usefulness of the ORSF model than the American Joint Committee on Cancer Stage. A nomogram and an online tool were given. Conclusion: The machine learning ORSF model could precisely predict the survival of patients with GINENs, with the ability to identify patients at high risk for death and probably guide clinical practice.
Article
Background Somatostatin analogs, molecular-targeted agents and cytotoxic anticancer agents are available as therapeutic agents for the systemic treatment of pancreatic neuroendocrine tumors, and we have developed a first-line treatment selection MAP to enable selection of the optimal treatment strategy for pancreatic neuroendocrine tumors. The purpose of this study was to validate the usefulness of the treatment selection MAP. Methods Patients who had received systemic therapy for a pancreatic neuroendocrine tumor between January 2017 and December 2020 were compared according to whether they had been treated as recommended by the MAP (matched patients) or not (unmatched patients) to determine whether better outcomes were achieved by the matched patients. The primary endpoint was progression-free survival of the matched group and unmatched groups in the somatostatin analog, molecular-targeted agent and cytotoxic anticancer agents areas of the MAP. Results There were 41 (55%) MAP-matched patients in all areas among the 74 patients registered at seven hospitals. The MAP-matched rates were 100, 77 and 38% in the somatostatin analog area, molecular-targeted agent area and cytotoxic anticancer agents area, respectively. All of the unmatched patients had been selected for less intensive treatment. The median progression-free survival in the matched group and unmatched group in the molecular-targeted agent area of the MAP were 46.6 and 15.4 months, respectively, and a multivariate analysis identified MAP-matched (hazard ratio 0.18 [95% confidence interval: 0.04–0.87], P = 0.032) as the only significant independent favorable predictive factor. Conclusion The usefulness of the MAP for treatment selection was validated in the molecular-targeted agent area of the MAP.
Article
Full-text available
Aim The aim of this study was to describe the long‐term safety and efficacy of lanreotide in Japanese patients with neuroendocrine tumors. Methods The final analyses of a 48‐week open‐label phase II study (n = 32) and its extension study (n = 17) were conducted. Patients received 4‐weekly subcutaneous injections of lanreotide autogel 120 mg. Safety was evaluated by adverse events. Efficacy endpoints included tumor response by RECIST and change in tumor size. Post hoc analyses including tumor growth rate were performed. Results The median (range) of lanreotide exposure in the safety analysis set (n = 17) and efficacy analysis set (n = 28) were 151.4 (52–181) and 52.7 (12–181) weeks, respectively. Sixteen patients developed adverse drug reaction; of these, upper abdominal pain and urticaria were not reported before 48 weeks. No patient discontinued lanreotide or died from an adverse event. Two serious events of bile duct stones in one patient were drug‐related. Partial response was observed in 2 patients (7.1%; at 60 and 108 weeks), stable disease in 20 (71.4%) and progressive disease in 6 (21.4%). The mean of the greatest change from baseline in the sum of diameters of target lesions was −5.5%. The mean (standard deviation) tumor growth rate before treatment and from baseline to last observation was 25.3% (35.7%)/month and 6.4% (9.6%)/month, respectively. Conclusion Lanreotide treatment had an acceptable safety profile and was effective over long‐term treatment in Japanese patients with neuroendocrine tumors. No unexpected serious adverse events developed during prolonged use of lanreotide.
Article
Full-text available
Background: Esophageal neuroendocrine carcinoma (NEC) is a rare malignant tumor. The role of surgery in resectable limited disease of esophageal NEC remains unclear. How to select a specific group of limited disease of esophageal NEC who might benefit from surgery remains to be answered. Methods: Patients undergoing esophagectomy for resectable limited disease of esophageal NEC in our department from January 2007 to June 2015 were analyzed. TNM staging system was applied to describe those patients, and according to their different long-term prognosis after surgery, those patients were subgrouped into surgery response limited disease (SRLD) group and surgery non-response limited disease (SNRLD) group. Both univariate and multivariate analyses were applied to identify potential prognostic factors. Results: A total of 72 patients with resectable limited disease of esophageal NEC were identified for analysis. The median survival time of those patients was 21.5 months. There was no significant survival differences among stage I, stage IIA, and stage IIB patients, but all these patients had significantly longer survival than stage III patients. Therefore, stage I, stage IIA, and stage IIB patients were aggregated together as SRLD group, and stage III patients were aggregated as SNRLD group. SRLD patients obtained significantly longer survival than SNRLD patients in both univariate analysis and multivariate analysis. Moreover, adjuvant therapy could significantly benefit SRLD patients (P = 0.004) but could not benefit SNRLD patients (P = 0.136). Conclusions: Different responses to surgery existed in resectable limited disease of esophageal NEC indicating the need of further subgrouping for those patients. The resectable limited disease of esophageal NEC could be further subgrouped into SRLD group and SNRLD group according to the TNM staging system.
Article
The better understanding of the biological behavior of MEN1 organ manifestations and the in-crease in clinical experience warrant a revision of previously published guidelines. DP-NENs are still the second most common manifestation in MEN1 and, besides NENs of the thymus, remain a leading cause of death. DP-NENs are thus of main interest in the effort to re-evaluate recommendations for their diagnosis and treatment. Especially over the last two years, more clinical experience has documented the follow-up of treated and untreated (natural-course) DP-NENs. It was the aim of the international consortium of experts in endocrinology, genetics, radiology, surgery, gastroenterology and oncology to systematically review the literature and to present a consensus statement based on the highest levels of evidence. Reviewing the literature published over the past decade, the focus was on the diagnosis of F- and NF-DP-NENs within the MEN1 syn-drome in an effort to further standardize and improve treatment and follow-up, as well as to es-tablish a "logbook" for the diagnosis and treatment of DP-NENs. This shall help further reduce complications and improve long-term treatment results in these rare tumors. The following international consensus statement builds upon the previously published guide-lines of 2001 and 2012 and attempts to supplement the recommendations issued by various na-tional and international societies.
Article
Background/objectives A number of therapeutic agents have been reported to be clinically useful for the management of the patients with unresectable pancreatic neuroendocrine tumors (PanNETs) including somatostatin analogues, molecular-targeted agents and cytotoxic agents. However, the optimal strategy for selection among those treatment modalities above in these patients has remained unexplored. Methods Japanese experts for PanNET discussed and determined the optimal treatment strategies according to the results of previously reported studies. Results The tumor volume of liver metastases and the Ki-67 labeling index were unanimously accepted as indicators of the tumor burden and tumor aggressiveness, respectively, which are two most clinically pivotal factors for determining the strategy of systemic treatment for unresectable PanNETs. In addition, for those with a relatively small tumor burden and slow disease progression, somatostatin analogues were selected as the first-line treatment agents. For those with a relatively large tumor burden and rapid tumor progression, cytotoxic agents were selected, possibly aiming at tumor shrinkage. For those of intermediate tumor volume and/or growth rate, molecular-targeted agents were selected as the first choice. Based on this strategy discussed among the experts, we tentatively prepared a MAP for proposing optimal treatment strategy and examined its validity in some patients with unresectable PanNETs. Results validated the usefulness of this MAP proposed for patients harbouring unresectable PanNETs. Conclusion We herein propose a tentative MAP for optimal treatment selection for the patients harbouring unresectable PanNETs. Further large scale studies are, however, warranted to validate the usefulness of this MAP proposed in this study.
Article
Metastatic duodenopancreatic neuro-endocrine tumors (dpNETs) are the most important disease-related cause of death in patients with multiple endocrine neoplasia type 1 (MEN1). Nonfunctioning pNETs (NF-pNETs) are highly prevalent in MEN1 and clinically heterogeneous. Therefore, management is controversial. Data on prognostic factors for risk stratification is limited. This systematic review aims to establish the current state of evidence regarding prognostic factors in MEN1-related NF-pNETs. We systematically searched four databases for studies assessing prognostic value of any factor on NF-pNET progression, development of distant metastases, and/or overall survival. In- and exclusion, critical appraisal and data-extraction were performed independently by two authors according to pre-defined criteria. Thirteen studies (370 unique patients) were included. Prognostic factors investigated were tumor size, timing of surgical resection, WHO grade, methylation, p27/p18 expression by immunohistochemistry (IHC), ARX/PDX1 IHC and alternative lengthening of telomeres. Results were complemented with evidence from studies in MEN1-related pNET for which data could not be separately extracted for NF-pNET and data from sporadic NF-pNET. We found that the most important prognostic factors used in clinical decision making in MEN1-related NF-pNETs are tumor size and grade. NF-pNETs <2 cm may be managed with watchful waiting, while surgical resection is advised for NF-pNETs ≥2cm. Grade 2 NF-pNETs should be considered high risk. The most promising and MEN1-relevant avenues of prognostic research are multianalyte circulating biomarkers, tissue based molecular factors and imaging-based prognostication. Multi-institutional collaboration between clinical, translation and basic scientists with uniform data and biospecimen collection in prospective cohorts should advance the field.
Article
Background To evaluate the outcome of duodenopancreatic reoperations in patients with multiple endocrine neoplasia type 1 (MEN1). Methods MEN1 patients who underwent reoperations for duodenopancreatic neuroendocrine neoplasms (dpNENs) were retrieved from a prospective database and retrospectively analyzed. Results Twelve of 101 MEN1 patients underwent up to three reoperations, resulting in a total of 18 reoperations for dpNEN recurrence. Patients initially underwent either formal pancreatic resections (n = 7), enucleations (n = 3), or duodenotomy with lymphadenectomy for either NF-pNEN (seven patients), Zollinger–Ellison syndrome (ZES, three patients), organic hyperinsulinism (one patient) or VIPoma (one patient). Six patients had malignant dpNENs with lymph node (n = 5) and/or liver metastases (n = 2). The indication of reoperations was NF-pNEN (five patients), ZES (five patients), organic hyperinsulinism (one patient), and recurrent VIPoma (one patient). Median time to first reoperation was 67.5 (range 6–251) months. Five patients required a second duodenopancreatic reoperation for 60–384 months after initial surgery, and one patient underwent a third reoperation after 249 months. The rate of complications (Clavien–Dindo ≥3) was 28%. Four patients required completion pancreatectomy. Six patients developed pancreoprivic diabetes. After a median follow-up of 18 (6–34) years after initial surgery, ten of 12 patients are alive, one died of metastatic pancreatic VIPoma, and one died of metastatic thymic NEN. Conclusion Reoperations are frequently necessary for dpNEN in MEN1 patients, but are not associated with an increased perioperative morbidity in specialized centers. Organ-sparing resections should be preferred as initial duodenopancreatic procedures to maintain pancreatic function and avoid completion pancreatectomy.
Article
Objective: The aim of this study was to evaluate clinical and morphological features related to nodal involvement in appendiceal neuroendocrine tumors (NETs), to identify patients who should be referred for oncological radicalization with hemicolectomy. Background: Appendiceal NETs are usually diagnosed accidentally after appendectomy; the indications for right hemicolectomy are currently based on several parameters (ie, tumor size, grading, proliferative index, localization, mesoappendiceal invasion, lymphovascular infiltration). Available guidelines are based on scarce evidence inferred by small, retrospective, single-institution studies, resulting in discordant recommendations. Methods: A retrospective analysis of a prospectively collected database was performed. Patients who underwent surgical resection of appendiceal NETs at 11 tertiary Italian centers, from January 1990 to December 2015, were included. Clinical and morphological data were analyzed to identify factors related to nodal involvement. Results: Four-hundred fifty-seven patients were evaluated, and 435 were finally included and analyzed. Of them, 21 had nodal involvement. Grading G2 [odds ratio (OR) 6.04], lymphovascular infiltration (OR 10.17), size (OR 18.50), and mesoappendiceal invasion (OR 3.63) were related to nodal disease. Receiver operating characteristic curve identified >15.5 mm as the best size cutoff value (area under the curve 0.747). On multivariate analysis, grading G2 (OR 6.98), lymphovascular infiltration (OR 8.63), and size >15.5 mm (OR 35.28) were independently related to nodal involvement. Conclusions: Tumor size >15.5 mm, grading G2, and presence of lymphovascular infiltration are factors independently related to nodal metastases in appendiceal NETs. Presence of ≥1 of these features should be considered an indication for oncological radicalization. Although these results represent the largest study currently available, prospective validation is needed.
Article
Background: Optimal management of rectal neuroendocrine tumors is not yet well defined. Various pathologic factors, particularly tumor size, have been proposed as prognostic markers. Objective: We characterized sequential patients diagnosed with rectal neuroendocrine tumors in a population-based setting to determine whether tumor size and other pathologic markers could be useful in guiding locoregional management. Design: This study is a retrospective analysis of data from the British Columbia provincial cancer registry. Settings: The study was conducted at a tertiary care center. Patients: Sequential patients diagnosed with rectal neuroendocrine tumors between 1999 and 2011 were identified. Neuroendocrine tumors were classified as G1 and G2 tumors with a Ki-67 ≤20% and/or mitotic count ≤20 per high-power field. Main outcome measures: Baseline clinicopathologic data including TNM staging, depth of invasion, tumor size, treatment modalities, and outcomes including survival data were measured. Results: Of 91 rectal neuroendocrine tumors, the median patient age was 58 years, and 35 were men. Median tumor size was 6 mm. Median length of follow-up was 58.1 months, with 3 patients presenting with stage IV disease. Treatment included local ablation (n = 5), local excision (n = 79), surgical resection (n = 4), and pelvic radiation (n = 1; T3N1 tumor). Final margin status was positive in 17 cases. Local relapse occurred in 8 cases and 1 relapse to bone 13 months after T3N1 tumor resection. Univariate analysis demonstrated an association between local relapse and Ki-67, mitotic count, grade, and lymphovascular invasion (p < 0.01). Larger tumor size was associated with decreased disease-free survival. Limitations: Sample size was 91 patients in the whole provincial population over a 13-year time period because of the low incidence of rectal neuroendocrine tumors. Conclusions: In this population-based cohort, rectal neuroendocrine tumors generally presented with small, early tumors and were treated with local excision or surgical resection without pelvic radiation. Pathologic markers play a role in risk stratification and prognostication. See Video Abstract at http://links.lww.com/DCR/A514.