Low- to intermediate-grade neuroendocrine tumor (NET) constitutes a group of indolent malignancies that share the capacity for secreting hormones and neuroamines. Until recently, there were few therapeutic options for oncologic control. The PROMID study showed that octreotide long-acting repeatable formulation can delay tumor growth in midgut NETs. And, recent phase III studies showed both everolimus and sunitinib improved progression-free survival in pancreatic NETs, validating the phosphoinositide 3-kinase/Akt/mTOR pathway and angiogenesis as important targets for further advances. Ongoing and planned pivotal studies targeting these pathways in other NET subtypes may widen their therapeutic application. Development of rational combinations may further improve therapeutic outcome. These successes and our improved understanding of the underlying molecular biology are likely to lead to further important advances on the horizon. Clin Cancer Res; 18(7); 1830–6. ©2012 AACR.

Neuroendocrine tumors (NET) are generally classified into low- to intermediate-grade versus high-grade tumors on the basis of pathology. Low- to intermediate-grade NETs, the focus of this review, are either functional or nonfunctional, depending on whether the tumor secretes bioactive substances that lead to clinical symptoms such as flushing and diarrhea. NETs are generally further divided into carcinoid and pancreatic NETs. Carcinoids develop from neuroendocrine cells at any location in the body and are grouped according to their embryonic origin: the foregut (lungs, thymus, stomach, and duodenum); the midgut (jejunum, ileum, appendix, and proximal large bowel); and the hindgut (distal colon and rectum; ref. 1). It is estimated that 64% of all NETs originated in the gastrointestinal tract and 28% originated in the lung (2). Pancreatic NET, traditionally called islet-cell carcinoma, represents 1.3% of all malignancies arising from the pancreas (3).

The incidence of NETs has increased significantly, from 1.09 to 5.25 per 100,000 individuals between 1973 and 2004 (4). A large, retrospective, case–control study identified only family history of malignancy as a significant risk factor (5). Pancreatic NETs are rare, occurring at an estimated incidence of 3 per 1,000,000 individuals (4). Infrequently, NETs arise as part of certain genetic cancer syndromes, including multiple endocrine neoplasia type 1, von Hippel-Lindau syndrome, neurofibromatosis, and tuberous sclerosis (TSC; ref. 6). Most NETs, however, are sporadic.

Apart from the tumor–node–metastasis staging system endorsed by the American Joint Committee on Cancer in 2010, tumor grade and primary site are important prognostic determinants of cancer-related outcomes (7–9). Low-grade NETs follow a relatively indolent course. The cells are well differentiated with a uniform appearance. They produce secretory granules and often express chromogranin A (CGA) and synaptophysin. On the other hand, high-grade NETs are aggressive tumors. Cells are poorly differentiated, appearing in a sheet-like structure, and usually nonfunctional. Indices of cell proliferation include the number of mitoses per 10 high-power microscopic fields or the percentage of tumor cells with immunostaining for Ki67. Because management of high-grade NETs fundamentally differs from that of low- and intermediate-grade NETs, uniformed reporting of grade is encouraged.

Imaging modalities frequently used for NETs include computed tomography (CT), MRI, and [111In-DTPA0]octreotide scintigraphy (10). Because of the vascular nature of NETs, tumors generally enhance intensely with i.v. contrast during the early arterial phase, with washout during the delayed venous phase. Therefore, multiphasic CT is recommended. MRI is a good modality to assess liver metastases, which are usually hypointense on T1-weighted images and hyperintense on T2-weighted images (11). Scintigraphy using the radiolabeled somatostatin analogue, octreotide, can detect NETs that express somatostatin receptor types 2 and 5, with an overall sensitivity of 80% to 90% (12, 13). Although scintigraphy provides useful information about the site of disease, it does not give accurate information about the size of tumor. 2[18F]fluoro-2-deoxy-D-glucose position emission tomography (FDG-PET) has been used in evaluating NETs. Although its applicability for low-grade NETs is limited because of their low metabolic activity, the use of FDG-PETs in intermediate- and high-grade NETs is still being evaluated.

NETs are characterized by their ability to produce bioactive peptides and neuroamines, such as serotonin, histamine, prostaglandins, substance P, insulin, gastrin, glucagon, and vasoactive intestinal polypeptide (14, 15). Most patients with early-stage NETs are asymptomatic; some may present with nonspecific abdominal symptoms because of the local effect of serotonin or the mechanical effect of the primary tumor. Patients with advanced-stage NETs can manifest symptoms of carcinoid syndrome, which classically consists of facial flushing, diarrhea, and occasionally bronchospastic wheezing. Carcinoid syndrome can be debilitating and is usually seen in the setting of advanced or metastatic disease.

Somatostatin (SST) analogues, such as octreotide and lanreotide, block sst2 and sst5, effectively reducing the release of bioactive peptides and neuroamines (Fig. 1). These agents are used for the management of carcinoid syndrome or symptoms of hormonal overproduction from NETs. Octreotide comes in 2 forms, the long-acting repeatable form and the short-acting rescue form. It is generally well tolerated. Side effects, including cholelithiasis, hypothyroidism, bradycardia, and hyperglycemia, are generally manageable, infrequently affecting the patients' quality of life.

Figure 1.

Targeting critical signaling pathways in NETs. Recent data from pivotal phase III studies have shown the importance of somatostatin, mTOR, and angiogenic pathways in NETs. RER, rough endoplasmic reticulum.

Figure 1.

Targeting critical signaling pathways in NETs. Recent data from pivotal phase III studies have shown the importance of somatostatin, mTOR, and angiogenic pathways in NETs. RER, rough endoplasmic reticulum.

Close modal

IFN-α binds to receptors on NET cells and can lead to degradation of secretory peptide and tumor growth suppression. It has been used to control carcinoid syndrome with or without octreotide (16–18). Because it has delayed onset of response and more side effects compared with octreotide, so it is not usually the first-line agent for symptomatic patients with NETs.

High-grade or poorly differentiated NETs are treated with platinum-based chemotherapy. For low- to intermediate-grade NETs, surgical resection of the primary tumor offers cure of localized disease; however, complete metastectomy has been associated with longer survival among patients with advanced disease and should be considered when possible (19, 20). Systemic therapy options for tumor control remain limited for NETs.

Systemic therapy for pancreatic NETs

Streptozocin-based chemotherapy was approved by the U.S. Food and Drug Administration for the treatment of pancreatic NET nearly 3 decades ago. Analyses of 2 large case series using Response Evaluation Criteria of Solid Tumors criteria have reported objective tumor response rates of 39% and 38%, respectively (21, 22).

In genetic cancer syndromes (TSC-2, neurofibromatosis-1, and von Hippel-Lindau syndrome), somatic mutations identified using an exome-sequencing approach, and expression profiling have consistently implicated a dysfunction of the mTOR pathway as a critical event in pancreatic NETs (Fig. 1; refs. 23–25). Everolimus, an oral inhibitor of mTOR, was studied in a multinational double-blind placebo-controlled phase III study (Table 1; ref. 26). The study showed that everolimus significantly prolonged median progression-free survival (PFS) from 4.6 to 11 months compared with placebo [HR = 0.35; 95% confidence interval (CI), 0.27–0.45; P < 0.0001]. Everolimus also significantly reduced insulin, glucagon, and gastrin secretions among patients with functional pancreatic NETs (27, 28).

Table 1.

Recent and ongoing phase III studies in advanced NETs

RegimenNMedian PFS and/or TTP (months)P-valueStudy status and reference
Pancreatic NETs 
Sunitinib 171 11.4 PFS 0.0001a Study completed 
Placebo  5.5 PFS  Raymond et al., 2011 (29) 
Everolimus 410 11.0 PFS <0.0001 Study completed 
Placebo  4.6 PFS  Yao et al., 2011 (26) 
Carcinoid tumors 
Octreotide LAR 90 14.3 TTP <0.0001 Study completed 
Placebo  6.0 TTP  Rinke et al., 2009 (34) 
Everolimus + octreotide LAR 429 16.4 PFS 0.026b Study completed 
Placebo + octreotide LAR  11.3 PFS  Pavel et al., 2011 (56) 
Lanreotide 200   Accrual completed 
Placebo     
Bevacizumab + octreotide LAR 400   Recruiting 
IFN-α + octreotide LAR     
Carcinoid syndrome 
Octreotide LAR 202   Recruiting 
Pasireotide LAR     
Lanreotide 100   Recruiting 
Placebo     
RegimenNMedian PFS and/or TTP (months)P-valueStudy status and reference
Pancreatic NETs 
Sunitinib 171 11.4 PFS 0.0001a Study completed 
Placebo  5.5 PFS  Raymond et al., 2011 (29) 
Everolimus 410 11.0 PFS <0.0001 Study completed 
Placebo  4.6 PFS  Yao et al., 2011 (26) 
Carcinoid tumors 
Octreotide LAR 90 14.3 TTP <0.0001 Study completed 
Placebo  6.0 TTP  Rinke et al., 2009 (34) 
Everolimus + octreotide LAR 429 16.4 PFS 0.026b Study completed 
Placebo + octreotide LAR  11.3 PFS  Pavel et al., 2011 (56) 
Lanreotide 200   Accrual completed 
Placebo     
Bevacizumab + octreotide LAR 400   Recruiting 
IFN-α + octreotide LAR     
Carcinoid syndrome 
Octreotide LAR 202   Recruiting 
Pasireotide LAR     
Lanreotide 100   Recruiting 
Placebo     

Abbreviations: LAR, long-acting repeatable; PFS, progression-free survival; TTP, time to progression.

aNot statistically significant due to unplanned analyses and early termination.

bNot statistically significant. Prespecified boundary is P ≤ 0.0246.

NETs are vascular tumors; therefore, there is also a strong rationale to study antiangiogenic agents in NETs (Fig. 1). Sunitinib, an inhibitor of VEGF and platelet-derived growth factor receptors, was also studied in a double-blind placebo-controlled phase III study (Table 1; ref. 29). Treatment with sunitinib 37.5 mg daily was associated with an improvement in median PFS from 5.5 to 11.4 months (HR = 0.42; 95% CI, 0.26–0.66; ref. 29). Although the study failed to achieve statistical significance owing to unplanned interim analyses and early termination, its results are supported by other studies of VEGF inhibitors in pancreatic NET (30–32).

Results from these 2 randomized phase III studies led to the approval of everolimus and sunitinib for the treatment of pancreatic NET and changed the treatment paradigm. However, it should be noted that the combination of daily everolimus and sunitinib is poorly tolerated and should not be combined in routine clinical use (33).

Systemic therapy for carcinoid

Cytotoxic chemotherapy has minimal benefit for advanced carcinoids. With no new agent approved over the past 3 decades, treatment options for these indolent tumors are very limited. The PROMID study (Fig. 1; Table 1) showed that long-acting repeatable octreotide significantly improved time to progression in treatment-naïve patients with midgut carcinoids (HR = 0.34; 95% CI, 0.2–0.59; ref. 34). However, the utility of somatostatin analogues such as octreotide for control of tumor growth in NETs of other primary sites remains undefined.

Antitumor efficacy and survival outcome with IFN in carcinoids are limited and controversial. On the basis of its antitumor activity observed in preliminary studies, IFN currently used either alone or in combination with somatostatin analogues for patients with symptomatic carcinoids (35).

Liver-directed therapy approaches

Taking advantage of the liver's regenerative capacity and its dual blood supply from both the hepatic artery and portal vein, liver-directed therapies have long been used in the management of NETs. These approaches associated with moderately higher complication rates are best employed for symptomatic NET patients with indolent disease course. Partial hepatectomy can be considered for patients when more than 90% of the tumor can be removed safely (36). Hepatic arterial embolization and chemoembolization can be considered as palliative measures in place of surgery (37). Newer radio-embolization techniques may have reduced acute toxicity but have not been prospectively compared with other established liver-directed modalities.

Improved diagnostics

With increasing recognition of the differences in biology and clinical behavior among pancreatic, thoracic, and gastrointestinal NETs, advanced imaging and molecular diagnostics will play progressively important roles in the classification and optimal management of NETs.

Much of the proof of concept for novel molecular diagnostics has already been done. For example, studies using comparative genomic hybridization and single-nucleotide polymorphism have shown distinct patterns of allelic alternation for pancreatic and ileal NETs (38, 39). These patterns of tissue-specific transcription factors, such as TTF1 and CDX2, could be leveraged to identify occult primary tumors.

Advances in cross-sectional and nuclear imaging are allowing us to peer into the human body with unprecedented spatial resolution and characterize abnormalities with increasing precision. Advanced techniques in CT using negative bowel contrast are permitting us to detect small primary tumors of the luminal gut that previously have been inaccessible to endoscopy examination. Novel PET agents, such as [11C]-5-HTP, 18F-FDOPA, 68Ga-DOTA-TOC, and 68Ga-DOTA-NOC, are offering significantly improved spatial resolution for functional imaging (40, 41).

Novel systemic therapy approaches

Major advances in the systemic therapy of NETs have been seen in the past several years. The recent completion of 3 randomized phase III studies has shown that rigorous evaluation of novel agents in this disease is feasible and can lead to practice-changing outcomes.

Therapy targeting phosphoinositide 3-kinase/Akt/mTOR pathway in pancreatic NET

The substantial efficacy of everolimus and the link of multiple germline and somatic mutations in the phosphoinositide 3-kinase (PI3K)/Akt/mTOR pathway genes to pancreatic NETs show the importance of this pathway in tumorigenesis (Fig. 1; refs. 24, 26). Nonetheless, therapeutic resistance frequently emerges over time. A number of strategies having potential to overcome such resistance are under development. Inhibition of TORC1 by everolimus may lead to upregulation of Akt through an insulin-like growth factor (IGF)/PI3K–dependent pathway. Strategies to downregulate IGF with somatostatin analogues such as octreotide and pasireotide, or inhibit IGF I signaling with a monoclonal antibody, such as cixutumumab, are being developed in combination therapy with everolimus. The PI3K/Akt/mTOR pathway can also be blocked at multiple points by using serine–threonine kinase inhibitors that simultaneously inhibit PI3K and mTOR, such as BEZ235 or agents that inhibit both TORC1 and TORC2, such as INC128.

Therapy targeting angiogenesis in pancreatic NET

Pancreatic NETs are also among the few malignancies in which the VEGF inhibitor sunitinib has shown benefit in a pivotal trial (29). Nonetheless, much of the initial promise of antiangiogenic therapy remains unfulfilled. Resistance to antiangiogenic therapy eventually develops. Preclinical studies suggest resistance mechanisms may involve upregulation of transcription factors that control the expression of multiple proangiogenic molecules (42–44). Further development strategies are taking advantage of a single agent or a combination of agents that target multiple proangiogenic pathways. Strategies targeting VEGF, along with fibroblast growth factor or MET, are already in development for other malignancies. Alternatively, targeting VEGF along with transcription factors, such as hypoxia-inducible factor or Sp1, can be another strategy against eventual therapeutic resistance.

Cytotoxic chemotherapy in pancreatic NET

In contrast with carcinoid tumors, recent prospective and retrospective studies have suggested that oral cytotoxic alkylating agent temozolomide is active in pancreatic NETs (45–47). In one retrospective series, for example, temozolomide-based therapy was associated with an overall response rate of 34% in patients with pancreatic NETs (45). In smaller case series, higher response rates have been reported for the combination of capecitabine and temozolomide (48). However, an adequate prospective controlled study to define the role of temozolomide or temozolomide-based combination in pancreatic NET is lacking, and promising activity of this cytotoxic agent awaits confirmation.

Therapy targeting angiogenesis in carcinoid

Several single-arm phase II studies have shown activity for angiogenesis inhibitors in advanced carcinoids. Although most studies using VEGF tyrosine kinase inhibitors have reported a lower response rate in carcinoids compared with pancreatic NETs, potential for delay of tumor growth remains (Fig. 1; refs. 30–32). In a small randomized run-in study, patients with advanced carcinoid tumors were randomly assigned to treatment with bevacizumab or pegylated IFN-α-2b (49). Clinical activity of bevacizumab was evidenced by a response rate of 18% and an improved PFS rate at week 18 (95% vs. 68%). These encouraging results led to the development of a pivotal phase III study led by the Southwest Oncology Group (Table 1), in which patients are randomized to receive either interferon-α-2b or bevacizumab in addition to octreotide. Results of this study will likely define the role of VEGF inhibitors in carcinoids.

Therapy targeting mTOR pathway in carcinoid

Parallel to its development in pancreatic NETs, everolimus was also evaluated in a phase III study among patients with progressive, well-differentiated NETs and carcinoid syndrome (Table 1). Patients received long-acting repeatable octreotide plus everolimus or placebo (50). The study showed a clinically important 5.1 months (from 11.3 to 16.4 months) improvement in PFS (HR = 0.77; 95% CI, 0.59–1.00; ref. 50). The observed P-value of 0.026, however, missed the prespecified boundary of 0.0246. The efficacy of everolimus in NETs of nonpancreatic origin will need to be confirmed in a future study.

Future development of immunotherapy in carcinoid

Immunotherapy is another promising area for advance. Past studies have shown relevant clinical activity for IFN and s.c. interleukin-2 (51, 52). The expression of multiple cancer-testis antigens provides additional rationale. Although there has been little systematic and rigorous development of immunotherapy in NETs, the recent advances in targeting of cytotoxic T-lymphocyte antigen 4 and PD-1 provide opportunities for future advances.

Personalizing therapy in NETs

NETs are heterogeneous in their biologic behavior and aggressiveness. This characteristic, along with the recent emergence of multiple active therapies, has led to considerable interest in predictive and prognostic biomarkers that may allow us to tailor therapy. In addition to the known prognostic value of tumor grade, recent studies have also confirmed the prognostic value of plasma levels of CGA and neuron-specific enolase (NSE). In a multinational study with central radiology review, patients with baseline elevated CGA or NSE had significantly shorter PFS and overall survival (53). Further, early CGA or NSE response was linked to favorable therapeutic benefit (21, 53). If these findings are validated in subsequent studies, biomarkers could be used to select patients for therapy versus active surveillance among newly diagnosed patients with significant tumor burden and to guide frequency of monitoring among patients on active therapy.

Emerging data also suggest that high methylguanine-DNA methyltransferase (MGMT) expression is associated with therapeutic resistance to temozolomide (45, 54). If confirmed in future prospective studies, low MGMT expression could help select patients for treatment.

Predictive biomarkers of therapeutic benefits for mTOR and VEGF inhibitors remain elusive. Multiple somatic mutations in the mTOR pathway, however, have been identified (24). Expression levels of PTEN and TSC2 have also recently been linked to outcome (25). Taken together, these findings suggest that future studies evaluating the function of the mTOR pathway may reveal predictive markers of benefit. For VEGF inhibitors, recent studies suggest that functional imaging holds promise for identifying patients likely to respond to treatment (55). Confirmation of these findings in a prospective study will open the way for a personalized approach to treatment.

J.C. Yao, commercial research grant, Novartis; consultant, Novartis, Pfizer, Ipsen. No potential conflicts of interest were disclosed by the other authors.

1.
Williams
ED
,
Sandler
M
. 
The classification of carcinoid tum ours
.
Lancet
1963
;
1
:
238
9
.
2.
Modlin
IM
,
Lye
KD
,
Kidd
M
. 
A 5-decade analysis of 13,715 carcinoid tumors
.
Cancer
2003
;
97
:
934
59
.
3.
Yao
JC
,
Eisner
MP
,
Leary
C
,
Dagohoy
C
,
Phan
A
,
Rashid
A
, et al
Population-based study of islet cell carcinoma
.
Ann Surg Oncol
2007
;
14
:
3492
500
.
4.
Yao
JC
,
Hassan
M
,
Phan
A
,
Dagohoy
C
,
Leary
C
,
Mares
JE
, et al
One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States
.
J Clin Oncol
2008
;
26
:
3063
72
.
5.
Hassan
MM
,
Phan
A
,
Li
D
,
Dagohoy
CG
,
Leary
C
,
Yao
JC
. 
Risk factors associated with neuroendocrine tumors: A U.S.-based case-control study
.
Int J Cancer
2008
;
123
:
867
73
.
6.
Jensen
RT
,
Berna
MJ
,
Bingham
DB
,
Norton
JA
. 
Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management, and controversies
.
Cancer
2008
;
113
[
Suppl
]:
1807
43
.
7.
Ferrone
CR
,
Tang
LH
,
Tomlinson
J
,
Gonen
M
,
Hochwald
SN
,
Brennan
MF
, et al
Determining prognosis in patients with pancreatic endocrine neoplasms: can the WHO classification system be simplified?
J Clin Oncol
2007
;
25
:
5609
15
.
8.
Jamali
M
,
Chetty
R
. 
Predicting prognosis in gastroentero-pancreatic neuroendocrine tumors: an overview and the value of Ki-67 immunostaining
.
Endocr Pathol
2008
;
19
:
282
8
.
9.
Pape
UF
,
Berndt
U
,
Müller-Nordhorn
J
,
Böhmig
M
,
Roll
S
,
Koch
M
, et al
Prognostic factors of long-term outcome in gastroenteropancreatic neuroendocrine tumours
.
Endocr Relat Cancer
2008
;
15
:
1083
97
.
10.
Rockall
AG
,
Reznek
RH
. 
Imaging of neuroendocrine tumours (CT/MR/US)
.
Best Pract Res Clin Endocrinol Metab
2007
;
21
:
43
68
.
11.
Semelka
RC
,
Custodio
CM
,
Cem Balci
N
,
Woosley
JT
. 
Neuroendocrine tumors of the pancreas: spectrum of appearances on MRI
.
J Magn Reson Imaging
2000
;
11
:
141
8
.
12.
Gibril
F
,
Jensen
RT
. 
Diagnostic uses of radiolabelled somatostatin receptor analogues in gastroenteropancreatic endocrine tumours
.
Dig Liver Dis
2004
;
36
[
Suppl 1
]:
S106
20
.
13.
Sundin
A
,
Garske
U
,
Orlefors
H
. 
Nuclear imaging of neuroendocrine tumours
.
Best Pract Res Clin Endocrinol Metab
2007
;
21
:
69
85
.
14.
Massironi
S
,
Sciola
V
,
Peracchi
M
,
Ciafardini
C
,
Spampatti
MP
,
Conte
D
. 
Neuroendocrine tumors of the gastro-entero-pancreatic system
.
World J Gastroenterol
2008
;
14
:
5377
84
.
15.
Yao
JC
,
Rindi
G
,
Evans
DB
. 
Pancreatic endocrine tumors
.
In
:
DeVita
VT
,
Lawrence
TS
,
Rosenberg
SA
,
editors
. 
Cancer: principles & practice of oncology
. 8th ed.
Philadelphia
:
Wolters Kluwer/Lippincott Williams & Wilkins
; 
2008
. p.
1702
21
.
16.
Smith
DB
,
Scarffe
JH
,
Wagstaff
J
,
Johnston
RJ
. 
Phase II trial of rDNA alfa 2b interferon in patients with malignant carcinoid tumor
.
Cancer Treat Rep
1987
;
71
:
1265
6
.
17.
Dirix
LY
,
Vermeulen
PB
,
Fierens
H
,
De Schepper
B
,
Corthouts
B
,
Van Oosterom
AT
. 
Long-term results of continuous treatment with recombinant interferon-alpha in patients with metastatic carcinoid tumors—an antiangiogenic effect?
Anticancer Drugs
1996
;
7
:
175
81
.
18.
Janson
ET
,
Rönnblom
L
,
Ahlström
H
,
Grandér
D
,
Alm
G
,
Einhorn
S
, et al
Treatment with alpha-interferon versus alpha-interferon in combination with streptozocin and doxorubicin in patients with malignant carcinoid tumors: a randomized trial
.
Ann Oncol
1992
;
3
:
635
8
.
19.
Knigge
U
,
Hansen
CP
,
Stadil
F
. 
Interventional treatment of neuroendocrine liver metastases
.
Surgeon
2008
;
6
:
232
9
.
20.
Yao
KA
,
Talamonti
MS
,
Nemcek
A
,
Angelos
P
,
Chrisman
H
,
Skarda
J
, et al
Indications and results of liver resection and hepatic chemoembolization for metastatic gastrointestinal neuroendocrine tumors
.
Surgery
2001
;
130
:
677
82
, discussion
682–5
.
21.
Kouvaraki
MA
,
Ajani
JA
,
Hoff
P
,
Wolff
R
,
Evans
DB
,
Lozano
R
, et al
Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas
.
J Clin Oncol
2004
;
22
:
4762
71
.
22.
Turner
NC
,
Strauss
SJ
,
Sarker
D
,
Gillmore
R
,
Kirkwood
A
,
Hackshaw
A
, et al
Chemotherapy with 5-fluorouracil, cisplatin and streptozocin for neuroendocrine tumours
.
Br J Cancer
2010
;
102
:
1106
12
.
23.
Yao
JC
. 
Neuroendocrine tumors. Molecular targeted therapy for carcinoid and islet-cell carcinoma
.
Best Pract Res Clin Endocrinol Metab
2007
;
21
:
163
72
.
24.
Jiao
Y
,
Shi
C
,
Edil
BH
,
de Wilde
RF
,
Klimstra
DS
,
Maitra
A
, et al
DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors
.
Science
2011
;
331
:
1199
203
.
25.
Missiaglia
E
,
Dalai
I
,
Barbi
S
,
Beghelli
S
,
Falconi
M
,
della Peruta
M
, et al
Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway
.
J Clin Oncol
2010
;
28
:
245
55
.
26.
Yao
JC
,
Shah
MH
,
Ito
T
,
Bohas
CL
,
Wolin
EM
,
Van Cutsem
E
, et al
RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3) Study Group
. 
Everolimus for advanced pancreatic neuroendocrine tumors
.
N Engl J Med
2011
;
364
:
514
23
.
27.
Kulke
MH
,
Bergsland
EK
,
Yao
JC
. 
Glycemic control in patients with insulinoma treated with everolimus
.
N Engl J Med
2009
;
360
:
195
7
.
28.
Yao
JC
,
Ito
T
,
Oberg
K
. 
Advances in pancreatic neuroendocrine tumor treatment
.
N Engl J Med
2011
;
364
:
1874
5
.
29.
Raymond
E
,
Dahan
L
,
Raoul
JL
,
Bang
YJ
,
Borbath
I
,
Lombard-Bohas
C
, et al
Sunitinib malate for the treatment of pancreatic neuroendocrine tumors
.
N Engl J Med
2011
;
364
:
501
13
.
30.
Kulke
MH
,
Lenz
HJ
,
Meropol
NJ
,
Posey
J
,
Ryan
DP
,
Picus
J
, et al
Activity of sunitinib in patients with advanced neuroendocrine tumors
.
J Clin Oncol
2008
;
26
:
3403
10
.
31.
Hobday
TJ
,
Rubin
J
,
Holen
K
,
Picus
J
,
Donehower
R
,
Marschke
R
, et al
MC044h, a phase II trial of sorafenib in patients (pts) with metastatic neuroendocrine tumors (NET): A Phase II Consortium (P2C) study
.
J Clin Oncol
2007
;
25
:
4504
.
32.
Phan
AT
,
Yao
JC
,
Fogelman
DR
,
Hess
KR
,
Ng
CS
,
Bullock
SA
, et al
A prospective, multi-institutional phase II study of GW786034 (pazopanib) and depot octreotide (sandostatin LAR) in advanced low-grade neuroendocrine carcinoma (LGNEC)
.
J Clin Oncol
2010
;
28
:
4001
.
33.
Molina
AM
,
Feldman
DR
,
Voss
MH
,
Ginsberg
MS
,
Baum
MS
,
Brocks
DR
, et al
Phase 1 trial of everolimus plus sunitinib in patients with metastatic renal cell carcinoma
.
Cancer
2011
Sep 6. [Epub ahead of print]
.
34.
Rinke
A
,
Müller
HH
,
Schade-Brittinger
C
,
Klose
KJ
,
Barth
P
,
Wied
M
, et al
PROMID Study Group
. 
Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group
.
J Clin Oncol
2009
;
27
:
4656
63
.
35.
Shah
T
,
Caplin
M
. 
Endocrine tumours of the gastrointestinal tract. Biotherapy for metastatic endocrine tumours
.
Best Pract Res Clin Gastroenterol
2005
;
19
:
617
36
.
36.
Que
FG
,
Sarmiento
JM
,
Nagorney
DM
. 
Hepatic surgery for metastatic gastrointestinal neuroendocrine tumors
.
Adv Exp Med Biol
2006
;
574
:
43
56
.
37.
O'Toole
D
,
Ruszniewski
P
. 
Chemoembolization and other ablative therapies for liver metastases of gastrointestinal endocrine tumours
.
Best Pract Res Clin Gastroenterol
2005
;
19
:
585
94
.
38.
Kim
H
,
Nagano
Y
,
Choi
IS
,
White
JA
,
Yao
JC
,
Rashid
A
. 
Allelic alterations in well-differentiated neuroendocrine tumors (carcinoid tumors) identified by genome-wide single nucleotide polymorphism analysis and comparison with pancreatic endocrine tumors
.
Genes Chromosomes Cancer
2008
;
47
:
84
92
.
39.
Wang
GG
,
Yao
JC
,
Worah
S
,
White
JA
,
Luna
R
,
Wu
TT
, et al
Comparison of genetic alterations in neuroendocrine tumors: frequent loss of chromosome 18 in ileal carcinoid tumors
.
Mod Pathol
2005
;
18
:
1079
87
.
40.
Boudreaux
JP
,
Klimstra
DS
,
Hassan
MM
,
Woltering
EA
,
Jensen
RT
,
Goldsmith
SJ
, et al
North American Neuroendocrine Tumor Society (NANETS)
. 
The NANETS consensus guideline for the diagnosis and management of neuroendocrine tumors: well-differentiated neuroendocrine tumors of the Jejunum, Ileum, Appendix, and Cecum
.
Pancreas
2010
;
39
:
753
66
.
41.
Orlefors
H
,
Sundin
A
,
Garske
U
,
Juhlin
C
,
Oberg
K
,
Skogseid
B
, et al
Whole-body (11)C-5-hydroxytryptophan positron emission tomography as a universal imaging technique for neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and computed tomography
.
J Clin Endocrinol Metab
2005
;
90
:
3392
400
.
42.
Yao
JC
,
Phan
A
. 
Overcoming antiangiogenic resistance
.
Clin Cancer Res
2011
;
17
:
5217
9
.
43.
Allen
E
,
Walters
IB
,
Hanahan
D
. 
Brivanib, a dual FGF/VEGF inhibitor, is active both first and second line against mouse pancreatic neuroendocrine tumors developing adaptive/evasive resistance to VEGF inhibition
.
Clin Cancer Res
2011
;
17
:
5299
310
.
44.
Zhang
J
,
Jia
Z
,
Li
Q
,
Wang
L
,
Rashid
A
,
Zhu
Z
, et al
Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors
.
Cancer
2007
;
109
:
1478
86
.
45.
Kulke
MH
,
Hornick
JL
,
Frauenhoffer
C
,
Hooshmand
S
,
Ryan
DP
,
Enzinger
PC
, et al
O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumors
.
Clin Cancer Res
2009
;
15
:
338
45
.
46.
Kulke
MH
,
Stuart
K
,
Earle
CC
,
Bhargava
P
,
Clark
JW
,
Enzinger
PC
, et al
A phase II study of temozolomide and bevacizumab in patients with advanced neuroendocrine tumors
.
J Clin Oncol
2006
;
24
:
4044
.
47.
Kulke
MH
,
Stuart
K
,
Enzinger
PC
,
Ryan
DP
,
Clark
JW
,
Muzikansky
A
, et al
Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors
.
J Clin Oncol
2006
;
24
:
401
6
.
48.
Strosberg
JR
,
Fine
RL
,
Choi
J
,
Nasir
A
,
Coppola
D
,
Chen
DT
, et al
First-line chemotherapy with capecitabine and temozolomide in patients with metastatic pancreatic endocrine carcinomas
.
Cancer
2011
;
117
:
268
75
.
49.
Yao
JC
,
Phan
A
,
Hoff
PM
,
Chen
HX
,
Charnsangavej
C
,
Yeung
SC
, et al
Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b
.
J Clin Oncol
2008
;
26
:
1316
23
.
50.
Pavel
ME
,
Hainsworth
JD
,
Baudin
E
,
Peeters
M
,
Hörsch
D
,
Winkler
RE
, et al
RADIANT-2 Study Group
. 
Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study
.
Lancet
2011
;
378
:
2005
12
.
51.
Schnirer
II
,
Yao
JC
,
Ajani
JA
. 
Carcinoid—a comprehensive review
.
Acta Oncol
2003
;
42
:
672
92
.
52.
Lissoni
P
,
Barni
S
,
Tancini
G
,
Mainini
E
,
Piglia
F
,
Maestroni
GJ
, et al
Immunoendocrine therapy with low-dose subcutaneous interleukin-2 plus melatonin of locally advanced or metastatic endocrine tumors
.
Oncology
1995
;
52
:
163
6
.
53.
Yao
JC
,
Pavel
M
,
Phan
AT
,
Kulke
MH
,
Hoosen
S
,
St Peter
J
, et al
Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus
.
J Clin Endocrinol Metab
2011
;
96
:
3741
9
.
54.
Kulke
MH
,
Siu
LL
,
Tepper
JE
,
Fisher
G
,
Jaffe
D
,
Haller
DG
, et al
Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting
.
J Clin Oncol
2011
;
29
:
934
43
.
55.
Yao
JC
,
Phan
AT
,
Fogleman
D
,
Ng
CS
,
Jacobs
CB
,
Dagohoy
CD
, et al
Randomized run-in study of Bevacizumab (B) and Everolimus (E) in low- to intermediate- grade neuroendocrine tumors (LGNETs) using perfusion CT as functional biomarker
.
J Clin Oncol
2010
;
28
:
4002
.
56.
Pavel
M
,
Hainsworth
JD
,
Baudin
E
,
Peeters
M
,
Hoersch
D
,
Anthony
L
, et al
A randomized, double-blind, placebo-controlled, multicenter phase III trial of everolimus + octreotide LAR vs. placebo + octreotide LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2)
.
Ann Oncol
2010
;
21
:
viii4
.