You are on page 1of 14

ENETS Consensus Guidelines

Neuroendocrinology 2016;103:172–185 Published online: January 5, 2016


DOI: 10.1159/000443167

ENETS Consensus Guidelines Update for the


Management of Distant Metastatic Disease of
Intestinal, Pancreatic, Bronchial Neuroendocrine
Neoplasms (NEN) and NEN of Unknown Primary Site
M. Pavel a D. O’Toole b F. Costa c J. Capdevila d D. Gross e R. Kianmanesh f
E. Krenning g U. Knigge h R. Salazar i U.-F. Pape a K. Öberg j
all other Vienna Consensus Conference participants
a
Department of Hepatology and Gastroenterology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin,
Berlin, Germany; b NET Centre, St. Vincent’s University and Department of Clinical Medicine, St James Hospital and Trinity
College, Dublin, Ireland; c Oncoclin Medicos Associados S/S Ltda, São Paulo, Brazil, d Institute of Oncology, Vall d’Hebron
University Hospital, Barcelona, Spain; e Department of Endocrinology and Metabolism, Hadassah University Hospital,
Mevasseret Tsion, Israel; f Department of Surgery, CHU Robert Debré, Reims, France, g Department of Internal Medicine,
Erasmus Medical Center, Rotterdam, The Netherlands; h Neuroendocrine Tumor Center of Excellence, Rigshospital,
Copenhagen University Hospital, Copenhagen, Denmark; i Institut Català d’Oncologia, Barcelona, Spain; j Department of
Medical Sciences, Endocrine Oncology Unit, University Hospital, Uppsala, Sweden

Introduction with increasing prevalence over time depending on initial


disease stage. Metastases are predominantly found in the
The goal of this paper is to update a more extensive liver and/or lymph nodes. In contrast, bone metastases
review and guidelines paper published in 2012 [1]. Gen- are reported in <15% of cases [5, 6]; however, the true
erally, any pertinent update pertaining to the diagnosis prevalence of bony metastases is probably underestimat-
and staging of individual primary tumors is provided in ed, since the reported figures are not based on the most
the relevant papers published elsewhere in this issue of sensitive imaging methods such as bone MRI or 68Ga-
updated guideline reviews. More specific issues with re- DOTATOC/NOC/TATE PET/CT. Other rare disease
spect to therapy of stage IV neuroendocrine neoplasms sites include the lung, brain and peritoneum, which have
(NEN) (focusing on grade1/2 tumors) are given below. A also been covered in guidelines [6–9].
separate guideline is provided for poorly differentiated Treatment options in metastatic disease consist of
neoplasms (grade 3 NEN). As some new large phase III liver surgery and/or locoregional and ablative therapies
trials have been published since the previous guidelines, (fig. 1). In general, these approaches are followed if ex-
this has indeed led to specific modifications in our ap- trahepatic disease is excluded or, in functional tumors,
proach to therapy. if the major tumor burden is located in the liver. Due to
Metastatic disease from NEN is very prevalent in in-
testinal and pancreatic NEN [2–4]. At initial diagnosis, For an alphabetical list of all other Vienna Consensus Conference par-
40–50% of NEN patients present with distant metastases, ticipants, see Appendix.
198.143.49.33 - 3/14/2016 7:08:07 PM

© 2016 S. Karger AG, Basel Marianne Pavel


0028–3835/16/1032–0172$39.50/0 Department of Hepatology and Gastroenterology, Campus Virchow Klinikum
Charité Universitätsmedizin Berlin
Downloaded by:

E-Mail karger@karger.com
Augustenburger Platz 1, DE–13353 Berlin (Germany)
www.karger.com/nen
E-Mail marianne.pavel @ charite.de
ENETS
Morphological and
functional imaging

Resection of primary

(a) Simple pattern of LMs (b) Complex pattern of LMs (c) Diffuse LMs
G1/G2 G1/G2 G1/G2
(unilobar or limited) (bilobar)

Or surgery Selected cases


contraindicated (<1%)

Resection Surgery One-step surgery Two-step surgery Small intestinal Pancreatic


(minor or contraindicated Major liver (1) Minor resection - SSA (IFN) - SSA (IFN)
anatomical) resection ± RFA ± RFA, RPVE, RPVL - PRRT - Chemotherapy
(2) Sequential major - Everolimus - Everolimus
liver resection - Sunitinib
- PRRT

Ablation TACE, TAE Liver


(RFA, LiTT) SIRT* transplantation
TACE, TAE
SIRT*

Fig. 1. Management of liver metastases without extrahepatic disease in G1/G2 NEN. * SIRT (selective internal
radiation therapy) is still an investigational method. LiTT = Laser-induced thermotherapy; LMs = liver metasta-
ses; RFA = radiofrequency ablation; RPVE = right portal vein embolization; RPVL = right portal vein ligation;
TACE = transarterial chemoembolization; TAE = transarterial embolization.

the rarity of the disease, the number of prospective ran- SSA. Furthermore, it has recently been reported that
domized trials is limited, and most recommendations three large randomized controlled drug trials (i.e. evero-
are based on uncontrolled studies, representing expert limus vs. placebo in lung and intestinal NET and NET
opinions. This is especially true for surgical treatment, of unknown primary tumor, RADIANT-4; 177Lu-
different locoregional or ablative therapies [emboliza- DOTATATE vs. high-dose octreotide in midgut NET,
tion, chemoembolization, radiofrequency ablation and NETTER-1, and telotristat etiprate vs. placebo in refrac-
selective internal radiation therapy (SIRT)] and system- tory carcinoid syndrome, TELESTAR) have reached
ic chemotherapy. Somatostatin analogues (SSA) and their primary endpoints [10–12]. These well-construct-
novel targeted drugs, such as the multiple tyrosine ki- ed phase III trials in NET have an impact on the current
nase inhibitor sunitinib and the mTOR inhibitor evero- treatment recommendations and therapeutic algorithm.
limus, are the only drugs that have been evaluated in In addition, there is novel information available on the
NEN within placebo-controlled trials. Based on the re- use of targeted drugs from application outside of ran-
sults of these trials, SSA, sunitinib and everolimus have domized clinical trials.
been approved and registered for antiproliferative ther- Given the variety of treatment options, the heteroge-
apy in different neuroendocrine tumor (NET) subtypes neity of NEN and the individual disease complexity, it is
excluding neuroendocrine carcinoma (NEC). Recent strongly recommended if not mandatory to discuss NEN
data from a placebo-controlled trial with lanreotide patients after accurate imaging and pathology review in a
(CLARINET study) in enteropancreatic NET have pro- multidisciplinary tumor board for appropriate therapeu-
vided novel evidence for the antiproliferative activity of tic decision making, especially to exploit surgical therapy
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 173


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
Table 1. Therapeutic options and conditions for preferential use as first-line therapy in advanced NEN

Drug Functionality Grading Primary site SSTR status Special considerations

Octreotide +/– G1 midgut + low tumor burden


Lanreotide +/– G1/G2 (–10%) midgut, + low and high (>25%) liver
pancreas tumor burden
IFN-alpha 2b +/– G1/G2 midgut if SSTR negative
STZ/5-FU +/– G1/G2 pancreas progressive in short-term* or
high tumor burden or
symptomatic
TEM/CAP +/– G2 pancreas progressive in short-term* or
high tumor burden or
symptomatic;
if STZ is contraindicated or not
available
Everolimus +/– G1/G2 lung atypical carcinoid and/or SSTR
negative
pancreas insulinoma or contraindication
for CTX
midgut if SSTR negative
Sunitinib +/– G1/G2 pancreas contraindication for CTX
PRRT +/– G1/G2 midgut + (required) extended disease; extrahepatic
disease, e.g. bone metastasis
Cisplatin§/ +/– G3 any all poorly differentiated NEC
etoposide

CAP = Capecitabine; TEM = temozolomide. * ≤6–12 months. § Cisplatin can be replaced by carboplatin.

in potentially resectable NEN patients and explore lo- Therapeutic Options


coregional therapies upfront. Choosing antiproliferative
therapies is also challenging depending on the tumor pri- In grade 1 (G1) and G2 NET, surgery with curative in-
mary, its functional status, its growth rate, grade and tent always has to be considered, even if liver and/ or
overall disease burden and the goal of individual thera- lymph node metastases are present (fig. 1). In non-resect-
pies within the patient’s choice and status. Variation of able disease, the following treatment options should be
treatment choices will also depend on physician exper- considered to control symptoms secondary to the hyper-
tise, the complexity of the treatment center and access to secretion of peptide hormones/amines leading to a func-
novel treatments. Recommendations for the preferential tional syndrome (carcinoid syndrome, diarrhea and oth-
use of targeted drugs or chemotherapy as first-line thera- er symptoms related to functionally active pancreatic
py are summarized in table 1. NEN) and/or tumor growth control. In some patients, it
This review focuses on intestinal and pancreatic NEN, may be necessary to combine therapies for example to
and it provides a therapeutic algorithm for both subtypes suppress symptoms using SSA in addition to locoregion-
(fig. 2, 3). The management of typical and atypical lung al therapies or other antiproliferative agents.
NET is similar to that of gastroenteropancreatic (GEP)
NEN taking into consideration pathological features (mi- Locoregional Therapies
totic count, Ki-67), somatostatin receptor (SSTR) expres- In the absence of any large comparative trials of dif-
sion, growth rate and disease extent. The best practice ferent locoregional or ablative therapies (bland embo-
recommendations for the management of typical and lization, chemoembolization, radioembolization, radio-
atypical bronchial NET are reported in a separate recent- frequency ablation or microwave destruction) or system-
ly published ENETS consensus paper [13]. ic treatment, the choice of treatment is based on
individual patient features (e.g. size, distribution and
198.143.49.33 - 3/14/2016 7:08:07 PM

174 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
Refractory
CS and SD
Consider debulking surgery of LM (see fig. 1)
Octreotide
CS or Refractory Consider locoregional/ablative therapy (see fig. 1)
Lanreotide CS and/or PD or SSA dose increase
or add-on IFN-alpha 2b
or pasireotide or a clinical trial
or PRRT
Complete Resect primary
resection if and metastases
Advanced feasible (G1/G2) (see fig. 1)
loco- Consider octreotide or lanreotide
regional (if prior watch and wait)
disease or or increase of SSA dose
Non-functional
distant Watch and wait
(G1, low tumor or locoregional therapy
metastases or
burden, no
Octreotide or lanreotide ध PD ध or PRRT (if SSTR positive)
symptoms, SD)
or everolimus
Non-functional Octreotide or lanreotide or IFN-alpha 2b
(G2 and/or high
tumor burden,
or PD or Everolimus or
symptoms) SSTR negative IFN-alpha or
Locoregional therapy

FOLFOX or
Cisplatin* + FOLFIRI or
NEC, G3 ध PD ध
Etoposide TEM/CAP or
Clinical trial

Fig. 2. Therapeutic algorithm for the management of intestinal (midgut) NEN with advanced locoregional disease
and/or distant metastases. CS = Carcinoid syndrome; LM = liver metastasis; PD = progressive disease; SD = sta-
ble disease; TEM/CAP = temozolomide/capecitabine. * Cisplatin may be replaced by carboplatin.

number of liver lesions, vascularization, proliferative in- surgery may be considered in patients with non-function-
dex) and local physicians’ expertise [14]. Locoregional al tumors if the disease is not progressive over a 6-month
therapies should be exploited early, following SSA thera- period and the patients are suffering from symptoms re-
py, to prevent carcinoid crisis in functionally active NET lated to tumor burden. Although some retrospective
(especially midgut NET with classical carcinoid syn- studies indicate that surgery for liver metastasis is associ-
drome), and they may be an alternative option to system- ated with improved survival [19–22], it remains unclear
ic therapies in patients with non-functional tumors if the whether debulking surgery is of benefit in asymptomatic
disease is limited to the liver. Locoregional therapies may patients, since comparative trials to systemic therapy are
be considered repetitively during the course of the dis- lacking. Even if surgery is performed with curative intent,
ease. There is consensus that SIRT is still investigational, there is a high rate of disease recurrence within 3–5 years
and that a comparative trial of SIRT to bland emboliza- [4, 23]. In patients with carcinoid syndrome, it is impor-
tion is required, as well as more safety data on long-term tant to control the hypersecretion of serotonin with SSA
tolerability of SIRT to establish this procedure for the prior to surgery, in order to prevent carcinoid crisis.
management of NEN [14–18].
Liver Transplantation
Debulking Surgery Transplantation is generally not recommended as a
This is an alternative option to locoregional therapies treatment option in advanced NEN; it may be an option
and could be considered in patients with uncontrolled in highly selected patients with carcinoid syndrome or
functional tumors, especially in patients with carcinoid other functional NET and extended liver disease, early
syndrome, refractory insulinoma, glucagonoma or vipo- refractory to multiple systemic treatments including SSA,
ma or PTH-related peptide-secreting tumors. Debulking interferon (IFN)-alpha, locoregional therapies and pep-
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 175


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
tide receptor-targeted radiotherapy (PRRT) [4]. A precise with Cushing’s disease or acromegaly. In a phase II trial,
preselection of patients (e.g. well-differentiated NET, the 27% of patients with carcinoid syndrome showed symp-
exclusion of extrahepatic disease by optimized staging, tom improvement with pasireotide following failure with
low serum total bilirubin) for liver transplantation may standard doses of octreotide LAR [32]; however, in a
increase the 5-year survival rates in patients with NET comparative trial, pasireotide LAR 60 mg was not supe-
undergoing liver transplantation [24–26]. rior to octreotide LAR 40 mg/month [33]. Since there are
limited treatment options available in refractory carci-
Minimal Consensus Statement on Therapeutic noid syndrome, pasireotide might be considered in indi-
Options vidual highly selected patients when other treatments
failed or are not feasible depending on accessibility, and
Surgery with curative intent and/or locoregional or ablative this includes locoregional therapies, debulking surgery,
therapies should be considered at initial diagnosis and during the
course of the disease as an alternative approach to systemic ther- IFN-alpha and novel drugs in clinical trials.
apies. In patients with functional NET, all liver-directed thera- Telotristat etiprate, an oral serotonin synthesis inhibi-
pies require the prior initiation of SSA therapy (or other specific tor, is a potential novel option in refractory carcinoid syn-
symptom-controlling measures). Debulking surgery is indicated drome [34, 35]. In a phase III placebo controlled trial
in selected patients with functional NET with predominant liver (TELESTAR), telotristat etiprate significantly reduced di-
disease for improved syndrome control, even if the liver tumor
burden can be reduced by <90%. Liver transplantation is an op- arrhea in patients with refractory carcinoid syndrome
tion in highly selected patients, preferably in young patients with while on SSA [12]. If approved, telotristat etiprate can be
functional syndromes demonstrating early resistance to medical recommended in addition to SSA for refractory diarrhea
therapy. in carcinoid syndrome patients.

SSA for Tumor Growth Control


Systemic Therapy SSA are an established therapy for antiproliferative
purposes in intestinal NET, based on 2 placebo-con-
SSA and Novel Compounds for Syndrome Control trolled trials (the PROMID study and the CLARINET
SSA are first-line therapy in functionally active NEN study). Both drugs, octreotide LAR and lanreotide auto-
including tumors associated with the carcinoid syn- gel, are recommended as first-line systemic therapy in
drome and functionally active endocrine pancreatic midgut NET to control tumor growth [36, 37]. There
NET (such as vipoma and glucagonoma). The commer- is consensus that SSA can be used as first-line systemic
cially available agents octreotide and lanreotide are therapy in pancreatic NET (Ki-67 <10%) in view of
considered equally effective for symptom control. In lack of toxicity, and although the antiproliferative effects
general, long-acting formulations (octreotide LAR 10– of SSA are considered a drug class effect, based on the
30 mg i.m. per month; lanreotide autogel 60–120 mg CLARINET study, lanreotide autogel should preferably
deeply s.c. per month) are used over a medium- to long- be used in pancreatic NET, since prospective data on the
term period. Initiating therapy with a lower dose of the use of octreotide LAR in pancreatic NET are lacking [37,
long-acting formulations or with octreotide 50–100 μg 38]. There are retrospective data supporting the use of
s.c. for 7–10 days twice to thrice per day is recommend- octreotide LAR in low-grade pancreatic NET [39].
ed [27–29], particularly in patients with severe symp- SSA can be recommended for the prevention or inhi-
toms. bition of tumor growth in both intestinal and pancreatic
In case of refractory syndrome, dose escalation above NET. Equally, based on the CLARINET study, the use of
the upper labeled dosages is an option [30, 31]. In gen- SSA in GEP NET is recommended up to a Ki-67 of 10%
eral, dose escalation is performed by shortening the injec- [37]. However, for the overall group of NEN, there was
tion interval from 4 to 3 weeks with long-acting SSA. no consensus among experts on a clear cut-off value for
There is consensus that dose escalation can be recom- the recommendation of SSA for antiproliferative purpos-
mended in refractory carcinoid syndrome for improve- es. When considering SSA as first-line therapy in intesti-
ment of symptoms. nal or pancreatic NET, some experts feel that 5% might
Pasireotide is a novel universal somatostatin ligand be a more appropriate Ki-67 cut-off threshold. Prospec-
that binds to 4 of 5 SSTR and that is not approved for the tive validation is required to determine the appropriate
treatment of carcinoid syndrome or other functional Ki-67 value for treatment stratification to SSA or more
NEN, but for the treatment of pituitary tumors associated aggressive therapies.
198.143.49.33 - 3/14/2016 7:08:07 PM

176 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
The recommendation for the use of SSA expands to level of evidence for the use of lanreotide autogel in pancreatic
patients with higher hepatic tumor burden (>25% liver NET; and based on the respective study designs, octreotide LAR
is approved for tumor control in midgut NET, whereas lanreo-
involvement) as supported by a subgroup analysis from tide autogel is approved for enteropancreatic NET. SSA may be
the CLARINET study [37]. Although no benefit in overall considered in low-grade NET of other sites. There is no estab-
survival could be demonstrated by the placebo-controlled lished Ki-67 threshold for the use of SSA, preferably SSA should
trials with SSA that allowed cross-over from the placebo be used if Ki-67 is ≤10%.
arm upon progression to open-label SSA, it is expected
that the use of SSA has an impact on the outcome of pa- Interferon-Alpha
tients [40]. It remains, however, controversial if SSA IFN-alpha is a second-line therapy in NEN that are
should be started at initial diagnosis or after the observa- functionally active. It is recommended to use IFN-alpha
tion of spontaneous tumor growth and be initiated in case as add-on therapy to SSA therapy in functional tumors.
that disease progression occurs. There is consensus that The recommended dose of IFN-alpha 2b is 3 × 3 to 3 ×
SSA should be started at diagnosis in cases of high liver 5 MU/week s.c. [28, 45]. In patients who do not tolerate
tumor burden and extended disease, since these are worse the conventional regimen, alternatively pegylated IFN-
prognostic factors. Another factor in favor of early SSA alpha (50–180 μg/week s.c.) can also be used [46]. IFN-
therapy is a pancreatic primary, given the fact that the alpha has antiproliferative activity and may be considered
overall 5-year survival rate in stage IV disease does not for antiproliferative purposes if other approved drugs are
exceed 40–60% [41, 42]. There is no data to support con- unavailable especially in midgut NEN. IFN-alpha has
tinued use of SSA when patients progress on SSA (they been explored in comparison to bevacizumab for antip-
may be required, however, for the continued suppression roliferative purposes in a large randomized trial of 400
of functionally active tumors). patients with carcinoids (including different primary
SSA may also be used in NET of other sites (e.g. rectal sites) who received octreotide LAR concomitantly (SWOG
or bronchial NET), when the SSTR status is positive (on trial); the primary endpoint, median progression-free sur-
somatostatin imaging or histology), if the tumor is slowly vival (PFS), was not different between those taking IFN-
growing, G1 or G2 and preferably with Ki-67 <10%. alpha and those taking bevacizumab [47]. This study,
Prospective ongoing clinical trials need to further evalu- however, confirms the antiproliferative activity of IFN-
ate the role of SSA (lanreotide autogel and pasireotide, alpha 2b in advanced G1/G2 NET, NET with progressive
respectively) in typical and atypical lung NEN (www. disease or with other poor prognostic features with a me-
clinicaltrials.gov). dian PFS of 15.4 months reached in the IFN-alpha arm.
SSA may be considered in SSTR-negative NEN, if a
small volume disease is present and it is expected that im- Minimal Consensus Statement
aging may have provided false negative information on
SSTR status. Immunostaining with SSTR2 antibodies IFN-alpha is an established and approved therapy for syn-
drome control, and primarily used as second-line (add-on) ther-
may also be useful [43, 44]. apy in refractory carcinoid syndrome or functional pancreatic
NET. IFN is an option for inhibiting tumor growth and, due to
Minimal Consensus Statement on Systemic Therapy limited therapy options in midgut NET, it may be considered an
antiproliferative option (less so in pancreatic NET).
SSA, octreotide and lanreotide, are effective drugs for syn-
drome control in functional NET. In refractory carcinoid syn-
drome or with insufficient syndrome control in pancreatic NET, Novel Targeted Drugs
a dose escalation of SSA may be recommended. The novel SSA Novel targeted drugs (everolimus and sunitinib) are
pasireotide might be considered in refractory carcinoid syn- approved for pancreatic NET based on the results of
drome in case all other treatment options including ablative pro-
two placebo-controlled trials on progressive pancreatic
cedures, transarterial embolization and IFN-alpha have failed,
and there is no clinical trial available. If approved, the oral sero- NET. The median PFS is around 11 months with either
tonin synthesis inhibitor telotristat etiprate will offer a novel of the drugs, while tumor remission occurs in 5% and
treatment option in refractory carcinoid syndrome. <10% of the patients with everolimus and sunitinib, re-
For antiproliferative purposes, SSA may be used in stable or spectively. The use of either everolimus or sunitinib is
progressive disease or in patients with unknown tumor behavior.
recommended in progressive G1/G2 pancreatic NET,
SSA are recommended as a first-line therapy in midgut NET and
can be considered in pancreatic NET as a first-line therapy (up irrespective of Ki-67 and tumor burden. The standard
to a Ki-67 of 10%). While the antiproliferative efficacy of both dose for everolimus is 10 mg/day and for sunitinib 37.5
available SSA is considered a drug class effect, there is a higher mg/day as continuous treatment. Side effects may re-
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 177


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
quire a dose reduction to 5 mg/day for everolimus or to imaging has an impact on therapy allocation to either
25 mg/day for sunitinib [48, 49]. While comparative everolimus or PRRT after failure of SSA.
data of both drugs are lacking, the selection of the tar- In the absence of approved drugs in metastatic lung
geted drug is based on the medical history of the patient, NET, everolimus may be recommended as a first-line
the side effect profile of the drug and accessibility to the therapy in progressive disease. However, in patients with
treatment. low proliferative activity (G1, typical carcinoid) with
Targeted drugs, everolimus or sunitinib, are one of the strong SSTR expression on imaging, SSA may be consid-
different treatment options in pancreatic NET and may ered as a first-line therapy. Although comprehensive clin-
be used as first- or second-line options with respect to ical data are lacking for the use of SSA in lung NET, it is
chemotherapy or subsequent to SSA therapy (table 1). Al- expected that the clinical behavior of typical carcinoids
though targeted drugs may be the first therapy choice in (mitotic count <2/10 HPF; G1 NET) is similar to G1 NET
pancreatic NET, there is consensus that targeted drugs of other sites. Ongoing and planned clinical trials (LUNA;
should not be broadly used as first-line therapy for their lanreotide vs. placebo) will further elucidate the role of
potential toxicity. There is no evidence on the exact se- SSA in advanced lung NET (www.clinicaltrials.gov).
quencing of different treatment options in pancreatic There is not sufficient data to support the use of other
NET. Potential toxicity needs to be considered when se- targeted drugs including bevacizumab, sorafenib, pazo-
quencing therapies, as indicated in a retrospective multi- panib or axitinib in either pancreatic or non-pancreatic
center study on 169 patients from Italy, where a mark- NEN. These drugs as well as sunitinib in midgut NET
edly increased toxicity was reported with everolimus in (SUNLAND study) are currently explored in prospective
patients previously treated either with PRRT and/or che- randomized clinical trials, but their results are not yet
motherapy [50]. In contrast, a smaller retrospective study available and their use should be restricted to clinical
from the Netherlands on 24 patients indicated that the trials.
safety of everolimus is not influenced by previous PRRT It is standard practice to combine targeted drugs with
[51]. An ongoing trial (SEQTOR) is currently investigat- SSA in functionally active NEN. The aim of a combina-
ing the antiproliferative efficacy of everolimus versus tion therapy of everolimus and SSA may not only be tu-
streptozotocin with 5-fluorouracil (STZ/5-FU) in pro- mor growth inhibition, but also improved syndrome con-
gressive pancreatic NET in a crossover design upon pro- trol, e.g. in patients with recurrent hypoglycemia related
gression (www.clinicaltrials.gov). to metastatic insulinoma. Although prospective trials
Everolimus can be recommended in advanced NET of with everolimus are lacking to demonstrate an improve-
non-pancreatic origin in case of disease progression (e.g. ment of hormone-related syndromes, the early use of
NET of intestinal or lung origin). It can be used in midgut everolimus may be justified to avoid hospitalization and
NET as second- or third-line therapy after failure of SSA sequelae related to hypoglycemia based on the experience
and/or IFN-alpha or PRRT. This recommendation is in few patients (fig. 3).
based on the results of the RADIANT-4 trial [10] that Although there might be a rationale to combine tar-
reached its primary endpoint and demonstrates superior geted drugs with SSA also in non-functional NET, given
PFS with everolimus compared to placebo in non-func- the SSTR expression in the majority of NET patients,
tional NET of intestinal or lung origin; and it is supported there is no robust evidence yet that the combination ther-
by the RADIANT-2 trial on advanced NET associated apy of targeted drugs with SSA is superior to monother-
with the carcinoid syndrome (that tended towards simi- apy with either everolimus or sunitinib for antiprolif-
lar results) [52]. erative purposes. A comparative trial on progressive pan-
The sequencing of everolimus as second- or third-line creatic NET (COOPERATE-2) with everolimus versus
therapy for advanced intestinal NET also depends on oth- everolimus and pasireotide, a novel SSA with a broader
er issues, including accessibility of PRRT. Individual pa- binding affinity to SSTR compared to first-generation
tient selection is important. A strong SSTR expression on SSA, failed to demonstrate superiority of the combination
imaging is necessary to achieve better results with PRRT, therapy with respect to PFS [53]. Although there might
while extensive hepatic and/or bone disease as well as de- be a potential benefit of a combination therapy using oth-
creased kidney function may limit its use. Otherwise, the er SSA, such as lanreotide or octreotide, and a recent
use of everolimus may be limited by comorbidities such open-label phase II study indicates favorable response
as uncontrolled diabetes or lung diseases. A comprehen- (disease control rate >90%) with everolimus in combi-
sive review of the patients’ medical history, pathology and nation with octreotide in a first-line setting in GEP NET
198.143.49.33 - 3/14/2016 7:08:07 PM

178 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
Refractory
syndrome
Diazoxide (insulinoma) Consider debulking surgery of LM (see fig. 1)
PPI (gastrinoma)
Functional
Octreotide or lanreotide‡ Consider locoregional*/ablative therapy (see fig. 1)
activity
or IFN-alpha 2b (if SSTR or SSA dose increase
negative) or add-on IFN-alpha 2b (if not already receiving)
or everolimus (insulinoma)‡
Complete Resect primary or PRRT
resection if and metastases
Advanced feasible (G1/G2) (see fig. 1)
loco-
regional
disease or Everolimus or sunitinib
distant Non-functional
metastases (G1, low G2§, or cytotoxic chemotherapy#
Lanreotide (octreotide)
low tumor
or ध PD ध ध PD ध
burden, SD or or locoregional therapies* PRRT**
Watch and wait
initial diagnosis, or
no symptoms) or lanreotide (octreotide) 2nd-line
(if prior watch and wait) CTX
or
Non-functional Clinical
(G2, high tumor trial
Cytotoxic Everolimus or
burden, and/or PD ध PD
chemotherapy# Sunitinib
PD or
symptoms)

Cisplatin† +
G3 NEC FOLFOX or
Etoposide
G3 NEN PD ध FOLFIRI or
G3 NET
STZ/5-FU Clinical trial
or TEM/CAP

Fig. 3. Therapeutic algorithm for the management of pancreatic ogy. 5-FU = 5-Fluorouracil; CS = carcinoid syndrome; CTX = che-
NEN with advanced locoregional disease and/or distant metasta- motherapy; LM = liver metastasis; PD = progressive disease; SD =
ses. § Ki-67 <5–10%; * locoregional therapies are contraindicated stable disease; TEM/CAP = temozolomide/capecitabine. The term
after Whipple procedure; # recommended chemotherapy includes ‘or’ indicates that the use of the other options at further progres-
STZ/5-FU or STZ/doxorubicin; TEM/CAP is an alternative che- sion should be considered, e.g. patients with G1 or low-grade G2
motherapy regimen if STZ-based chemotherapy is not available; NET and/ or low tumor burden who received everolimus may be
** if SSTR imaging is positive; ‡ patients should be closely moni- treated with standard cytotoxic chemotherapy upon progression
tored for paradoxical reaction (increasing hypoglycemia); † cispla- before unapproved drugs, second-line chemotherapy or a clinical
tin may be replaced by carboplatin; G3 NET is coined for tumors trial is considered.
with Ki-67 >20% but well- or moderately differentiated morphol-

[54], in the absence of a comparative study of targeted tinib are generally recommended after failure of SSA or chemo-
drugs with either octreotide or lanreotide compared to therapy in pancreatic NET. In intestinal NET, everolimus may be
used as a second-line therapy after failure of SSA or as a third-line
the targeted drug alone, the upfront combination therapy therapy after failure of PRRT; while in progressive lung NET,
of targeted drugs with SSA cannot be recommended. Fur- everolimus is recommended as a first-line therapy, unless SSA
thermore, data are lacking to support the use of SSA be- may be considered as a first-line therapy (e.g. in typical carcinoid
yond progression in combination with targeted drugs. with slow growth expressing SSTR). The combined use of tar-
geted drugs with SSA for antiproliferative purpose is not recom-
mended in non-functional NET. Antiangiogenic drugs including
Minimal Consensus Statement sunitinib are not recommended in non-pancreatic NEN outside
of clinical trials.
Everolimus and sunitinib are approved antiproliferative ther-
apies in progressive pancreatic NET, and they represent one of
the different treatment options next to SSA and systemic chemo-
therapy. They can be considered as a first-line therapy, especially Systemic Chemotherapy
if SSA is not an option, and if systemic chemotherapy is not clini- Systemic chemotherapy is indicated in progressive or
cally required, not feasible or not tolerated. Everolimus or suni- bulky pancreatic NET and in G3 NEN. The term G3 NEN
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 179


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
comprises well- or moderately differentiated tumors with After failure of STZ-based chemotherapy in pancre-
Ki-67 >20% that are not termed in the WHO 2010 clas- atic NET, the following are alternative chemotherapeutic
sification and large or small cell tumors with Ki-67 >20% options: temozolomide +/– capecitabine and oxaliplatin-
(G3 NEC; presented in detail elsewhere). Chemotherapy based chemotherapy + 5-FU or capecitabine. It remains
may be considered in NET of other sites (lung, thymus, unclear which treatment option is superior; however, in
stomach, colon or rectum) under certain conditions [e.g. pancreatic NET there are data supporting the preferential
when Ki-67 is at a high level (upper G2 range), in rapidly use of temozolomide +/– capecitabine with promising re-
progressive disease and/or after failure of other therapies, sponse rates and a low toxicity profile [62, 67, 68].
or if SSTR imaging is negative]. Systemic chemotherapy is not recommended in non-
Chemotherapy is one of different treatment options pancreatic NET unless G2 NET (Ki-67 >15%), tumors
in pancreatic NET and can be used in G1 or G2 tumors. displaying aggressive biological behavior (RECIST pro-
Cytotoxic therapy combinations include: STZ/5-FU (an gression in 3–6 months) or in those which are SSTR neg-
established therapy) and doxorubicin with STZ as an ative. Metronomic chemotherapy may be an option using
alternative option; however, the use of doxorubicin is temozolomide and/or capecitabine +/– SSA in G2 NET
limited by a cumulative dose of 500 mg/m2 (due to the or in SSTR-negative NET, or capecitabine + bevacizumab
risk of cardiotoxicity). Therapeutic regimens can be after failure of other treatments (such as locoregional
recommended according to Moertel et al. (cycles every therapies, IFN-alpha or everolimus) [69–72]. Given the
6 weeks) or Fjallskog et al. (cycles every 3 weeks) [55– limited treatment options in bronchial carcinoids, temo-
57]. Data do not support three-drug regimen associa- zolomide is a therapeutic option based on data from small
tions including cisplatin, nor the replacement of 5-FU studies [73, 74]. Prospective validation is needed, as well
by capecitabine [58–60]. Systemic chemotherapy may as evaluation of the best sequencing of therapies in bron-
be considered without prior progression in patients chial NET including SSA, everolimus and temozolomide.
with high tumor burden. There is no established Ki-67 In G3 NEC, cisplatin-based chemotherapy (e.g. cispla-
cut-off value for the recommendation of chemotherapy. tin/etoposide) is standard therapy and recommended as a
Patients with pancreatic NET with Ki-67 of 5–20% can first-line therapy (see guidelines on poorly differentiated
be treated with chemotherapy. Other factors that favor tumors). Cisplatin might be replaced by carboplatin, based
chemotherapy compared to targeted drugs include: on the data from the Nordic NEC trial [75]. Although ob-
bulky disease; a symptomatic patient; rapid tumor pro- jective remission rates are high (40–67%), the median PFS
gression in ≤6–12 months, and patients with a possible is limited with 4–6 months [76–78]. Second-line systemic
chance of achieving a response to allow for surgery therapy options include FOLFOX and FOLFIRI [79, 80],
(neoadjuvant option). while topotecan is not effective in G3 NEC [81]. Temo-
Although replacing STZ/5-FU by temozolomide/ zolomide-based chemotherapy should be preferably used
capecitabine is gaining popularity, this approach cannot in pancreatic G3 NET or in gastrointestinal NEC with Ki-
be categorically recommended, since data for temozo- 67 <55% [67, 78]; prospective studies are underway to as-
lomide are still limited. However, temozolomide +/– sess the activity of temozolomide in this setting. Targeted
capecitabine may be considered as an alternative regimen drugs in combination with chemotherapy are under evalu-
depending on the availability of STZ/5-FU. Reported ob- ation in clinical trials on G3 NEN. Further details on the
jective response rates from small prospective and retro- management of G3 NEN, including recommendations for
spective studies achieved with temozolomide either com- different primary tumor sites, are summarized in a recent-
bined with antiangiogenic drugs or capecitabine range ly published comprehensive review on G3 NEC and are
between 15 and 70% [61–63]. The value of temozolomide provided in a separate consensus paper [82, 83].
either as mono- or combination therapy with capecitabi-
ne or antiangiogenic drugs is further explored in prospec- Minimal Consensus Statement
tive clinical trials (www.clinicaltrials.gov). Few studies in-
dicate that the MGMT status is correlated with tumor re- STZ-based chemotherapy is one of the treatment options in
sponse to alkylating agents [64–66]; however, determin- pancreatic G1/G2 NET next to SSA and novel targeted drugs. It
ing MGMT expression or methylation can currently not is preferably recommended in patients with higher tumor burden
with or without associated clinical symptoms and/or in patients
be recommended as selection criterion for the use of che- with significant tumor progression in ≤6–12 months. Although
motherapy in NEN, since studies are small, and prospec- data for temozolomide-based chemotherapy are still limited, it
tive validation is lacking. may replace the STZ/5-FU regimen in case this is not available,
198.143.49.33 - 3/14/2016 7:08:07 PM

180 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
in pancreatic NET and it may be considered in G3 NET and in localized in the intestine or the lung. Additional tools
high-risk NET of other primary site (e.g. pulmonary NET). In G3 should be exploited to identify the primary tumor. These
NEC, platinum-based chemotherapy is recommended as a first-
line therapy. include immunohistochemistry of transcription factors
(CDX-2, Islet-1, TTF-1) [88], PET/CT (e.g. Ga68-SR, 11C-
5-hydroxytryptophan or 18F-DOPA) [89, 90] and upper
Peptide Receptor Radionuclide Therapy and lower gastrointestinal endoscopy and optionally cap-
PRRT is a therapeutic option in progressive SSTR-pos- sule endoscopy [91, 92]. If the primary tumor site remains
itive NET with homogenous SSTR expression (all lesions unknown, therapeutic decision making is essentially
are positive). In general, the use of PRRT follows failed based on grading, functionality, SSTR status, tumor ex-
first-line medical therapy. Radionuclide therapy with ei- tent and hepatic tumor burden.
ther 90Y and/or 177Lu-labeled SSA is most frequently used Further information is provided in the consensus
in NET, but 177Lu-labelled SSA is increasingly used due guideline updates for other GEP NET [83, 93–97, this is-
to lower kidney toxicity. The minimum requirements sue].
for PRRT are reported in a separate consensus guideline
[84]. Until recently, there were no results from prospec-
tive randomized trials available. The registrational trial Appendix
of 177Lu-DOTATATE in progressive midgut NET
All Other Vienna Consensus Conference Participants
(NETTER-1) has reached its primary endpoint with a sig- Anlauf, M. (Institut für Pathologie und Zytologie, St. Vincenz
nificant prolongation of PFS compared to high-dose oc- Krankenhaus, Limburg, Germany); Bartsch, D.K. (Department of
treotide (60 mg/month). Based on this trial, and cumula- Surgery, Philipps University, Marburg, Germany); Baudin, E. (In-
tive data from prospective and retrospective trials over stitut Gustave Roussy, Villejuif, France); Caplin, M. (Neuroendo-
the last 15 years, PRRT may be recommended in midgut crine Tumour Unit, Royal Free Hospital, London, UK); Cwikla,
J.B. (Department of Radiology, Faculty of Medical Sciences, Uni-
NET as a second-line therapy after failure of SSA if the versity of Warmia and Mazury, Olsztyn, Poland); De Herder,
general requirements for applying PRRT are fulfilled [84– W.W. (Department of Internal Medicine, Division of Endocrinol-
87] or as a third-line therapy after failure of everolimus. ogy, Erasmus Medical Center, Rotterdam, The Netherlands); Del-
Given the different established and approved therapeu- le Fave, G. (Department of Digestive and Liver Disease, Ospedale
tic options in pancreatic NET and the lack of a prospective Sant’Andrea, Rome, Italy); Eriksson, B. (Department of Endocrine
Oncology, University Hospital, Uppsala, Sweden); Falconi, M.
trial with PRRT in pancreatic NET, PRRT (if available) is (Department of Surgery, San Raffaele Hospital, Università Vita e
in general recommended in G1/G2 NET after failure of Salute, Milan, Italy); Ferolla, P. (NET Center, Umbria Regional
medical therapy including SSA, chemotherapy or novel Cancer Network, Università degli Studi di Perugia, Perugia, Italy);
targeted drugs. However, potential increasing toxicity, e.g. Ferone, D. (Department of Endocrine and Metabolic Sciences,
after prior chemotherapy or targeted therapy, needs to be University of Genoa, Genoa, Italy); Garcia-Carbonero, R. (Medical
Oncology Department, Hospital Universitario Doce de Octubre,
considered, requires close surveillance and might justify Madrid, Spain); Ito, T. (Pancreatic Diseases Branch, Kyushu Uni-
an earlier use of PRRT in selected patients (table 1). versity Hospital, Fukuoka, Japan); Jensen, R.T. (Digestive Diseases
Branch, NIH, Bethesda, Md., USA); Kaltsas, G. (Department of
Minimal Consensus Statement Pathophysiology, Division of Endocrinology, National University
of Athens, Athens, Greece); Kelestimur, F. (Department of Endo-
PRRT is recommended after failure of medical therapy. Data crinology, Erciyes University Medical School, Kayseri, Turkey);
from a prospective trial in midgut NET support its role as a second- Kos-Kudla, B. (Department of Endocrinology, Medical University
line therapy option in intestinal NET if the general requirements of Silesia, Katowice, Poland); Kwekkeboom, D. (Department of
for PRRT are fulfilled and as an alternative option to everolimus. Internal Medicine, Division of Nuclear Medicine, Erasmus Medi-
The optimal sequencing with targeted drugs and/or chemotherapy cal Center, Rotterdam, The Netherlands); Niederle, B. (Depart-
needs to be defined in pancreatic NET when data from prospective ment of Surgery, Medical University of Vienna, Vienna, Austria);
randomized trials with PRRT in pancreatic NET become available. O’Connor, J. (Department of Clinical Oncology, Institute Alexan-
der Fleming, Buenos Aires, Argentina); Pascher, A. (Department
of Visceral and Transplant Surgery, Campus Virchow Klinikum,
Charité Universitätsmedizin Berlin, Berlin, Germany); Perren, A.
Management of NET with Unknown Primary Tumor (Institute of Pathology, University of Bern, Bern, Switzerland); Ra-
mage, J.K. (Gastroenterology Department, Hampshire Hospitals
NHS Trust, Hampshire, UK); Raymond, E. (Oncologie Médicale,
In approximately 13% of patients who are diagnosed Hôpitaux Universitaires Paris Nord Val de Seine, Paris, France);
as having NEN, the primary site is not known. In patients Reed, N. (Beatson Oncology Centre, Gartnavel General Hospital,
with unknown primary tumor, the site is most frequently Glasgow, UK); Rindi, G. (Institute of Anatomic Pathology, Poli-
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 181


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
clinico A. Gemelli, Università Cattolica del Sacro Cuore, Rome, Lijnden, The Netherlands); Toumpanakis, C. (Neuroendocrine
Italy); Ruszniewski, P. (Department of Gastroenterology, Beaujon Tumour Unit, Royal Free Hospital, London, UK); Weber, W. (De-
Hospital, Clichy, France); Sedlackova, E. (Department of Oncol- partment of Radiology, Memorial Sloan Kettering Cancer Center,
ogy, First Faculty of Medicine and General Teaching Hospital, New York, N.Y., USA); Wiedenmann, B. (Department of Hepatol-
Prague, Czech Republic); Sorbye, H. (Department of Oncology, ogy and Gastroenterology, Campus Virchow Klinikum, Charité
Haukeland University Hospital, Bergen, Norway); Sundin, A. (De- Universitätsmedizin Berlin, Germany); Zheng-Pei, Z. (Depart-
partment of Radiology, Section for Molecular Imaging, University ment of Endocrinology, Peking Union Medical College Hospital,
Hospital, Uppsala, Sweden); Taal, B. (Netherlands Cancer Centre, Beijing, China).

References
1 Pavel M, Baudin E, Couvelard A, Krenning E, 8 Pavel M, Grossman A, Arnold R, Perren A, 13 Caplin ME, Baudin E, Ferolla P, Filosso P,
Öberg K, Steinmüller T, Anlauf M, Wieden- Kaltsas G, Steinmüller T, et al: ENETS con- Garcia-Yuste M, Lim E, Oberg K, Pelosi G,
mann B, Salazar R; Barcelona Consensus sensus guidelines for the management of Perren A, Rossi RE, Travis WD; ENETS con-
Conference participants: ENETS consensus brain, cardiac and ovarian metastases from sensus conference participants: Pulmonary
guidelines for the management of patients neuroendocrine tumors. Neuroendocrinolo- neuroendocrine (carcinoid) tumors: Europe-
with liver and other distant metastases from gy 2010;91:326–332. an Neuroendocrine Tumor Society expert
neuroendocrine neoplasms of foregut, mid- 9 Kianmanesh R, Ruszniewski P, Rindi G, consensus and recommendations for best
gut, hindgut, and unknown primary. Neuro- Kwekkeboom D, Pape UF, Kulke M, et al: practice for typical and atypical pulmonary
endocrinology 2012;95:157–176. ENETS consensus guidelines for the manage- carcinoids. Ann Oncol 2015;26:1604–1620.
2 Niederle MB, Hackl M, Kaserer K, Niederle B: ment of peritoneal carcinomatosis from neu- 14 de Baere T, Deschamps F, Tselikas L, Ducreux
Gastroenteropancreatic neuroendocrine tu- roendocrine tumors. Neuroendocrinology M, Planchard D, Pearson E, Berdelou A, Le-
mours: the current incidence and staging 2010;91:333–340. boulleux S, Elias D, Baudin E: GEP-NETS
based on the WHO and European Neuroen- 10 Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi update: interventional radiology: role in the
docrine Tumour Society classification: an C, Wolin E, Tomasek J, Raderer M, Lahner H, treatment of liver metastases from GEP-
analysis based on prospectively collected pa- Voi M, Pacaud LB, Rouyrre N, Sachs C, Valle NETs. Eur J Endocrinol 2015; 172:R151–
rameters. Endocr Relat Cancer 2010; 17: 909– JW, Fave GD, Van Cutsem E, Tesselaar M, R166.
918. Shimada Y, Oh DY, Strosberg J, Kulke MH, 15 Fiore F, Del Prete M, Franco R, Marotta V,
3 Lawrence B, Gustafsson BI, Chan A, Svejda B, Pavel ME; RAD001 in Advanced Neuroendo- Ramundo V, Marciello F, Di Sarno A, Carratù
Kidd M, Modlin IM: The epidemiology of crine Tumours, Fourth Trial (RADIANT-4) AC, de Luca di Roseto C, Colao A, Faggiano
gastroenteropancreatic neuroendocrine tu- Study Group: Everolimus for the treatment of A: Transarterial embolization (TAE) is equal-
mors. Endocrinol Metab Clin North Am advanced, non-functional neuroendocrine ly effective and slightly safer than transarte-
2011;40:1–18, vii. tumours of the lung or gastrointestinal tract rial chemoembolization (TACE) to manage
4 Frilling A, Modlin IM, Kidd M, Russell C, (RADIANT-4): a randomised, placebo-con- liver metastases in neuroendocrine tumors.
Breitenstein S, Salem R, Kwekkeboom D, Lau trolled, phase 3 study. Lancet 2015, Epub Endocrine 2014;47:177–182.
WY, Klersy C, Vilgrain V, Davidson B, Siegler ahead of print. 16 Maire F, Lombard-Bohas C, O’Toole D, Vul-
M, Caplin M, Solcia E, Schilsky R; Working 11 Strosberg J, Wolin E, Chasen B, Kulke M, lierme MP, Rebours V, Couvelard A, Pelletier
Group on Neuroendocrine Liver Metastases: Bushnell D, Caplin M, Baum RP, Mittra E, AL, Zappa M, Pilleul F, Hentic O, Hammel P,
Recommendations for management of pa- Hobday T, Hendifar A, Oberg K, Lopera Ruszniewski P: Hepatic arterial embolization
tients with neuroendocrine liver metastases. Sierra M, Ruszniewski P, Kwekkeboom D: versus chemoembolization in the treatment
Lancet Oncol 2014;15:e8–e21. 177-Lu-Dotatate significantly improves pro- of liver metastases from well-differentiated
5 Van Loon K, Zhang L, Keiser J, Carrasco C, gression-free survival in patients with midgut midgut endocrine tumors: a prospective ran-
Glass K, Ramirez MT, Bobiak S, Nakakura neuroendocrine tumours: results of the phase domized study. Neuroendocrinology 2012;
EK, Venook AP, Shah MH, Bergsland EK: III NETTER-1 trial . Eur J Cancer 2015; 96:294–300.
Bone metastases and skeletal-related events 51(suppl 3):S710. 17 Engelman ES, Leon-Ferre R, Naraev BG,
from neuroendocrine tumors. Endocr Con- 12 Kulke MH, Hörsch D, Caplin M, Anthony L, Sharma N, Sun S, O’Dorisio TM, Howe J, But-
nect 2015;4:9–17. Bergsland E, Oberg K, Welin S, Warner R, ton A, Zamba G, Halfdanarson TR: Compar-
6 Kos-Kudla B, O’Toole D, Falconi M, Gross D, Lombard-Bohas C, Kunz P, Grande E, Valle ison of transarterial liver-directed therapies
Klöppel G, Sundin A, et al: ENETS consensus J, Fleming D, Lapuerta P, Banks P, Jackson for low-grade metastatic neuroendocrine tu-
guidelines for the management of bone and S, Wheeler D, Zambrowicz B, Sands A, Pav- mors in a single institution. Pancreas 2014;43:
lung metastases from neuroendocrine tu- el M: Telotristat etiprate is effective in treat- 219–225.
mors. Neuroendocrinology 2010;91:341–350. ing patients with carcinoid syndrome that is 18 Benson AB 3rd, Geschwind JF, Mulcahy MF,
7 O’Toole D, Rindi G, Plöckinger U, Wieden- inadequately controlled by somatostatin an- Rilling W, Siskin G, Wiseman G, Cunning-
mann B: ENETS consensus guidelines for the alog therapy (the phase 3 TELESTAR clini- ham J, Houghton B, Ross M, Memon K, An-
management of patients with rare metastases cal trial). Eur J Cancer 2015; 51(suppl 3): drews J, Fleming CJ, Herman J, Nimeiri H,
from digestive neuroendocrine tumors: ratio- S728. Lewandowski RJ, Salem R: Radioembolisa-
nale and working framework. Introduction. tion for liver metastases: results from a pro-
Neuroendocrinology 2010;91:324–325. spective 151 patient multi-institutional phase
II study. Eur J Cancer 2013;49:3122–3130.
198.143.49.33 - 3/14/2016 7:08:07 PM

182 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
19 Franko J, Feng W, Yip L, Genovese E, Moser 31 Al-Efraij K, Aljama MA, Kennecke HF: As- 39 Jann H, Denecke T, Koch M, Pape UF, Wie-
AJ: Non-functional neuroendocrine carcino- sociation of dose escalation of octreotide denmann B, Pavel M: Impact of octreotide
ma of the pancreas: incidence, tumor biology, long-acting release on clinical symptoms and long-acting release on tumour growth control
and outcomes in 2,158 patients. J Gastrointest tumor markers and response among patients as a first-line treatment in neuroendocrine tu-
Surg 2010;14:541–548. with neuroendocrine tumors. Cancer Med mours of pancreatic origin. Neuroendocri-
20 Yuan CH, Wang J, Xiu DR, Tao M, Ma ZL, 2015;4:864–870. nology 2013;98:137–143.
Jiang B, Li ZF, Li L, Wang L, Wang H, Zhang 32 Kvols LK, Oberg KE, O’Dorisio TM, Mohi- 40 Shen C, Shih YC, Xu Y, Yao JC: Octreotide
TL: Meta-analysis of liver resection versus deen P, de Herder WW, Arnold R, Hu K, long-acting repeatable use among elderly pa-
nonsurgical treatments for pancreatic neuro- Zhang Y, Hughes G, Anthony L, Wieden- tients with carcinoid syndrome and survival
endocrine tumors with liver metastases. Ann mann B: Pasireotide (SOM230) shows effica- outcomes: a population-based analysis. Can-
Surg Oncol 2016;23:244–249. cy and tolerability in the treatment of patients cer 2014;120:2039–2049.
21 Watzka FM, Fottner C, Miederer M, Schad A, with advanced neuroendocrine tumors re- 41 Rindi G, Falconi M, Klersy C, Albarello L,
Weber MM, Otto G, Lang H, Musholt TJ: Sur- fractory or resistant to octreotide LAR: results Boninsegna L, Buchler MW, et al: TNM stag-
gical therapy of neuroendocrine neoplasm from a phase II study. Endocr Relat Cancer ing of neoplasms of the endocrine pancreas:
with hepatic metastasis: patient selection and 2012;19:657–666. results from a large international cohort
prognosis. Langenbecks Arch Surg 2015;400: 33 Wolin E, Jarzab B, Eriksson B, Walter T, study. J Natl Cancer Inst 2012;104:764–777.
349–358. Toumpanakis C, Morse M, Tomassetti P, et al: 42 Yao JC, Hassan M, Phan A, Dagohoy C, Leary
22 Mayo SC, Herman JM, Cosgrove D, Bhagat N, Phase III study of pasireotide long-acting re- C, Mares JE, Abdalla EK, Fleming JB, Vauthey
Kamel I, Geschwind JF, Pawlik TM: Emerging lease in patients with metastatic neuroendo- JN, Rashid A, Evans DB: One hundred years
approaches in the management of patients crine tumors and carcinoid symptoms refrac- after ‘carcinoid’: epidemiology of and prog-
with neuroendocrine liver metastasis: role of tory to available somatostatin analogues. nostic factors for neuroendocrine tumors in
liver-directed and systemic therapies. J Am Drug Des Devel Ther 2015;9:5075–5086. 35,825 cases in the United States. J Clin Oncol
Coll Surg 2013;216:123–134. 34 Kulke MH, O’Dorisio T, Phan A, Bergsland E, 2008;26:3063–3072.
23 Sarmiento JM, Heywood G, Rubin J, Ilstrup Law L, Banks P, Freiman J, Frazier K, Jackson 43 Kaemmerer D, Peter L, Lupp A, Schulz S, Sän-
DM, Nagorney DM, Que FG: Surgical treat- J, Yao JC, Kvols L, Lapuerta P, Zambrowicz B, ger J, Prasad V, Kulkarni H, Haugvik SP,
ment of neuroendocrine metastases to the liv- Fleming D, Sands A: Telotristat etiprate, a Hommann M, Baum RP: Molecular imaging
er: a plea for resection to increase survival. J novel serotonin synthesis inhibitor, in pa- with 68Ga-SSTR PET/CT and correlation to
Am Coll Surg 2003;197:29–37. tients with carcinoid syndrome and diarrhea immunohistochemistry of somatostatin re-
24 Nobel YR, Goldberg DS: Variable use of mod- not adequately controlled by octreotide. En- ceptors in neuroendocrine tumours. Eur J
el for end-stage liver disease exception points docr Relat Cancer 2014;21:705–714. Nucl Med Mol Imaging 2011;38:1659–1668.
in patients with neuroendocrine tumors met- 35 Pavel M, Hörsch D, Caplin M, Ramage J, 44 Papotti M, Bongiovanni M, Volante M, Allìa
astatic to the liver and its impact on patient Seufferlein T, Valle J, Banks P, Lapuerta P, E, Landolfi S, Helboe L, Schindler M, Cole SL,
outcomes. Transplantation 2015;99:2341– Sands A, Zambrowicz B, Fleming D, Wieden- Bussolati G: Expression of somatostatin re-
2346. mann B: Telotristat etiprate for carcinoid syn- ceptor types 1–5 in 81 cases of gastrointestinal
25 Chung H, Chapman WC: Liver transplanta- drome: a single-arm, multicenter trial. J Clin and pancreatic endocrine tumors. A correla-
tion for metastatic neuroendocrine tumors. Endocrinol Metab 2015;100:1511–1519. tive immunohistochemical and reverse-tran-
Adv Surg 2014;48:235–252. 36 Rinke A, Müller HH, Schade-Brittinger C, scriptase polymerase chain reaction analysis.
26 Fan ST, Le Treut YP, Mazzaferro V, Bur- Klose KJ, Barth P, Wied M, Mayer C, Aminos- Virchows Arch 2002;440:461–475.
roughs AK, Olausson M, Breitenstein S, Frill- sadati B, Pape UF, Bläker M, Harder J, Arnold 45 Oberg K: Interferon-alpha versus somatosta-
ing A: Liver transplantation for neuroendo- C, Gress T, Arnold R; PROMID Study Group: tin or the combination of both in gastro-
crine tumour liver metastases. HPB (Oxford) Placebo-controlled, double-blind, prospec- enteropancreatic tumours. Digestion 1996;
2015;17:23–28. tive, randomized study on the effect of octreo- 57(suppl 1):81–83.
27 Kvols LK: Metastatic carcinoid tumors and tide LAR in the control of tumor growth in 46 Pavel ME, Baum U, Hahn EG, Schuppan D,
the malignant carcinoid syndrome. Ann NY patients with metastatic neuroendocrine Lohmann T: Efficacy and tolerability of pe-
Acad Sci 1994;733:464–470. midgut tumors: a report from the PROMID gylated IFN-alpha in patients with neuroen-
28 Eriksson B, Klöppel G, Krenning E, Ahlman Study Group. J Clin Oncol 2009; 27: 4656– docrine gastroenteropancreatic carcinomas. J
H, Plöckinger U, Wiedenmann B, et al: Con- 4663. Interferon Cytokine Res 2006;26:8–13.
sensus guidelines for the management of pa- 37 Caplin ME, Pavel M, Ćwikła JB, Phan AT, Ra- 47 Yao J, Guthrie K, Moran C, Strosberg J, Kulke
tients with digestive neuroendocrine tumors derer M, Sedláčková E, Cadiot G, Wolin EM, M, Chan J, et al: SWOG S0518: phase III pro-
– well-differentiated jejunal-ileal tumor/car- Capdevila J, Wall L, Rindi G, Langley A, Mar- spective randomized comparison of depot oc-
cinoma. Neuroendocrinology 2008;87:8–19. tinez S, Blumberg J, Ruszniewski P; CLARI- treotide plus interferon alpha-2b versus depot
29 Ruszniewski P, Ish-Shalom S, Wymenga M, NET Investigators: Lanreotide in metastatic octreotide plus bevacizumab (NSC #704865)
O’Toole D, Arnold R, Tomassetti P, et al: Rap- enteropancreatic neuroendocrine tumors. N in advanced, poor prognosis carcinoid pa-
id and sustained relief from the symptoms of Engl J Med 2014;371:224–233. tients (NCT00569127). J Clin Oncol 2015;
carcinoid syndrome: results from an open 38 Martín-Richard M, Massutí B, Pineda E, 33(suppl):abstr 4004.
6-month study of the 28-day prolonged-re- Alonso V, Marmol M, Castellano D, Fonseca 48 Raymond E, Dahan L, Raoul JL, Bang YJ, Bor-
lease formulation of lanreotide. Neuroendo- E, Galán A, Llanos M, Sala MA, Pericay C, bath I, Lombard-Bohas C, et al: Sunitinib ma-
crinology 2004;80:244–251. Rivera F, Sastre J, Segura A, Quindós M, Mai- late for the treatment of pancreatic neuroendo-
30 Strosberg JR, Benson AB, Huynh L, Duh MS, sonobe P; TTD (Tumores del Tracto Digesti- crine tumors. N Engl J Med 2011;364:501–513.
Goldman J, Sahai V, Rademaker AW, Kulke vo) Study Group: Antiproliferative effects of 49 Yao JC, Shah MH, Ito T, Bohas CL, Wolin
MH: Clinical benefits of above-standard dose lanreotide autogel in patients with progres- EM, Van Cutsem E, et al; RAD001 in Ad-
of octreotide LAR in patients with neuroen- sive, well-differentiated neuroendocrine tu- vanced Neuroendocrine Tumors, Third Trial
docrine tumors for control of carcinoid syn- mours: a Spanish, multicentre, open-label, (RADIANT-3) Study Group: Everolimus for
drome symptoms: a multicenter retrospective single arm phase II study. BMC Cancer 2013; advanced pancreatic neuroendocrine tumors.
chart review study. Oncologist 2014; 19: 930– 13:427. N Engl J Med 2011;364:514–523.
936.
198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 183


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS
50 Panzuto F, Rinzivillo M, Fazio N, de Braud F, 60 Turner NC, Strauss SJ, Sarker D, Gillmore R, 69 Brizzi MP, Berruti A, Ferrero A, Milanesi E,
Luppi G, Zatelli MC, et al: Real-world study of Kirkwood A, Hackshaw A, Papadopoulou A, Volante M, Castiglione F, Birocco N, Bom-
everolimus in advanced progressive neuroen- Bell J, Kayani I, Toumpanakis C, Grillo F, baci S, Perroni D, Ferretti B, Alabiso O, Ciuf-
docrine tumors. Oncologist 2014;19:966–974. Mayer A, Hochhauser D, Begent RH, Caplin freda L, Bertetto O, Papotti M, Dogliotti L:
51 Kamp K, Gumz B, Feelders RA, Kwekkeboom ME, Meyer T: Chemotherapy with 5-fluoro- Continuous 5-fluorouracil infusion plus long
DJ, Kaltsas G, Costa FP, de Herder WW: Safe- uracil, cisplatin and streptozocin for neuroen- acting octreotide in advanced well-differenti-
ty and efficacy of everolimus in gastrointesti- docrine tumours. Br J Cancer 2010;102:1106– ated neuroendocrine carcinomas. A phase II
nal and pancreatic neuroendocrine tumors 1112. trial of the Piemonte oncology network. BMC
after (177)Lu-octreotate. Endocr Relat Can- 61 Chan JA, Stuart K, Earle CC, Clark JW, Bhar- Cancer 2009;9:388.
cer 2013;20:825–831. gava P, Miksad R, Blaszkowsky L, Enzinger 70 Koumarianou A, Kaltsas G, Kulke MH, Oberg
52 Pavel ME, Hainsworth JD, Baudin E, Peeters PC, Meyerhardt JA, Zheng H, Fuchs CS, Kul- K, Strosberg JR, Spada F, Galdy S, Barberis
M, Hörsch D, Winkler RE, et al; RADIANT-2 ke MH: Prospective study of bevacizumab M, Fumagalli C, Berruti A, Fazio N: Temo-
Study Group: Everolimus plus octreotide plus temozolomide in patients with advanced zolomide in advanced neuroendocrine neo-
long-acting repeatable for the treatment of neuroendocrine tumors. J Clin Oncol 2012; plasms: pharmacological and clinical aspects.
advanced neuroendocrine tumours associat- 30:2963–2968. Neuroendocrinology 2015;101:274–288.
ed with carcinoid syndrome (RADIANT-2): 62 Strosberg JR, Fine RL, Choi J, Nasir A, Cop- 71 Mitry E, Walter T, Baudin E, Kurtz JE,
a randomised, placebo-controlled, phase 3 pola D, Chen DT, Helm J, Kvols L: First-line Ruszniewski P, Dominguez-Tinajero S, Ben-
study. Lancet 2011;378:2005–2012. chemotherapy with capecitabine and temo- grine-Lefevre L, Cadiot G, Dromain C, Farace
53 Kulke MH Ruszniewski P, Van Cutsem E, zolomide in patients with metastatic pancre- F, Rougier P, Ducreux M: Bevacizumab plus
Valle J, De Herder W, Pavel M, Degtyarev E, atic endocrine carcinomas. Cancer 2011; 117: capecitabine in patients with progressive ad-
Lam D, et al: A randomized open-label phase 268–275. vanced well-differentiated neuroendocrine
II study of everolimus alone or in combina- 63 Kulke MH, Stuart K, Enzinger PC, Ryan DP, tumors of the gastro-intestinal (GI-NETs)
tion with pasireotide LAR in advanced, pro- Clark JW, Muzikansky A, Vincitore M, Mi- tract (BETTER trial) – a phase II non-ran-
gressive pancreatic neuroendocrine tumors chelini A, Fuchs CS: Phase II study of temo- domised trial. Eur J Cancer 2014; 50: 3107–
(pNET): COOPERATE-2 Trial. 12th Annual zolomide and thalidomide in patients with 3115.
ENETS Conference for the Diagnosis and metastatic neuroendocrine tumors. J Clin 72 Berruti A, Fazio N, Ferrero A, Brizzi MP, Vo-
Treatment of Neuroendocrine Tumor Dis- Oncol 2006;24:401–406. lante M, Nobili E, Tozzi L, Bodei L, Torta M,
ease, March 11–13, 2015, Barcelona, Spain. 64 Walter T, van Brakel B, Vercherat C, Hervieu D’Avolio A, Priola AM, Birocco N, Amoroso
54 Bajetta E, Catena L, Fazio N, Pusceddu S, V, Forestier J, Chayvialle JA, Molin Y, Lom- V, Biasco G, Papotti M, Dogliotti L: Bevaci-
Biondani P, Blanco G, et al: Everolimus in bard-Bohas C, Joly MO, Scoazec JY: O6- zumab plus octreotide and metronomic
combination with octreotide long-acting re- Methylguanine-DNA methyltransferase sta- capecitabine in patients with metastatic well-
peatable in a first-line setting for patients with tus in neuroendocrine tumours: prognostic to-moderately differentiated neuroendocrine
neuroendocrine tumors: an ITMO group relevance and association with response to tumors: the XELBEVOCT study. BMC Can-
study. Cancer 2014;120:2457–2463. alkylating agents. Br J Cancer 2015; 112: 523– cer 2014;14:184.
55 Kouvaraki MA, Ajani JA, Hoff P, Wolff R, 531. 73 Crona J, Fanola I, Lindholm DP, Antonodi-
Evans DB, Lozano R, Yao JC: Fluorouracil, 65 Schmitt AM, Pavel M, Rudolph T, Dawson H, mitrakis P, Öberg K, Eriksson B, Granberg D:
doxorubicin, and streptozocin in the treat- Blank A, Komminoth P, Vassella E, Perren A: Effect of temozolomide in patients with meta-
ment of patients with locally advanced and Prognostic and predictive roles of MGMT static bronchial carcinoids. Neuroendocri-
metastatic pancreatic endocrine carcinomas. protein expression and promoter methylation nology 2013;98:151–155.
J Clin Oncol 2004;22:4762–4771. in sporadic pancreatic neuroendocrine neo- 74 Ekeblad S, Sundin A, Janson ET, Welin S,
56 Moertel CG, Lefkopoulo M, Lipsitz S, Hahn plasms. Neuroendocrinology 2014; 100: 35– Granberg D, Kindmark H, Dunder K, Kozlo-
RG, Klaassen D: Streptozocin-doxorubicin, 44. vacki G, Orlefors H, Sigurd M, Oberg K,
streptozocin-fluorouracil or chlorozotocin in 66 Kulke MH, Stuart K, Enzinger PC, Ryan DP, Eriksson B, Skogseid B: Temozolomide as
the treatment of advanced islet-cell carcino- Clark JW, Muzikansky A, Vincitore M, Mi- monotherapy is effective in treatment of ad-
ma. N Engl J Med 1992;326:519–523. chelini A, Fuchs CS, Kulke MH, Hornick JL, vanced malignant neuroendocrine tumors.
57 Fjallskog ML, Janson ET, Falkmer UG, Vatn Frauenhoffer C, Hooshmand S, Ryan DP, Clin Cancer Res 2007;13:2986–2991.
MH, Oberg KE, Eriksson BK: Treatment with Enzinger PC, Meyerhardt JA, Clark JW, Stu- 75 Sorbye H, Welin S, Langer SW, Vestermark
combined streptozotocin and liposomal doxo- art K, Fuchs CS, Redston MS: O6-methylgua- LW, Holt N, Osterlund P, Dueland S, Hof-
rubicin in metastatic endocrine pancreatic tu- nine DNA methyltransferase deficiency and sli E, Guren MG, Ohrling K, Birkemeyer E,
mors. Neuroendocrinology 2008;88:53–58. response to temozolomide-based therapy in Thiis-Evensen E, Biagini M, Gronbaek H,
58 Dilz LM, Denecke T, Steffen IG, Prasad V, patients with neuroendocrine tumors. Clin Soveri LM, Olsen IH, Federspiel B, Assmus J,
von Weikersthal LF, Pape UF, Wiedenmann Cancer Res 2009;15:338–345. Janson ET, Knigge U: Predictive and prog-
B, Pavel M: Streptozocin/5-fluorouracil che- 67 Welin S, Sorbye H, Sebjornsen S, Knappskog nostic factors for treatment and survival in
motherapy is associated with durable re- S, Busch C, Oberg K: Clinical effect of temo- 305 patients with advanced gastrointestinal
sponse in patients with advanced pancreatic zolomide-based chemotherapy in poorly dif- neuroendocrine carcinoma (WHO G3): the
neuroendocrine tumours. Eur J Cancer 2015; ferentiated endocrine carcinoma after pro- NORDIC NEC study. Ann Oncol 2013; 24:
51:1253–1262. gression on first-line chemotherapy. Cancer 152–160.
59 Meyer T, Qian W, Caplin ME, Armstrong G, 2011;117:4617–4622. 76 Moertel CG, Kvols LK, O’Connell MJ, Rubin
Lao-Sirieix SH, Hardy R, Valle JW, Talbot DC, 68 Fine RL, Gulati AP, Krantz BA, Moss RA, J: Treatment of neuroendocrine carcinomas
Cunningham D, Reed N, Shaw A, Navalkis- Schreibman S, Tsushima DA, Mowatt KB, with combined etoposide and cisplatin. Evi-
soor S, Luong TV, Corrie PG: Capecitabine Dinnen RD, Mao Y, Stevens PD, Schrope B, dence of major therapeutic activity in the ana-
and streptozocin ± cisplatin in advanced gas- Allendorf J, Lee JA, Sherman WH, Chabot JA: plastic variants of these neoplasms. Cancer
troenteropancreatic neuroendocrine tu- Capecitabine and temozolomide (CAPTEM) 1991;68:227–232.
mours. Eur J Cancer 2014;50:902–911. for metastatic, well-differentiated neuroen-
docrine cancers: the Pancreas Center at Co-
lumbia University experience. Cancer Che-
mother Pharmacol 2013;71:663–670.
198.143.49.33 - 3/14/2016 7:08:07 PM

184 Neuroendocrinology 2016;103:172–185 Pavel  et al.


 

DOI: 10.1159/000443167
Downloaded by:
ENETS
77 Sorbye H, Strosberg J, Baudin E, Klimstra DS, 85 Imhof A, Brunner P, Marincek N, Briel M, 92 Wang SC, Parekh JR, Zuraek MB, Venook
Yao JC: Gastroenteropancreatic high-grade Schindler C, Rasch H, Mäcke HR, Rochlitz C, AP, Bergsland EK, Warren RS, Nakakura EK:
neuroendocrine carcinoma. Cancer 2014; Müller-Brand J, Walter MA: Response, sur- Identification of unknown primary tumors in
120:2814–2823. vival, and long-term toxicity after therapy patients with neuroendocrine liver metasta-
78 Heetfeld M, Chougnet CN, Olsen IH, Rinke with the radiolabeled somatostatin analogue ses. Arch Surg 2010;145:276–280.
A, Borbath I, Crespo G, Barriuso J, Pavel M, [90Y-DOTA]-TOC in metastasized neuroen- 93 Delle Fave G, O’Toole D, Sundin A, Taal B,
O’Toole D, Walter T; other Knowledge Net- docrine cancers. J Clin Oncol 2011; 29: 2416– Ferolla P, Ramage JK, Ferone D, Ito T, Weber
work members: Characteristics and treat- 2423. W, Zheng-Pei Z, De Herder WW, Pascher A,
ment of patients with G3 gastroenteropancre- 86 Bushnell DL Jr, O’Dorisio TM, O’Dorisio MS, Ruszniewski P; all other Vienna Consensus
atic neuroendocrine neoplasms. Endocr Relat Menda Y, Hicks RJ, Van Cutsem E, Baulieu Conference participants: ENETS consensus
Cancer 2015;22:657–664. JL, Borson-Chazot F, Anthony L, Benson AB, guidelines update for gastroduodenal neuro-
79 Hadoux J, Malka D, Planchard D, Scoazec JY, Oberg K, Grossman AB, Connolly M, Bouter- endocrine neoplasms. Neuroendocrinology
Caramella C, Guigay J, Boige V, Leboulleux S, fa H, Li Y, Kacena KA, LaFrance N, Pauwels 2016;103:119–124.
Burtin P, Berdelou A, Loriot Y, Duvillard P, SA: 90Y-edotreotide for metastatic carcinoid 94 Niederle B, Pape UF, Costa F, Gross D, Kele-
Chougnet CN, Déandréis D, Schlumberger refractory to octreotide. J Clin Oncol 2010;28: stimur F, Knigge U, Öberg K, Pavel M, Perren
M, Borget I, Ducreux M, Baudin E: Post-first- 1652–1659. A, Toumpanakis C, O’Connor J, O’Toole D,
line FOLFOX chemotherapy for grade 3 neu- 87 Kwekkeboom DJ, de Herder WW, Kam BL, Krenning E, Reed N, Kianmanesh R; all other
roendocrine carcinoma. Endocr Relat Cancer van Eijck CH, van Essen M, Kooij PP, Feelders Vienna Consensus Conference participants:
2015;22:289–298. RA, van Aken MO, Krenning EP: Treatment ENETS consensus guidelines update for neu-
80 Hentic O, Hammel P, Couvelard A, Rebours with the radiolabeled somatostatin analog roendocrine neoplasm of the jejunum and
V, Zappa M, Palazzo M, Maire F, Goujon G, [177 Lu-DOTA 0, Tyr3]octreotate: toxicity, ileum. Neuroendocrinology 2016; 103:125–
Gillet A, Lévy P, Ruszniewski P: FOLFIRI reg- efficacy, and survival. J Clin Oncol 2008; 26: 138.
imen: an effective second-line chemotherapy 2124–2130. 95 Ramage JK, De Herder WW, Delle Fave G,
after failure of etoposide-platinum combina- 88 Lin X, Saad RS, Luckasevic TM, Silverman JF, Ferolla P, Ferone D, Ito T, Ruszniewski P,
tion in patients with neuroendocrine carcino- Liu Y: Diagnostic value of CDX-2 and TTF-1 Sundin A, Weber W, Zheng-Pei Z, Taal B,
mas grade 3. Endocr Relat Cancer 2012; 19: expressions in separating metastatic neuroen- Pascher A; all other Vienna Consensus Con-
751–757. docrine neoplasms of unknown origin. Appl ference participants: ENETS consensus
81 Olsen IH, Knigge U, Federspiel B, Hansen CP, Immunohistochem Mol Morphol 2007; 15: guidelines update for colorectal neuroendo-
Skov A, Kjær A, Langer SW: Topotecan 407–414. crine neoplasms. Neuroendocrinology 2016;
monotherapy in heavily pretreated patients 89 Nakamoto Y, Sano K, Ishimori T, Ueda M, 103:139–143.
with progressive advanced stage neuroendo- Temma T, Saji H, Togashi K: Additional in- 96 Pape UF, Niederle B, Costa F, Gross D, Kele-
crine carcinomas. J Cancer 2014;5:628–632. formation gained by positron emission to- stimur F, Kianmanesh R, Knigge U, Öberg K,
82 Ilett E, Langer SW, Olsen I, Federspiel B, Kjær mography with (68)Ga-DOTATOC for sus- Pavel M, Perren A, Toumpanakis C, O’Connor
A, Knigge U: Neuroendocrine carcinomas of pected unknown primary or recurrent neuro- J, Krenning E, Reed N, O’Toole D; all other
the gastroenteropancreatic system: a compre- endocrine tumors. Ann Nucl Med 2015; 29: Vienna Consensus Conference participants:
hensive review. Diagnostics 2015;5:119–176. 512–518. ENETS consensus guidelines for neuroendo-
83 Garcia-Carbonero R, Sorbye H, Baudin E, 90 Tan TH, Lee BN, Hassan SZ: Diagnostic value crine neoplasms of the appendix (excluding
Raymond E, Wiedenmann B, Niederle B, Sed- of (68)Ga-DOTATATE PET/CT in liver me- goblet cell carcinomas). Neuroendocrinology
lackova E, Toumpanakis C, Anlauf M, Cwikla tastases of neuroendocrine tumours of un- 2016;103:144–152.
JB, Caplin M, O’Toole D, Perren A; all other known origin. Nucl Med Mol Imaging 2014; 97 Falconi M, Eriksson B, Kaltsas G, Bartsch DK,
Vienna Consensus Conference participants: 48:212–215. Capdevila J, Caplin M, Kos-Kudla B, Kwek-
ENETS consensus guidelines for high-grade 91 Frilling A, Smith G, Clift AK, Martin J: Cap- keboom D, Rindi G, Klöppel G, Reed N, Kian-
gastroenteropancreatic neuroendocrine tu- sule endoscopy to detect primary tumour site manesh R, Jensen RT; all other Vienna Con-
mors and neuroendocrine carcinomas. Neu- in metastatic neuroendocrine tumours. Dig sensus Conference participants: ENETS con-
roendocrinology 2016;103:186–194. Liver Dis 2014;46:1038–1042. sensus guidelines update for the management
84 Kwekkeboom DJ, Krenning EP, Lebtahi R, of patients with functional pancreatic neuro-
Komminoth P, Kos-Kudła B, de Herder WW, endocrine tumors and non-functional pan-
et al: ENETS consensus guidelines for the creatic neuroendocrine tumors. Neuroendo-
standards of care in neuroendocrine tumors: crinology 2016;103:153–171.
peptide receptor radionuclide therapy with
radiolabeled somatostatin analogs. Neuroen-
docrinology 2009;90:220–226.

198.143.49.33 - 3/14/2016 7:08:07 PM

ENETS Consensus Guidelines Update for Neuroendocrinology 2016;103:172–185 185


Distant Metastatic Disease DOI: 10.1159/000443167
Downloaded by:
ENETS

You might also like