Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 1.
Published in final edited form as: Expert Opin Pharmacother. 2018 May 24;19(8):909–928. doi: 10.1080/14656566.2018.1476492

Everolimus in treatment of neuroendocrine tumors: efficacy, side-effects, resistance and factors affecting its place in the treatment sequence.

Lingaku Lee 1, Tetsuhide Ito 2, Robert T Jensen 1,
PMCID: PMC6064188  NIHMSID: NIHMS977115  PMID: 29757017

Abstract

Introduction:

Since the initial approval of everolimus in 2011, there have been a number of important changes in therapeutic/diagnostic modalities as well as classification/staging systems of neuroendocrine tumors (NETs), which can significantly impact the use of everolimus in patients with advanced NETs.

Areas covered:

The efficacy of everolimus monotherapy and combination therapy demonstrated in clinical studies involving patients with advanced NETs are reviewed. Several factors affecting everolimus’ use are described including: the development and routine use of NET classification/staging systems; widespread use of molecular imaging modalities; side-effects; drug resistance; and the availability of other treatment options. Furthermore, the current position of everolimus in the treatment approach is discussed, taking into account the recommendations from the recent guidelines.

Expert opinion:

Although everolimus demonstrated its high efficacy and tolerability in the RADIANT trials and other clinical studies, there still remain a number of controversies related to everolimus treatment in the management of NETs. The synergistic anti-growth effect of other agents in combination with everolimus or its effect on overall survival have not been established. The appropriate order of the use of everolimus in the treatment of advanced NETs still remains unclear, which needs to be defined in further studies and will be addressed in the new guidelines.

Keywords: Neuroendocrine tumor (NET), everolimus, mammalian target of rapamycin (mTOR) inhibitor, RADIANT trials

1. Background

Neuroendocrine tumors (NETs) are rare malignancies derived from the neuroendocrine system throughout the body [1]. The annual incidence of NETs is increasing and was 6.98 per 100,000 persons in 2012 reported from Surveillance, Epidemiology, and End Results (SEER) program[2], with most NETs commonly arising from lung and gastroenteropancreatic (GEP) sites[2,3]. NETs comprise a heterogeneous group of neoplasms with a wide spectrum of clinical and malignant behaviors depending on various clinicopathological factors. NETs are usually indolent by nature and slow growing, however, poor prognosis of those in advanced stage have not improved dramatically until recently[24], and thus effective therapeutic options are highly warranted.

The phosphatidylinositol-3-kinase (PI3K)/Akt and mammalian target of rapamycin (mTOR) signaling pathway is well known to play a crucial role in controlling cancer cell-cycle and growth[5], and its activation is involved in the pathogenesis of NETs[6]. Upregulation of mTOR signaling components[711], downregulation of its upstream negative regulators; phosphatase and tensin homolog deleted on chromosome 10 [PTEN] and tuberous sclerosis complex 2 [TSC2] [1214], and genomic mutation of mTOR pathway[15,16] in NETs were reported from numerous groups. The anti-proliferative effect of mTOR pathway inhibition was reported in preclinical models of NETs [12,17], and thus it was a promising therapeutic target[6,18]. The efficacy and safety of everolimus, an oral inhibitor of the mTOR pathway[19], in patients with advanced NETs of different origins was examined in a series of phase II/III clinical studies; RADIANT trials (Table 1) [2023]. Of these, RADIANT-3 trial was a randomized, double-blind, placebo-controlled, phase III trial that demonstrated significant clinical benefit in the largest number of patients with advanced pancreatic NETs (panNET) [22]. Based on this result, the Food and Drug Administration (FDA) approved everolimus as an anti-tumoral drug for the treatment of panNETs with progressive disease on May 5, 2011, and it is now commercially available in >110 countries.

Table 1.

RADIANT 1–4; studies related to the efficacy of everolimus in NETs and sub-analyses

Study, phase
(year)
n Treatment (n) Population (n)
[comment]
Progression-free
Survival (PFS)
Overall
Survival (OS)
Objective
response rate (ORR)
Stable
disease
RADIANT-1, phase II;
Yao et al (2008)[20]
60 Eve 5 or 10 mg + Oct (n=30, each) Low- to intermediate-grade NET; pancreas (n=29), small bowel (n=16), lung (n=4) 60 weeks
(72 weeks for 10-mg cohort)
Not reached
(3-yr: 78%)
22%
(30% PRs in 10-mg cohort)
70%
(67% in 10-mg cohort)
RADIANT-2, phase III;
Pavel et al (2011)[21]
429 Eve + Oct (n=216) vs placebo + Oct (n=213) NET with carcinoid syndrome; small bowel (n=224), lung (n=44), colon (n=28), pancreas (n=26) 16.4 vs 11.3 mo
(HR 0.77, p = 0.026)
Not reached
(HR 1.22)
2% vs 2% 84% vs 81%
 Sub-analysis;
 Pavel et al
 (2017)[59]
429 Final OS [High baseline CgA level was associated with poor prognosis, but not predictive for Eve effect on OS] - 29.2 vs 35.2 mo
(HR 1.17)
- -
 Sub-group analysis;
 Fazio et al
 (2013)[60]
44 Eve + Oct (n=33) vs placebo + Oct (n=11) Lung NET (n=44) 13.6 vs 5.6 mo
(HR 0.72, p = 0.228)
- (Tumor shrinkage a;
67% vs 27%)
-
 Sub-group analysis;
 Castellano et al
 (2013)[61]
39 Eve + Oct (n=19) vs placebo + Oct (n=20) Colorectal NET (n=39) 29.9 vs 6.6 mo
(HR 0.34, p = 0.011)
- (Tumor shrinkage a;
67% vs 37%)
-
 Sub-analysis;
 Anthony et al
 (2015)[62]
429 Comparison of previous SSA use (n=339)
with SSA-naïve (n=90)
[Previous SSA use did not affect PFS] SSA(+); 14.3 vs 11.1 mo
SSA(−); 25.2 vs 13.6 mo
- - -
 Sub-analysis;
 Strosberg et al
 (2015)[63]
196 Efficacy of Oct; comparison of previous SSA use (n=155) with SSA-naïve (n=41) NET in placebo arm (n=196); midgut (n=143), foregut (n=32), hindgut (n=21) SSA(+); 11.1 mo
SSA(−); 13.6 mo
(22.2 mo for
SSA-naïve midgut NET)
SSA(+); 33.5 mo
SSA(−); 50.6 mo
- -
 Sub-analysis;
 Pavel et al
 (2017)[64]
Effect of Eve co-administration on Oct pharmacokinetics [Co-administration of Eve and Oct increased Cmin of Oct, but did not impact efficacy] - - - -
RADIANT-3, phase III;
Yao et al (2011)[22]
410 Eve (n=207) vs placebo (n=203) panNET 11.0 vs 4.6 mo
(HR 0.35, p < 0.001)
Not reached 5% vs 2% 73% vs 51%
 Sub-analysis;
 Yao et al
 (2016)[65]
410 Final OS [High baseline CgA, NSE, PIGF and sVEGFR1 levels were associated with poor OS] - 44.0 vs 37.7 mo
(HR 0.94, p = 0.30)
- -
 Sub-group analysis;
 Ito et al (2012)[66]
40 Eve (n=23) vs placebo (n=17) Japanese panNET (n=40) 19.5 vs 42.8 mo
(HR 0.19, p < 0.001)
- 4% vs 6% 83% vs 29%
 Sub-analysis;
 Lombard-Bohas et al
 (2015)[67]
410 Comparison of previous CTx (n=206) with CTx-naïve (n=204) [Previous CTx did not affect PFS] CTx(+); 11.0 vs 3.2 mo
CTx(−); 11.4 vs 5.4 mo
- CTx(+); 5% vs 2%
CTx(−); 5% vs 2%
CTx(+); 73% vs 45%
CTx(−); 73% vs 56%
RADIANT-4, phase III;
Yao et al (2016) [23]
302 Eve (n=205) vs placebo (n=97) NF-NET; GI (n=175) and lung (n=90) 11.0 vs 3.9 mo
(HR 0.48, p < 0.001)
Not reached
(HR 0.64)
2% vs 1% 80% vs 64%
 Sub-group analysis;
 Singh et al
 (2017)[68]
211 Eve (GI; n=118, unknown; n=23) vs placebo (GI; n=57, unknown; n=13) NF-NET; GI (n=175) and unknown primary (n=36) GI; 13.1 vs 5.4 mo (HR 0.56), unknown primary; 13.6 vs 7.5 mo (HR 0.60) - - -
 Sub-group analysis;
 Fazio et al
 (2017)[69]
90 Eve (n=63) vs placebo (n=27) NF-NET; lung (n=90) 9.2 vs 3.6 mo
(HR 0.50)
- 2% vs 4%
(Tumor shrinkage a;
58% vs 13%)
79% vs 56%
 Sub-analysis;
 Buzzoni et al
 (2017)[70]
302 Effect of prior SSA therapy (n=163), CTx (n=77) and RTx (n=63) on Eve activity [Prior therapy did not affect PFS] SSA(+); 11.1 vs 4.5 mo
SSA(−); 9.5 vs 3.7 mo
CTx(+); 9.2 vs 2.1 mo
CTx(−); 11.2 vs 5.4 mo
RTx(+); 9.2 vs 5.4 mo
RTx(−); 11.0 vs 3.0 mo
- SSA(+); 1% vs 1%
SSA(−); 3% vs 0%
CTx(+); 2% vs 0%
CTx(−); 2% vs 1% RTx(+); 0% vs 5%
RTx(−); 3% vs 0%
SSA(+); 78% vs 65%
SSA(−); 83% vs 63%
CTx(+); 78% vs 44%
CTx(−); 82% vs 70% RTx(+); 75% vs 47%
RTx(−); 82% vs 68%
 Sub-analysis;
 Pavel et al
 (2016)[71]
Report of health related quality of life (HRQOL) [HRQOL was maintained in both Eve and placebo group]

CgA, chromogranin A; Cmin, minimum concentration; CTx, chemotherapy; Eve, everolimus; GI, gastrointestinal; HR, hazard ratio; NET, neuroendocrine tumor; NF, non-functioning; Oct, octreotide; NSE, neuron specific enolase; PIGF; placental growth factor, panNET, pancreatic neuroendocrine tumor; PR, partial response; RTx, radiotherapy; SSA, somatostatin analogue; St, stratum; sVEGFR; soluble vascular endothelial growth factor receptor.

a

Included decrease in tumor size less than 30%.

The purpose of this article is to evaluate at present (7 years post initial approval), the clinical utility of everolimus in NET patients. This is not possible without first considering the advances in the diagnosis and treatment of NETs that are currently influencing anti-tumor treatment approaches to advanced NETs, which have occurred since everolimus’ initial approval in 2011 (Table 2). An understanding of these is essential to exploring the current options that are available and the place of everolimus within these for the current treatment of NETs.

Table 2.

Recent important advances in the management/treatment of NETs since initial approval of everolimus by FDA (5/5/2011)[22]

Changes in anti-tumor treatment
 1. Approval of sunitinib by FDA for anti-growth therapy in panNETs (5/20/2011) [24]
 2. Approval of lanreotide by FDA for anti-growth therapy in GEP-NET (12/16/2014) [25]
 3. Additional indication of everolimus in GI and lung NETs (FDA approved on 5/26/2016) [23]
 4. Increased use of PRRT and its approval by FDA in GEP-NETs (1/26/2018) [26,27]
 5. Increased use of new liver directed therapy (SIRT using 90Yttrium microspheres)[28,29]
Changes in treatment of clinical symptoms in functioning NETs
 6. Approval of telotristat ethyl by FDA for symptom control of carcinoid syndrome refractory to SSA (2/28/2017) [30]
 7. Additional indication of lanreotide for treatment of carcinoid syndrome (FDA approved on 9/18/2017) [31]
Changes in imaging and management
 8. Development and routine use of classification/staging system (WHO, ENETS, AJCC) with prognostic value [3436]
 9. Development and routine use of classification/staging system (WHO, ENETS, AJCC) for treatment selection [3436]
 10. Change in classification systems (WHO 2015, 2017) affecting prognosis/treatment (i.e. NET G3 vs NEC [G3]) [46]
 11. Improvement and widespread use of molecular imaging modalities for diagnosis and tumor location[4749]
 - SRS [111Indium-pentetreotide with SPECT/CT] (FDA approved on 1994)
 - 68Gallium-DOTATATE with PET/CT (FDA approved on 6/1/2016)
 12. Increased use of molecular imaging to predict aggressiveness, survival, and grade (18Fluorine-FDG PET/CT) [50,51]

AJCC, American Journal of Critical Care; ENETS, European Neuroendocrine Tumor Society; FDA; Food and Drug Administration, FDG; fluorodeoxyglucose, GEP, gastroenteropancreatic; NET, neuroendocrine tumor; panNET, pancreatic neuroendocrine tumor; PET, positron emission tomography; PRRT, peptide receptor radionuclide therapy; SIRT, selective internal radiation therapy; SPECT, single-photon emission computed tomography; SRS; somatostatin receptor scintigraphy; SSA, somatostatin analogue; WHO,World Health Organizationpan

Shortly after the approval in May, 2011 of everolimus for treatment of patients with advanced panNETs, sunitinib, an oral multi-targeted tyrosine kinases inhibitor, was shown to have efficacy in patients with advanced panNETs, prolonging progression free survival (PFS) in a phase III trial[24], and it was approved as a standard therapeutic agent in those patients worldwide (Table 2), thus offering an additional treatment choice. Three years later, lanreotide autogel was shown to improve PFS in a phase III trial (CLARINET) in patients with advanced grade 1 or 2 NETs, and it was approved by FDA as an anti-growth therapy in GEP-NET[25](Table 2). Therefore, by 2014, there were three new anti-tumor medications (everolimus, sunitinib, and somatostatin analogues [SSAs]) available to treat patients with advanced panNETs, in addition to older therapies such as chemotherapy. In 2016, the RADIANT-4 trial showed the benefit of everolimus in lung and all gastrointestinal (GI) NETs to extend PFS in patients with advanced disease[23], leading to its additional approval for extrapancreatic NETs. In 2018, peptide receptor radionuclide therapy (PRRT) using 177 Lutetium (Lu) -DOTATATE was globally approved for the treatment of GEP-NETs[26,27](Table 2). This approval was based on the results of the phase III (NETTER-1) trial in which 177 Lu-DOTATATE markedly extended PFS in patients with advanced somatostatin-receptor (SSTR) -positive midgut NETs with acceptable safety[26], and on data from 504 patients treated with advanced panNETs and other NETs in Rotterdam[27]. In addition, over the last 7 years, therapeutic options have been developed for the treatment of patients with liver predominant metastatic disease from malignant NETs. These treatments include the increasing use of selective internal radiation therapy (SIRT) with 90Yttrium (Y) labeled microspheres to reduce the hepatic tumor burden[28,29](Table 2).

There have also been additional drugs approval to treat the clinical symptoms of functional NETs (F-NETs), such as the carcinoid syndrome or F-panNETs syndromes. In the management of carcinoid syndrome, one new drug became available recently (Table 2). Telotristat ethyl, a peripheral-acting tryptophan hydroxylase inhibitor, which blocks serotonin synthesis, has been shown to cause a significant reduction in bowel movement frequency in a phase III trial (TELESTAR) in patients with carcinoid syndrome[30]. In combination with SSA, it was approved by FDA for the treatment of SSA-refractory carcinoid syndrome in February, 2017 (Table 2). In addition, SSAs such as octreotide and lanreotide have been approved for the treatment of F-NETs, and in a recent phase III trial (ELECT), lanreotide autogel reduced bowel movement frequency in patients with carcinoid syndrome, regardless of primary location (Table 2)[31].

In addition to new treatments for patients with NETs, there was a number of changes in the management of these patients over the past 7 years since the initial approval of everolimus which affect current practices (Table 2). Histopathological classification and TNM staging systems proposed by World Health Organization (WHO), European Neuroendocrine Tumor Society (ENETS) and American Joint Committee on Cancer (AJCC) are now routinely used in clinical practice to predict prognosis and to select optimal treatments. The WHO classification system separates NETs into several groups based on the proliferative index and tumor differentiation assessed by Ki-67 labeling index and mitotic counts, including NET G1, NET G2 and G3 for GEP-NETs[32], and typical carcinoids (TC), atypical carcinoids (AC) and neuroendocrine carcinoma (NEC) for lung NETs[33], respectively (Table 3). The prognostic value of these systems was validated by several studies[3436], however, heterogeneity remained in the high-grade G3 that showed different prognosis, genetic profiles, and treatment response to platinum-based chemotherapy in different groups of G3 patients[3745]. To address this issue, the recent 2017 WHO classification for panNETs has divided the G3 group for panNETs into well-differentiated G3 NETs (WDNEC) as a distinct subgroup from poorly-differentiated NEC (PDNEC, Table 2,3)[46]. The use of these classification/grading systems is now mandatory in all recent guidelines, and their widespread use is having an effect on the management of these patients[3436]. It affects anti-tumor drug use, because cisplatin-based regimens are recommended for G3 NEC and the role of everolimus in NET G3 patients, as well as other therapeutic options, in these patients is still being defined.

Table 3.

WHO classification of NETs of pancreas, GI tract, and lung.

2017, Pancreas [46]
2010, GI tract [32]
2015, Lung [33]
Classification Differentiation  Ki-67 index
 (%)
Mitotic count
(/10 HPF)
Classification  Ki-67 index
 (%)
Mitotic count
(/10 HPF)
Classification Mitotic count
(/10 HPF)
Necrosis
NET G1 Well < 3 < 2 NET G1 ≤ 2 < 2 Typical carcinoid < 2 Absence
NET G2 Well 3–20 2–20 NET G2 3–20 2–20 Atypical carcinoid 2–10 Absence or punctiform
NET G3 Well > 20 > 20 NEC > 20 > 20 NEC,
large cell type
> 10 Large
NEC (G3) Poor
(large cell or small cell type)
> 20 > 20 NEC,
small cell type
> 10 Large

GI; gastrointestinal, HPF; high power field, NEC; neuroendocrine carcinoma, NET; neuroendocrine tumor, WHO; World Heath Organization.

Diagnostic modalities, especially molecular imaging, have now been validated[1], and are now routinely used with 68Gallium (Ga) -DOTATATE positron emission tomography (PET/CT) imaging receiving FDA approval on June 2016, supported by several studies[4749] that demonstrated its high sensitivity/specificity for the detection of location and extent of SSTR positive NETs (Table 2). In addition, 18F-fluorodeoxyglucose (FDG) PET is being increasingly used in combination with 68Ga-DOTATATE to define patients with aggressive tumors, usually of high grade (Table 2)[50,51]. These modalities can influence treatment options, because they are better defining the extent of the disease as well as pattern of growth, which can be important in selecting different anti-tumor treatments.

Despite these many recent advances (Table 2), mentioned above, the treatment of advanced NETs still has many unanswered questions. These will be discussed in a separate, later section. Prior to discussion of the current controversies as well as the clinical use of everolimus at present in patients with advanced NETs, the findings from studies of the efficacy/safety of everolimus in these patients will be briefly reviewed.

2. An overview of the current market

As described in depth in the previous background section, since the approval by the FDA of everolimus for its anti-growth effect in patients with advanced panNETs in May 5, 2011, and the subsequent approval for GI and lung NETs in May 26, 2016, there have been numerous changes in the diagnosis and treatment of these diseases which affects the potential use of everolimus at present. These are summarized in Table 2 and includes both the introduction of other anti-tumor agents (sunitinib, lanreotide, PRRT, and liver-directed therapy) and changes in the management of NETs that affect its treatment. These include the demonstration of efficacy of everolimus in controlling the hormone-excess state, as well as changes in tumor imaging and tumor classification which provide prognostic information which can affect the anti-tumor treatment.

3. Introduction to the Compound

As reviewed in the background, numerous in vitro and in vivo studies in animals with NETs, as well as preliminary human studies (Table 4) provide evidence that the PI3K/Akt/mTOR pathway plays an important role in proliferation of NETs. Everolimus (40-O-[hydroxyethyl]-rapamycin), also known as RAD001, is a rapamycin analog that directly inhibits the mTORC1 pathway by binding to intracellular receptor FK506-binding protein (FKBP-12), which subsequently inhibits activation of its downstream mediators such as p70S6K/4E-BP1 (Box 1)[5,52]. Its efficacy in patients with NETs was established in numerous studies (Table 1,3). Before the approval of everolimus for the treatment of panNETs in 2011, it was already approved by FDA for its anti-growth effect in patients with renal cell carcinoma in 2009[53] and TSC-associated subependymal glial cell astrocytoma in 2010[54], as well as for immunosuppression to prevent organ rejection in kidney transplants recipients in 2010[55], and had proven safe/effective in these patients. Subsequent to its approval for patients with advanced NETs, everolimus also received FDA approval for the anti-proliferative treatment of hormone receptor-positive breast cancer (in combination with exemestane)[56] and TSC-associated renal angiomyolipoma[54] in 2012.

Table 4.

Phase I/II studies related to the efficacy of everolimus treatment with SSAs or studies with everolimus in combination with other agents in NETs

Study, phase
(year)
n Treatment (n) Population (n)
[comment]
PFS OS ORR SD
Phase II;
Yao et al (2010)[72]
160 Statrum 1; Eve,
Statrum 2; Eve + Oct
panNET; Oct-naïve (St 1, n=115), patients on Oct for ≥3 mo (St 2, n=45) St 1; 9.7 mo,
St 2; 16.7 mo
St 1; 24.9 mo,
St 2; not reached
(2-yr; 54.7%)
St 1; 9.6%,
St 2; 4.4%
St 1; 68%,
St 2; 80%
Phase II:
Bajetta et al
(2014) [73](2017)[74]
50 Eve + Oct NET; GI (n=11), pancreas (n=14), lung (n=11) TTP; 33.6 mo 61.9 mo 18% 74%
COOPERATE-2, phase II;
Kulke et al (2017)[75]
160 Eve + Pas (n=79) vs Eve (n=81) panNET 16.8 vs 16.6 mo
(HR 0.99, p = 0.488)
Not reached
(HR 0.93, p = 0.410)
20% vs 6% 57% vs 77%
LUNA, Phase II;
Ferolla et al (2017)[76]
124 Pas (n=41) vs Eve (n=42) vs Eve + Pas (n=41) NET; lung (n=116),
thymus (n=8)
8.5 vs 12.5 vs 11.8 mo
(39% vs 33% vs 59% at 9 mo)
- 2.4% in each group
(Tumor shrinkage a; 31% vs 49% vs 73%)
34% vs 31% vs 49%
Phase I;
Chan et al (2012)[77]
22 Eve + Pas NET; GI (n=14), pancreas (n=4), bronchus (n=2) 76% at 6 mo
65% at 12 mo
- (Tumor shrinkagea; 81%) 90%
CALGB 80701, phase II;
Kulke et al (2017)[75]
150 Eve + Oct + Bv (n=75) vs Eve + Oct (n=75) panNET 16.7 vs 14.0 mo
(HR 0.80, p = 0.12)
36.7 vs 35.0 mo
(HR 0.75, p = 0.16)
31% vs 12%
(p = 0.005)
-
Phase I;
Dasari et al (2015)[78]
19 Eve + Oct + cixutumumab NET; small bowel (n=6), pancreas (n=4), lung (n=4) 43.6 weeks 25.5 mo (Tumor shrinkage a; 89%) 89%
Phase I;
Kim et al (2018)[79]
13 Eve + Pas + SIRT NET; GI (n=6), pancreas (n=3), lung (n=2) 18.6 mo 46.3 mo 46% 23%
Phase I/II;
Chan et al (2013)[83]
43 Eve 5 or 10 mg + temozolomide panNET 15.4 mo - 40% 53%
Phase I;
Chan et al (2013)[84]
21 Eve + sorafenib NET; GI (n=13), bronchus (n=3), pancreas (n=3) 79% at 6 mo - 6% (Tumor shrinkage a; 62%) 76%
NETTLE, Phase I;
Claringbold et al (2015)[85]
16 Eve + PRRT NET; GI (n=11),
pancreas (n=5)
-
(No disease progression in 6 mo treatment)
Not reached
(1-yr; 88%, 2-yr; 63%)
44% 50%
Phase II;
Salazar et al (2017)[86]
62 BEZ235 (n=31) vs Eve (n=31) panNET 8.2 vs 10.8 mo (HR 1.53) (6-mo; 96.6% vs 90.3%) 9.7% vs 9.7% 52% vs 81%
Phase II;
Oh et al (2012)[87]
27 Eve NF-GEP-NET 17.1 mo Not reached 12% 88%

Bv, bevacizumab; Eve, everolimus; GI, gastrointestinal; HR, hazard ratio; NET, neuroendocrine tumor; NF; non-functioning, Oct, octreotide; ORR, objective response rate; OS, overall survival; Pas, pasireotide; panNET, pancreatic neuroendocrine tumor; PFS, progression free survival; PRRT, Peptide Receptor Radionuclide Therapy; SD, stable disease; St, statrum; TTP; time to progression.

a

Included decrease in tumor size less than 30%.

4. Pharmacokinetics and Pharmacodynamics

Oral everolimus is rapidly absorbed after oral administration, and reaches a peak serum concentration after 1.3–1.8 hours[57]. Steady-state serum concentrations are reached within a week, and both maximum serum concentration and the area under the curve, increased in a dose-proportional manner[58]. Everolimus is metabolized mainly by cytochrome P450 (CYP) 3A1, 3A5 and 2C8 in the gut and liver, and is a substrate for P-glycoprotein (P-gp). Approximately 98% and 2% of everolimus is excreted in the bile in the form of metabolites and urine, respectively (for detail, see[57]).

5. Efficacy of everolimus

5.1. RADIANT trials (Table 1)

The results of the RADIANT trial series[2023] and sub-analyses[5971] are summarized in Table 1. The efficacy of everolimus in NETs was initially investigated in the RADIANT-1 trial, an open-label, phase II pilot dose-finding study involving patients with metastatic, low- to intermediate-grade NETs of pancreas, small bowel, and lung origin[20]. Sixty patients were treated with either everolimus 5 mg or 10 mg in combination with octreotide. Among those received everolimus 10 mg, median PFS was 72 weeks, and 30% achieved a partial response[PR]. Based on these results, further phase III studies with large number of patients with NETs were carried out.

The RADIANT-2 trial was an international randomized, double-blind, placebo-controlled phase III trial that compared everolimus plus octreotide and octreotide alone in 426 patients with NETs with carcinoid syndrome[21]. PFS in everolimus arm was 5.1 months longer than the control arm with a 23% risk reduction of disease progression, but prolongation of PFS did not reach the predefined level of statistical significance (P = 0.026 reached, needed P = 0.024) (Table 1). This result may have been affected by the imbalance of patient characteristics between the everolimus and the control arms (i.e., WHO performance status, lung origin). Several sub-group analyses were performed from the results of the RADIANT-2 trial (Table 1)[5964]. Although the numbers were small, everolimus plus octreotide demonstrated superior anti-tumor activity in patients with lung[60] and colorectal NETs[61]. Another sub-group analysis showed the efficacy of everolimus regardless of prior SSA exposure[62]. These results suggest that everolimus may be beneficent in patients with extrapancreatic NETs.

The RADIANT-3 trial was an international randomized, double-blind, placebo-controlled phase III trial involving patients with advanced panNETs. Patients were randomized (1:1) to receive everolimus or placebo plus best supportive care, including concomitant SSA use in 40% of patients[22](Table 1). Everolimus significantly prolonged PFS (11.0 vs 4.6 months), representing 65% risk reduction of PFS. Sub-group analyses revealed that prolonged PFS with everolimus treatment was irrespective of WHO performance status, age, sex, ethnicity, tumor grade, and prior chemotherapy/SSA therapy[22,66,67]. Everolimus treatment was associated with low objective response rate (ORR 5% [i.e. sinficant decrease in tumor size by RECIST criteria]), however, patients treated with everolimus achieved a high rate of disease stabilization (i.e. stable disease (SD) achieved in 73%, Table 1). The final analysis of overall survival (OS) showed a 6.3 month prolongation in the everolimus arm (44.0 vs 37.7 months), however, this difference was not statistically significant, likely due to the high proportion (85%) of patients in the placebo arm who crossed over to everolimus treatment after disease progression.

The RADIANT-4 trial was an international randomized, double-blind, placebo-controlled phase III trial that studied everolimus efficacy in 302 patients with advanced GI and lung NETs without carcinoid syndrome (Table 1)[23]. Everolimus significantly improved the median PFS by 7.1 months compared to placebo (11.0 vs 3.9 months), with risk reduction of disease progression or death by 52%. High disease stabilization was observed in the everolimus arm (81%), which was comparable to that reported in the RADIANT-3 trial[22]. Sub-group analyses showed clinically meaningful anti-tumor activity of everolimus regardless of its primary site[68,69] and prior therapies[70].

5.2. Other phase I/II trials and retrospective studies related to efficacy of everolimus (Table 4)

Table 4 summarize the results of other phase I/II studies in NETs related to the efficacy of everolimus, commonly combined with SSA[7275,7579], which were performed following the evidence from preclinical studies that showed enhanced anti-tumor effect of everolimus in combination with SSA[8082].

An open-label phase II study assessed the efficacy of everolimus in 160 patients with advanced panNETs who progressed after prior systemic chemotherapy[72]. In a total of 45 patients who were taking octreotide for at least 3 months at study entry (documentation of disease progression was required), when treated with everolimus plus octreotide, the median PFS and ORR were 16.7 months and 4.4%, respectively. In 115 patients who were not being treated with octreotide, everolimus responses for PFS and ORR were 9.7 months and 9.6%, respectively[72]. Another multicenter phase II trial with a single arm everolimus plus octreotide treatment in a first-line setting was reported from Italian group[73,74]. Among 50 patients with GEP and lung NETs, ORR was 18%, and median time to progression(TTP) and OS were 33.6 months and 61.0 months, respectively[73,74]. Although the patient numbers included in these studies were small, these studies raise the possibility that the use of octreotide combined with everolimus may have additional anti-tumor benefit in NETs, similar to the results shown in the RADIANT-2 trial[21].

A number of studies have examined the efficacy of everolimus with other non-octreotide anti-tumor agents (Table 4). Theses includes the combination of everolimus with pasireotide[7577,79], bevacizumab[75], SIRT[79], temozolomide[83], sorafenib[84], or PRRT[85](Table 4). In a phase I study of pasireotide plus everolimus in 22 patients with GEP and lung NETs[77], the PFS at 6 months was 76%. Disease stability was achieved in 90%, and 81% showed some tumor shrinkage (which did not meet the RECIST PR criteria [less than 30% decrease in tumor size])[77]. In the COOPERATE-2 trial, an open-label phase II study[75], 160 patients were randomized to everolimus plus pasireotide or everolimus alone. The primary endpoint was PFS, but the addition of pasireotide to everolimus failed to show significant superiority in the prolongation of PFS compared to everolimus alone (16.8 vs 16.6 months)[75]. The LUNA trial was a three-arm, randomized, open-label phase II study comparing everolimus plus pasireotide with either agent alone in 112 patients with advanced lung or thymus NETs[76]. All three arms achieved the pre-planned statistical objective of a 9-month PFS rate >20%, with a median PFS of 11.8, 12.5 and 8.5 months in combination arm, everolimus arm, and pasireotide arm, respectively[76]. Taken together with the higher ORR observed in the combination therapy in the COOPERATE-2 (20% vs 6%)[75], combination therapy of pasireotide plus everolimus may have enhanced activity in panNETs and extrapancreatic NETs. Further studies are warranted to elucidate the synergistic effect of these agents.

Other phase I/II trials have investigated the efficacy of other therapeutic agents alone or in combination with everolimus (Table 4) [75,78,8387]. Of these, the CALGB 80701 trial was a randomized phase II trial that compared the vascular-endothelial growth factor (VEGF) inhibitor, bevacizumab, plus everolimus and everolimus alone in panNETs[75]. Despite encouraging results from preliminary phase II studies[8890], the results of this study were disappointing in not showing enhanced value of bevacizumab[75]. Accordingly, bevacizumab plus everolimus showed a 2.7 months prolongation in PFS compared to everolimus alone (P = 0.12). Significantly higher ORR were also observed[75] (Table 4), which is consistent with another study evaluating efficacy of temsirolimus combined with bevacizumab in panNETs (ORR 41%) [88]. Although prolongation in PFS was deemed statistically significant (pre-determined P value threshold; 0.15), this was not a clinically satisfying improvement. The high type I error rate of this study design requires further well-designed trial to confirm the true efficacy of this combination therapy. Other studies showed the positive results of everolimus combination therapy in the small number of patients with other therapeutic agents including temozolomide[83], PRRT[85], and SIRT[79], and further studies were planned in some cases.

Since the global approval of everolimus, numerous retrospective studies from various countries have reported the efficacy of everolimus in NETs in the real clinical practice, particularly in those of GEP origin (Table 5)[91100]. Overall, the median PFS (12–27.5 months) and ORR (0–28%) among these study populations were similar or superior to those reported in RADIANT-3 trials[22]. In addition, several patients remained on treatment more than 5 years after everolimus initiation[94,101]. Clinically meaningful prolonged PFS reported from these studies might contribute to the improvement of OS[102]. Considering that the anti-tumor activity of everolimus is mainly presented as high disease stabilization, long-term continuation of everolimus might be important in maximizing its efficacy. Although several limitations such as study design and small number of patients could have overestimated everolimus’s efficacy, these results were important in supporting the conclusion that everolimus can provide significant and prolonged clinical benefit not only in the clinical trials but also in the real-world clinical setting.

Table 5.

Other prospective/retrospective studies related to the efficacy of everolimus in NETs

Study, (year) Design n Treatment Primary PFS OS ORR SD
Panzuto (2014) [91] Retro 169 Eve P, GI, L 12 mo 32 mo 8% 68%
Berardi (2017) [92] Retro 116 Eve P 19 mo 44 mo 10% 73%
Liu (2016) [93] Retro 53 Eve P, GI 18.9 mo 63.9 mo 28% 55%
Lee (2017) [94] Retro 47 Eve P 27.5 mo 60.8 mo 19% 66%
Lee (2017) [95] Retro 40 Eve P 20 mo - 0% 65%
Yao (2015) [96] Pro 39 Bv or Eve a
→ Bv + Eve
P, GI, L 14.6 mo 27.9 mo 25% 69%
Kamp (2013) [97] Retro 24 Eve P, GI 13.1 mo - 17% 63%
Angelousi (2017) [98] Retro 21
11
Eve; 1st line
Eve; 2nd line
P
P
16.3 mo
15.5 mo
(2-yr; 83%)
(2-yr; 69%)
-
-
-
-
Yoo (2017) [99] Retro 11
6
Eve
Eve
P
GI
16.6 mo
14.7 mo
Not reached
27.7 mo
9%
17%
73%
83%
Capdevila (2015) [100] Retro 57 Eve + lanreotide P, GI 25.8 mo b 26.4 mo 18% 61%

Bv,bevacizumab; Eve, everolimus,; GI,gastrointestinal; L, lung; NET, neuroendocrine tumor; ORR, objective response rate; OS, overall survival; P, pancreas; PFS,progression free survival; Pro prospective; Retro, retrospective; SD, stable disease.

a

Treated with either agents alone for 3 weeks prior to combination therapy.

b

Reported as TTP (time to progression).

5.3. Efficacy of everolimus in functioning NETs

A number of studies demonstrated in some patients with refractory F-NETs, everolimus may be helpful for treatment of hormone-excess state, independent of its anti-growth effect[103], which will be discussed in more detail below.

To understand the possible importance of everolimus for control of symptoms in F-NET patients, the current treatment options need to be briefly reviewed. In the management of F-NETs, control of the hormone hypersecretory syndrome is a key factor for maintaining quality of life (QOL) and also survival in many cases[103,104]. Carcinoid-syndrome is the most frequent ectopic hormonal syndromes in NETs[104], and SSA is the standard initial treatment [104106]. In the recent phase III TELESTAR trial, telotristat ethyl, a peripheral tryptophan hydroxylase inhibitor, in combination with SSA demonstrated significant reduction in bowel movement frequency in NETs with SSA refractory carcinoid syndrome[30]. For some of the other F-NET symptoms, there are reports of the effectiveness of everolimus when other antisecretory therapies fail. In Zollinger–Ellison syndrome (ZES) in patients with gastrinomas, which invariably have hyperchlorhydria/gastrin hypersecretion [107109], the standard therapy for control of the hypersecretion is the use of proton pump inhibitors (PPIs), occasionally high-dose histamine H2 receptors, and rarely SSA[110113]. These are generally successful in all patients, thus everolimus is not needed to control the acid hypersecretion in these patients[110,113,114]. Most patients with insulinomas are cured surgically, thus the medical therapy of the hormone-excess state is usually only needed during the preoperative period or the small percentage with malignant tumors(5–15%)[103,114,115]. The primary treatment is frequent small feedings and the use of diazoxide and SSA[103,111], however, SÁ can cause severe hypoglycemia by attenuating counter-regulatory hormone release[116]. In patients with malignant insulinomas, control of the hypoglycemia can be difficult and may not be satisfactory[103,114,115]. VIPomas are uncommon, but the hormone excess state can be difficult to treat. The first option is SSA, however, continuation may lose its effectiveness and additional treatment is needed[103,111].

Some reports and small series provide evidence that everolimus can be helpful in some patients with refractory F-NET symptoms[103]. Recently, the effect of everolimus on gastrin levels was reported in patients with panNETs from RADIANT-1 and RADIANT-3 trials [117]. Accordingly, everolimus rapidly reduced median gastrin levels to approximately 60% in the RADIANT-1 trial and by greater than 50% in the RADIANT-3 trial (everolimus vs placebo, p < 0.001), respectively. In addition, these down-regulatory effects of everolimus were irrespective of prior or concomitant SSA use. Whether this will also apply to ZES patients is unclear. Several studies in small number of patients and case reports also indicated the usefulness of everolimus in controlling carcinoid syndrome[118,119]. Inhibition of mTOR pathway was reported to attenuate insulin secretion in an insulinoma cell line[120]. Furthermore, everolimus controlled hypoglycemic symptoms in patients with insulinomas[103,121,121127].

5.4. Efficacy of everolimus in G3 NETs/NEC

Chemotherapy with platinum-based regimens is widely accepted as a standard treatment among patients with PDNEC[1,128]. In contrast, patients with WDNEC/G3 NETs usually do not responds well to this chemotherapy regimen[3741], and the best treatment as for this group of patients is not resolved. In a retrospective study including 15 patients with well to moderately differentiated panNET G3 treated with everolimus, median PFS and OS were 6 and 28 months, respectively, and 40% achieved disease stabilization for >1 year [129]. Other groups also reported the favorable response to everolimus in small number of patients with panNET G3[42,94,130132]. Taken together, everolimus could be a potential anti-tumor therapy in panNET G3 as well as possibly extrapancreatic WDNEC.

With respect to the PDNEC, several immunohistochemical analyses reveal high expression and activation of mTOR [7,133], and an anti-proliferative effect of everolimus was shown in a preclinical study[134]. A case study reported a patient with PDNEC who was successfully treated with everolimus for 15 months[135]. Although everolimus alone had limited efficacy[136], combination therapy with paclitaxel showed preliminary anti-tumor activity in patients with previously treated small-cell lung NEC[137]. In a phase II trial, everolimus combined with paclitaxel and carboplatin was active in 49 patients with large-cell lung NEC[138]. Accordingly, the median PFS, OS and ORR were 4.4 months, 9.9 months and 45%, respectively. Considering the poor prognosis due to its aggressive nature of PDNEC[139], further studies are warranted to evaluate possible synergistic effects of everolimus with platinum-based regimen in PDNEC.

6. Markers predicting efficacy of everolimus

The recent increase in therapeutic options for patients with advanced NETs show variable responses in different patients, and thus have raised the importance of biomarkers in predicting response to treatment that could enable optimal treatment selection and change in therapy[140]. In the RADIANT-1 trial, early response in serum chromogranin A (CgA) or neuron-specific enolase (NSE) levels was a significant predictor for prolonged PFS with everolimus treatment[72]. In addition, elevated baseline CgA and NSE levels were associated with shorter PFS and OS with everolimus treatment[141]. Similarly, sub-analysis of RADIANT-3 trial also showed that high baseline CgA, NSE, placental growth factor (PIGF) or soluble vascular endothelial growth factor receptor 1 (sVEGFR1) levels were associated with poor OS[65], however, baseline CgA levels were not predictive for everolimus treatment effect on OS in the RADIANT-2 trial[59]. Another study genotyped the fibroblast growth factor receptor 4 (FGFR4) using DNA isolated from tissue or blood in panNETs, and found that patients homozygous for FGFR-G388 demonstrated greater tumor shrinkage with everolimus treatment than those harboring only one FGFR4-R388 allele[142]. Although longer PFS and OS were also observed in those homozygous for FGFR4-G388, these were not statistically significant due to small number of patients, which is consistent with the result from another group showing that FGFR4 polymorphism did not have predictive value on everolimus treatment[143].

The efficacy of other markers, specifically the components of mTOR pathway, has been evaluated by several groups. In 17 patients with NETs treated with everolimus and octreotide, high tumor p-Akt levels in pre-treatment (R = 0.476, P = 0.053) and on-treatment (R = 0.604, P = 0.015) tumor biopsy specimens correlated with longer PFS[144]. In an immunohistochemical analysis of bronchial NETs in 21 patients, protein levels of the total and phosphorylated form of mTOR and its component (p70S6K [ribosomal protein S6 kinase, 70 kDa], Akt and ERK 1/2) were upregulated in everolimus sensitive patients compared to everolimus resistant patients[145]. The efficacy of monitoring intra-tumor p-p70S6K [80,146148] was suggested by other groups, however, the relationship to PFS and tumor response with everolimus were conflicting. Other clinicopathologic factors including histological grade [22,9395], Ki-67 index[91,94] and hepatic metastasis burden[93] were also reported to affect PFS of everolimus treatment. In addition, the results of imaging modalities, such as 89Zirconium (Zr)-bevacizumab PET[149] and perfusion CT[96], in predicting response to everolimus was suggested by several groups. Furthermore, patients experienced stomatitis within 8 weeks of everolimus initiation had longer PFS than those without stomatitis in the RADIANT-3 trial (13.9 vs 8.3 months, hazard ratio [HR] 0.70) and in RADIANT-2 trial (HR 0.87) [150]. Similar to this, occurrence of hypercholesterolemia was also suggested as a possible marker of response[146].

7. Safety of everolimus (Table 6)

Table 6.

Common adverse events (AEs) associated with everolimus in previous studies

Study (year) n Regimen AEs, all grade (grade 3/4), %
Stomatitis Rash Diarrhea Fatigue Infection Pneumonitis Hyperglycemia Plt decrease Anemia
Eve monotherapy
Yao et al (2011)[22] 207 Eve 64(7)* 49(<1) * 34(3)* 31(2)* 23(2)* 17(2)* 13(5) 13(4)* 17(6) *
Yao et al (2016)[65] 225 Eve 60(4) 37(1) 26(2) 20(3) 28(5) 10(<1) - 10(4) 14(0)
Yao et al (2016)[23] 205 Eve 63(9) * 27(1) * 31(7) * 31(3) 29(7) * 16(1) * 10(3) -(<1) 16(4) *
Panzuto et al [91] 169 Eve 22(2) - - - - 19(8) 17(1) 22(8) 20(5)
Pavel et al (2016) [166] 123 Eve (panNET) 19(3) 7(2) 32(9) 15(8) 39(19) 5(2) 5(3) 5(2) 11(3)
117 Eve (non panNET) 19(3) 7(2) 32(9) 15(8) 39(19) 5(2) 5(3) 5(2) 11(3)
Berardi et al (2017)[92] 116 Eve -(4) -(1) -(3) -(2) - -(2) -(3) a -(4) -(1)
Liu et al (2016) [93] 53 Eve 36(2) 19(0) - 15(2) - 9(2) 23(9) 9(0) 13(8)
Lee et al (2017) [94] 47 Eve 85(0) 57(0) 21(6) - 53(21) 15(4) 34(4) 28(9) 34(6)
Lee et al (2017) [95] 40 Eve 48(5) 33(3) 10(3) - - 3(0) - 8(0) 13(3)
Oh et al (2012)[87] 32 Eve 18(6) 29(0) 27(3) - - - 12(6) 15(15) 12(6)
Salazar et al (2017)[86] 31 Eve 65(6) 42(0) 55(3) 32(36) - 16(3) 35(6) 23(0) 35(10)
Angelousi et al
(2017)[98]
20
11
Eve (1st line)
Eve (2nd line)
10(0)
9(0)
25(0)
18(0)
15(0) b
45(9) b
10(0)
18(0)
-
-
25(5)
18(9)
25(5)
18(0)
25(5)c
27(5)c
25(5)c
27(5)c
Kamp et al (2013) [97] 24 Eve 42(4) 25(4) 4(4) 33(8) 8(0) 38(0) 25(13) 17(8) 21(0)
Yoo et al (2017) [99] 17 Eve 59(6) 41(0) 29(0) 29(0) - 18(12) - 12(6) 29(0)
Eve-combined therapy
Pavel et al (2011)[21] 216 Eve + Oct 62(7) * 37(1) * 27(6) * 31(7) 20(5) * 12(2) * 12(5) * 14(5) * 15(1) *
Yao et al (2010)[72] 115
45
Eve
Eve + Oct
45(4)
49(2)
40(1)
44(0)
39(4)
31(0)
31(4)
36(2)
-
-
6(0)
13(0)
13(4)
13(4)
8(3)
13(9)
13(4)
16(4)
Yao et al (2008)[20] 60 Eve + Oct 69(8) 64(5) - -(11) - 7(2) 70(9) 35(5) -(3)
Bajetta et al (2014)[73] 50 Eve + Oct 62(10) 48(2) 60(22) - - 6(0) 18(0) 12(0) 8(2)
Dasari et al (2015)[78] 19 Eve + Oct
+ cixutumumab
63(11) 42(0) 58(0) 74(21) 16(5) - 63(11) 53(0) 11(5)
Kulke et al (2017)[75] 79
81
Eve + Pas
Eve
59(9)
63(9)
24(0)
30(0)
63(5)
53(4)
27(4)
35(4)
15(0)
14(0
8(1)
12(1)
76(37)
27(11)
-
-
27(5)
26(5
Dasari et al (2015)[76] 42
41
Eve
Eve + Pas
72(10)
37(5)
33(7)
12(0)
50(7)
76(20)
19(2)
34(10)
9(2)
12(0)
5(5)
15(5)
46(17)
83(24)
23(2)
17(0)
31(2)
25(5)
Chan et al (2012)[77] 22 Eve + Pas 48(5) 48(5) 62(5) 76(0) - - 90(38) 76(14) 76(0)
Chan et al (2013)[83] 43 Eve
+ temozolomide
63(2) 53(5) 51(2) 77(2) - 7(0) 72(19) 67(16) 65(0)
Yao et al (2015)[96] 39 BV or Eve
→ Bv + Eve
61(2) 15(2) 49(15) 61(7) 56(7) - 44(10) 20(5) 32(5)
Chan et al (2013)[84] 21 Eve + sorafenib 38(0) 81(14) 67(14) 76(0) 10(0) 5(5) 76(10) 86(14) 86(0)

Bv, bevacizumab; Eve, everolimus; Plt, platelet; panNET, pancreatic NET; Oct, octreotide; Pas, pasireotide.

*

Frequencies of all grade AEs more than 10% higher than control arm.

a

Reported as metabolic toxicities.

b

Reported as gastrointestinal intolerance.

c

Reported as haematologic toxicities.

Treatment with everolimus is well known to cause a spectrum of adverse events (AEs) in various patients. According to meta-analyses including patients with NETs, treatment with everolimus as well as other mTOR inhibitors, is most commonly associated with an increased risk of stomatitis [150153], rash[151,154], fatigue[155], infection[156], pulmonary toxicities [152,157,158], hyperglycemia[155,159], anemia[160,161], and thrombocytopenia[160]. The pharmacokinetic analysis of RADIANT-2 trial also revealed that an increased everolimus minimum concentration was associated with higher risk for pulmonary and metabolic events[64]. In addition, the development of severe AEs, such as infection and pneumonitis[162], could result in treatment discontinuation[163] and fatal outcome[162,164]. The frequencies of common AEs associated with everolimus treatment observed in the previous studies were listed in Table 6. The majority of AEs in the different studies were mild to moderate, and treatment-related mortality did not occur in most of the studies (Table 6). Pulmonary toxicity (n=5) and infection/sepsis (n=3) were the most common fatal AEs that caused everolimus-related death[22,23,75,76,93]. Though patient-reported tolerance with everolimus treatment was reported to be poorer than SSA[165], health-related QOL was well maintained in large clinical trials[71,79,166]. These results strongly support the general tolerability of everolimus by most patients in both the clinical trial setting and real-world setting.

However, everolimus-associated AEs can be significant and have led 5–35% of patients to treatment discontinuation (Table 7). In addition, dose reduction and/or interruption are frequently (13–100%) required to control the severity of the AEs (Table 7). Like other targeted agents, the importance of appropriate dose modification for the management of everolimus-associated AEs has been emphasized, especially for the long-term continuation of everolimus[94]. This is in line with the pharmacokinetic analysis including patients with NETs that showed significant correlation between elevated everolimus trough concentration (Cmin) and increased risk of toxicity[152,167]. Regarding efficacy, higher Cmin were associated with improved tumor size reduction[152], whereas lower Cmin increased risk of progression[167]. It was reported that patients with high cumulative dose (>3000 mg) and dose intensity (> 9 mg/day) of everolimus experienced significantly longer OS[92]. Although dose intensities were reported to be well maintained in most previous studies[21,72,73,75,76,85], therapeutic drug monitoring (TDM) may be useful in prevention of developing severe AEs without lowering the efficacy of everolimus[167], in addition to the patients’ education and physicians careful monitoring[168].

Table 7.

Treatment-related outcomes and dose adjustment of everolimus in previous studies

Study, (year) n Regimen (n) Treatment
duration,
median
Reasons of discontinuation
Dose adjustment of Eve
Dose intensity
of Eve, median (mg/day)
Eve-related mortality
AEs Disease progression Reduction (R) Interruption (I)
Phase I-III studies
 RADIANT-1, phase II;
Yao et al (2008)[20]
60 Eve + Oct - 5% - 13% - - -
 RADIANT-2, phase III;
Pavel et al (2011)[21]
216 Eve + Oct 9.2 mo 19% 44% 65% (R+I) 8.3 0%
 RADIANT-3, phase III;
Yao et al (2011)[22]
207 Eve 8.8 mo 17% 44% 59% (R+I) <1% a
  Sub-analysis
Yao et al (2016)[65]
225 Eve (open label) 44 wk 24% 55% - - - 0%
 RADIANT-4, phase III;
Yao et al (2016)[23]
205 Eve 40.4 wk 12% 37% 67% (R+I) - 1% b
 Phase II; Yao et al (2010)[72] 115
45
Eve
Eve + Oct
-
-
-
-
-
-
-
-
-
-
9.9
9.4
-
-
 Phase II; Bajetta et al (2014)[73] 50 Eve + Oct 74 wk 16% - 26% 0% 9.4 0%
 COOPERATE-2, phase II;
 Kulke et al (2017)[75]
79
81
Eve + Pas
Eve
49.4 wk
48.3 wk
23%
21%
40%
43%
53%
36%
65%
56%
7.8
9.8
3% c
1% c
 LUNA, Phase II;
Ferolla et al (2015)[76]
42
41
Eve
Eve + Pas
- 35%
32%
17%
24%
67% (R+I)
54% (R+I)
9.4
8.4
2% d
5% d
 Phase I; Chan et al (2012)[77] 22 Eve + Pas 6 cycles (4-wk) 5% 36% 14% - - 0%
 Phase II; Salazar et al (2017)[86] 31 Eve 39.4 wk 16% - - - - 0%
 Phase I/II; Chan et al (2013)[83] 43 Eve + temozolomide 8.5 cycles (4-wk) 21% 49% - - - 0%
 Phase I; Chan et al (2013)[84] 21 Eve + sorafenib 4 cycles (4-wk) 14% 57% 14% - - 0%
 Phase I; Dasari et al (2015)[78] 19 Eve + Oct
+ cixutumumab
10 cycles (3-wk) 21% 42% - - - 0%
 NETTLE, Phase I;
Claringbold et al (2015)[85]
16 Eve + PRRT 24 wk (Eve) + 4 cycles of PRRT (8-wk) - 0% 100% (R+I)
in 10mg cohort
8.3, 8.0, 4.8 in 5mg, 7.5mg, 10mg cohort 0%
Prospective /retrospective studies
 Yao et al (2015)[96] 39 BV or Eve
→ Bv + Eve
- - - 25% (R+I) - 0%
 Pavel et al (2016) [166] 123
117
Eve (PANNET)
Eve (non PANNET)
12.1 wk
24 wk
17%
25%
-
-
39% (R+I)
53% (R+I)
-
-
0%
0%
 Panzuto et al [91] 169 Eve 6 mo 9% 50% 17% 63% - 0%
 Berardi et al (2017)[92] 116 Eve - - - 22% 66% - 0%
 Capdevila et al (2015) [100] 73 Eve + lanreotide 4.7mo 14% 32% - - - -
 Liu et al (2016) [93] 53 Eve - 6% 51% - - - 2% e
 Lee et al (2017) [94] 47 Eve 50 wk 34% 43% - - - 0%
 Lee et al (2017) [95] 40 Eve 8 mo 5% - - - - 0%
 Angelousi et al (2017)[98] 20
11
Eve (1st line)
Eve (2nd line)
13.6 mo
7 mo
10%
9%
90%
63%
-
-
-
-
-
-
0%
0%
 Kamp et al (2013) [97] 24 Eve 10.2 mo 17% 29% 17% 29% - 0%
 Yoo et al (2017) [99] 17 Eve - 24% - 29% - - 0%

AEs, adverse events; Bv,bevacizumab; Eve, everolimus; Plt, platelet; panNET, pancreatic NET; Oct,octreotide; Pas, pasireotide; PRRT, peptide receptor radionuclide therapy .

a

Included acute respiratory distress syndrome (n=1).

b

Included respiratory failure (n=1), septic shock (n=1), and cardiac failure (n=1).

c

Included ketoacidosis (n=1) and sepsis (n=1) in combination arm; pulmonary embolism (n=1) in Eve arm.

d

Included acute kidney injury associated with diarrhea(n=1) in Eve arm; diarrhea and urinary sepsis (n=1), acute renal failure and respiratory failure (n=1) in combination arm.

e

Included interstitial pneumonitis (n=1).

8. Mechanism of resistance to everolimus

Despite the high anti-tumor activity of everolimus, disease progression can occur early after everolimus initiation in some patients, and even in those who once experienced tumor response with everolimus treatment. In the previous studies, up to 57% of the patients terminated everolimus treatment due to disease progression (Table 7). Primary resistance, as well as acquired (secondary) resistance to everolimus is thought to play a role, thus understanding and overcoming the mechanisms underlying everolimus resistance may prove important in preventing patients from treatment discontinuation due to disease progression.

mTOR is the core components of 2 complexes, mTORC1 and mTORC2, which have distinct signaling pathways and functions[19,169]. The activation of mTORC1 subsequently phosphorylates its downstream p70S6K and 4E-BP1[5,52]. Everolimus specifically directly inhibits the mTORC1 and its downstream p70S6K/4E-BP1[170,171], resulting in attenuation of protein synthesis as well as cell growth[12,52,147,148]. The PI3K/Akt/mTORC1 pathway is normally regulated by in part from its upstream insulin-like growth factor 1 (IGF-1) receptors with IGF-1-dependent activation of the PI3K/Akt pathway leading to inhibition of signaling due to mTOR-mediated phosphorylation and degradation of insulin receptor substrate-1 (IRS-1) [80,81,172,173].

Suppression of these feedback loops by inhibiting mTORC1 causes over-activation of upstream signaling, including PI3K/Akt[174,175]. Several studies showed that everolimus may only partially inhibit mTORC1 and incompletely block its certain downstream target, especially 4E-BP1[176,177]. The inability of everolimus to block mTORC2 induces upstream Akt phosphorylation[19,170,178,179]. In addition, a genetic mutation in FKBP12 is reported to affect the sensitivity of mTOR inhibitor[180]. Other mechanisms of resistance to mTOR inhibitors have also demonstrated including; activation of mitogen activated protein kinase(MAPK); up-regulation of pro-angiogenic factors; and activation of Ras pathway[92,174,181,182]. All these mechanisms potentially counteract the anti-proliferative effects of mTORC1 inhibitors and lead to drug resistance[80,172,173].

Consistent with this hypothesis, histopathological analyses and preclinical studies reveal the increased activation of Akt in everolimus-sensitive cell lines and patients[80,144,183]. On the other hand, several studies have showed contrary results that high p-Akt levels were significantly associated with poor survival with everolimus treatment[9,12]. In addition, overexpression p70S6K was reported to be associated with shorter PFS[10,146]. Discrepancies between these studies may partly result from the small number of patients, and due to the heterogeneity in patient’s background, especially their genetic profile[8,16,145,184].

Emerging evidence shows that dual inhibition of these pathways, particularly PI3K/Akt and the mTOR pathway, could be a novel therapeutic approach to overcome everolimus resistance [6,178,183187]. The efficacy and safety of the dual PI3K/mTOR inhibitor, BEZ235, was examined in patients with advanced NETs[86,188,189]. In a phase II trial, 62 patients with mTOR inhibitor-naïve advanced panNETs were randomized to receive BEZ235 (n=31) or everolimus (n=31)[86]. Unfortunately, BEZ235 did not demonstrated superior efficacy compared with everolimus (Table 4). Despite the acceptable tolerability observed in a phase I trial in patients with advanced solid tumors[188], high toxicity of BEZ235 observed in this study led patients to require frequent dose modifications and treatment discontinuations. As a result, the shorter treatment duration of BEZ235 compared to everolimus (22.9 vs 39.4 weeks) might have negative impact on its anti-tumor activity. Poor tolerability of BEZ235 was also reported from a single-arm phase II trial in advanced patients with everolimus-refractory panNETs[189], which has led to its development being stopped. Further studies using other agents with better tolerability are warranted to clarify the effect of pan-PI3K inhibitors against resistance to everolimus.

9. Controversies in everolimus treatment in advanced NETs

9.1. General

To discuss the current controversies involving everolimus as well as the later discussion on expert opinion, some additional information related to current efficacy of competing therapies and current guideline recommendations are needed and will be briefly covered below.

9.2. Current efficacy of various therapeutic modalities

In general, prolonged treatment of NETs requires a multidisciplinary approach which may include biotherapies, targeted therapies, chemotherapies, liver directed therapies, and radiotherapies.

SSAs are generally the initial therapeutic agents used to control the tumor growth, in addition to the hormone excess symptoms as mentioned earlier, not only because they are effective, but they are well tolerated, as shown in a recent survey of patient-reported outcomes[165]. The anti-growth effects of octreotide have been shown in 2 phase III trials. In the PROMID trials, octreotide demonstrated prolonged TTP (14.3 vs 6.0 months, HR 0.34, p < 0.001) in patients with well-differentiated small intestine NETs[190]. Lanreotide autogel also showed further prolonged PFS in the CLARINET study (not reached vs 18.0 months in placebo arm, HR 0.47, p < 0.001) in NETs originated in pancreas, midgut and hindgut[25]; however, it is important to mention that 96% of the patients did not have disease progression in the 3–6 months before randomization. These studies demonstrated the efficacy of SSAs to control tumor growth in both advanced panNETs and GI-NETs.

In the pivotal phase III trial including 171 patients with advanced panNETs[24,191], sunitinib demonstrated significant improvement in PFS (11.4 versus 5.5 months, HR 0.42, p < 0.001) and a trend in prolongation of OS (38.6 vs 29.1 months, HR 0.73, p = 0.094) versus placebo. This study supports the use of sunitinib in advanced panNETs only.

Recently, a markedly prolonged PFS (estimated PFS rate at 20 months; 65.2% vs 10.8%) and high ORR (ORR 18% vs 3%, p < 0.001) of PRRT/octreotide compared to octreotide alone were shown in the phase III NETTER-1 trial in patients with advanced midgut NETs[26]. Because of the additional data showing efficacy/safety from the large, single-group trial (non-prospective) in 310 patients with advanced GEP-NETs (ORR 29%)[27] from the Rotterdam studies, the FDA approved 177Lutetium-DOTATATE for both panNETs and GI-NETs.

The efficacy of streptozotocin- and temozolomide-based regimens in patients with advanced panNETs, especially in causing tumor reduction (range 28–70%), were confirmed by several retrospective studies and small numbers of patients in prospective studies[115,192195]. Other studies demonstrated the possible activity of temozolomide-based chemotherapies in patients with extrapancreatic NETs, specifically originated from bronchus[196] and thymus[197]. Therefore, the high cytotoxic effect of chemotherapy may be beneficial in these patients with high tumor burden. However, it should be noted that almost all studies report lower response rates of GI-NETs to temozolomide-based chemotherapies compared to panNETs (ORR 0–7%) [90,198].

Liver directed therapies, including transarterial embolization/chemoembolization and SIRT using 90Y-microspheres, are also generally used for the treatment of liver-dominant metastases and high tumor burden[28,29]. Although high level evidence (i.e., randomized, placebo-control, phase III trial) has been lacking, these options can provide clinically significant benefit in a subset of patients, particularly for those with progressive disease[199].

Surgical removal of advanced NETs is recommended whenever possible[1,200204]. The effect of this in subsequent treatment with other anti-tumor options has in general not been well studied.

All of the above therapeutic options are well to moderately tolerated[2429,190,192194]. Similar to everolimus, treatment with sunitinib had no impact on health-related QOL in a recent phase III trial involving patients with advanced panNETs[205]. Furthermore, SIRT[79] and PRRT[206] were reported to improve QOL in patients with advanced NETs. On the other hand, patient-reported tolerance was reported to be poor with sunitinib, intravenous chemotherapy, and liver directed therapy, whereas PPRT had better tolerance compared to SSAs[165].

9.3. Current position of everolimus; recommendations from guidelines

Since the initial approval of everolimus in 2011, guidelines provided from several scientific societies have been updated reflecting recent advances in the treatment of NETs. In this section, important changes in the latest guidelines were reviewed and highlighted, especially by focusing on the position of everolimus.

According to the latest ENETS consensus guideline published in 2016[207], everolimus is generally recommended as second-line therapy after failure of SSA or cytotoxic chemotherapy in panNETs. However, it can be considered as first-line therapy if SSA is not an option, and if systemic chemotherapy is not clinically required, not feasible, or not tolerated. Similarly, in GI-NETs, everolimus may be used as a second-line therapy after failure of SSA or as a third-line therapy after failure of PRRT[207]. Due to the lack of approved therapeutic drugs in advanced lung NETs, everolimus is recommended as a first-line therapy in progressive disease, unless SSA may be considered as a first-line therapy (e.g. G1, TC, slow growth, strong SSTR expression)[207]. In these guidelines, the combination therapy of everolimus plus SSA for anti-proliferative purpose is not recommended in nonfunctional NETs regardless of their primary sites[207].

The National Comprehensive Cancer Network (NCCN) guidelines was upgraded in 2017 (version 3. 2017) [208]. In the treatment algorithm regarding panNETs, everolimus alone was recommended as second-line therapy after SSA failure, in patients with symptomatic, or clinically significant tumor burden/progressive disease. In GI-NET, everolimus is also recommended as second-line therapy after progression with SSA regardless of its clinical behavior[208]. Similar to the ENETS guidelines, everolimus with or without SSAs was recommended as first-line treatment of lung and thymus NETs in patients with AC or TC with high tumor burden[208]. All these recommendations were categorized as 2A (i.e. the intervention is appropriate based upon lower-level evidence).

In 2017, the North American Neuroendocrine Tumor Society (NANETS) provided consensus-based guidelines for the treatment of advanced midgut NETs[106]. Although SSAs are the generally recommended initial therapy, everolimus, as well as liver embolization, can be considered as first-line therapy in patients who are not indicated for radiolabeled SSAs (i.e. weak or absent SSTR expression). Everolimus is also recommended as an appropriate therapy in patients presenting massive extrahepatic metastases with weak/absent SSTR expression. The guidelines note that possibly stronger evidence of everolimus efficacy in non-midgut NETs (the majority of patients in the RADIANT-4 trial) than in midgut NETs (the majority of patients in the RADIANT-2 trial) should be taken into account when deciding second-line treatment after SSAs[106].

Similarly, consensus guidelines from Nordic neuroendocrine group in 2014[209] and Canadian experts in 2015[202] also considered everolimus as second-line treatment in panNETs and GI-NETs with progressive disease on SSAs. In the European Society of Medical Oncology (ESMO) guideline in 2012[203], SSAs are the standard recommended first-line treatment, whereas the treatment algorithm shows everolimus as first-line therapy in panNETs G1/2 (Ki-67 2–20%) and GI-NETs (Ki-67 10–20%).

When interpreting these recommendations, it should be noted that some of these guidelines were published several years ago[202,203,209,210], thus do not reflect the important findings from recent clinical trials, specifically RADIANT-4 and NETTER-1 trials, as well as other important changes in management of NETs patients listed in Table 2, which can strongly affect the current position of everolimus.

With respect to F-NETs, a number of guidelines shows the efficacy of everolimus in controlling symptoms of hormone-excess state in a number of F-NETs including insulinomas, and carcinoid syndrome[103105,208210]. In these patients, it is usually used when SSAs and other approved therapies have failed. The NANETS guidelines in 2017 also concluded that everolimus should be considered an option for patients with progressive midgut NETs, even if there is a history of carcinoid syndrome[106].

Common recommendation for the standard treatment of G3 NEC (PDNEC) is systemic platinum-based chemotherapy [128,202,203,207,208,211]. Because the disease concept of WDNEC/NET G3 was recently established, high-level evidence is lacking for the preferred treatment in these patients. However, several guidelines proposed that temozolomide-based chemotherapy[207] and PRRT[212] may be potential treatment options. Even though everolimus has been successful in some patients with G3 tumors, no guidelines have made a recommendation for everolimus use in these patients.

10. Regulatory affairs

For the patients with NETs, everolimus (AFINITOR®) has been approved by the FDA for the treatment of panNETs on May 5, 2011, and for GI and lung NETs on May 26, 2016, respectively. The initial dose of AFINITOR® is 10 mg, to be taken once daily at the same time every day, either consistently with food or consistently without food. Dose reduction is recommended for patients with hepatic impairment depending on its severity and those who required co-administration of moderate inhibitors of CYP3A4 and/or P-gp. If patients require concomitant use of strong CYP3A4 and P-gp inducers, increase dose using increments of 5 mg or less increments to a maximum of 20 mg/day. Dose reduction is also used in controlling side effects.

11. Conclusion

Everolimus is a key therapeutic agent in the treatment of advanced NETs, which covers the spectrum of this heterogeneous groups of neoplasms, regardless of its primary site, histological grade, functional status, and radiological aggressiveness. Further studies are required to define its efficacy and safety in combination with other agents, mechanisms of resistance, and to better define its place in the current treatment options with the recent release of other anti-tumor agents such as PRRT.

12. Expert opinion

The RADIANT trials have corroborated the basic science results, isolated cell studies and studies in animals showing the anti-growth mechanisms of everolimus on NETs, and established its efficacy in patients with advanced NETs[2023]. However, there still remain a number of controversies related to everolimus treatment in the management of NETs.

Despite the clinically meaningful prolongation of PFS with everolimus treatment demonstrated in the RADIANT trials, its effect on overall survival has not yet been established[22,23]. Treatment of advanced NETs requires a multidisciplinary approach, thus appropriate timing for everolimus initiation is important to evaluate long-term outcome. The exact order of the use of everolimus in the treatment cascade of patients with advanced NETs is at present unclear and being affected by a number of factors, such as the recent widespread use of WHO classification with its prognostic value and importance in treatment selection; everolimus’s effectiveness in combination with other anti-tumor treatments; the recognition of long-term potential resistance to everolimus and other targeted therapies; side effects of long-term everolimus treatment; and the increasing development of competing therapies. The use of biomarkers that predict the sensitivity and response to each treatment could be useful in selecting optimal therapeutic options, however, their utility in real clinical practice is so far, not clear.

In recent guidelines, everolimus is generally recommended as second-line therapy behind SSAs[201203,207209], because of the low toxicity with high tolerability of SSAs treatment[165]. At present, everolimus is one of the most frequently used second line treatments in patients presenting clinically significant tumor burden and disease progression on SSAs. Other competing therapeutic agents including sunitinib for panNETs, PRRT, chemotherapy, and liver directed therapy for panNETs and other NETs, may also be indicated in these settings[201203,207209]. Of these, the recent approval of PRRT is most likely to change everolimus’s order in the anti-tumor therapeutic approach. PRRT results in a longer PFS than everolimus, is well-tolerated by patients, a significant percentage of patients show a cytotoxic effect with reduced tumor burden[26,27], and thus it will likely have a significant impact on the use of cytostatic agents including everolimus and sunitinib, especially in patients with aggressive tumors. Severe AE/toxicity of PRRT is reported to be low (1–2%) (leukemia, myeloproliferative effects) [26,27,213]. With PRRT’s general availability and use in all groups of NETs, AE/toxicity will be better defined, and this could play a role in its general use and the position of everolimus in the treatment sequence. It should be noted that most guidelines have not reflected the recent availability of PRRT, shown in the NETTER-1 trial and other recent studies[202,203,209,210].

The underlying mechanisms of primary and/or acquired resistance to everolimus occurring in as many as 17–90% of patients stopping treatment due to loss of efficacy is not entirely clear (Table 7) [80,172,173]. It has a strong impact on the long-term continuation of monotherapy with everolimus, as well as combination and subsequent treatment with other therapeutic agents. Emerging evidences from the preclinical studies have shown a number of likely mechanisms of resistance to everolimus, and several potent agents with or without combination with everolimus have been examined in clinical trials for the purpose of overcoming the resistance to everolimus as well as adding a synergistic anticarcinogenic effect. Some of these show encouraging results, however high toxicity may limit their anti-tumor effect[86,189]. The efficacy of combination therapy with everolimus still needs to be explored more widely in further studies.

It is reported that percentage of patients, which is generally low, stopped everolimus treatment in various regimens owing to AEs (Table 7). Everolimus is generally well tolerated, and most of these AEs/side effects are manageable with dose reduction and interruption. In contrast, decreased dose intensity of everolimus may impair its anti-tumor activity. Balancing the anti-tumor activity and treatment-associated toxicity, as well as maintaining QOL is important in term of long-term continuation. Although it is not required in the real clinical practice, therapeutic drug monitoring may provide important information in this regard[167], as recommended for the use of immunosuppressants after organ transplantation[214]. Development of severe AEs, especially pulmonary toxicities and infections (Table 7)[22,23,75,76,93], can result in fatal outcomes, thus screening of occult infections and pulmonary function is mandatory before everolimus initiation. Adequate control of comorbidities, such as diabetes and hyperlipidemia, could also prevent patients from unnecessary treatment discontinuation. From this viewpoint, one possible strategy for treatment decision-making could be based on patient tolerance, comorbidities, and toxicity profiles. Specifically, everolimus is not indicated in patients with impaired pulmonary function, uncontrolled infections and metabolic disorders[152,155159].

Although a number of unanswered questions remained in this area, in the near future with more widespread use of PRRT and other treatment options, it is likely new guidelines will become available addressing some of these issues.

Acknowledgments

Funding: This manuscript was partially supported by intramural funds of NIDDK, NIH.

Footnotes

Declaration of interests: The authors have no conflict of interests to disclose.

References

Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

  • 1. Ito T, Hijioka S, Masui T , et al. Advances in the diagnosis and treatment of pancreatic neuroendocrine neoplasms in Japan. J Gastroenterol 2017;52:9–18 [DOI] [PubMed] [Google Scholar]
  • 2. Dasari A, Shen C, Halperin D , et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol 2017;3:1335–1342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Yao JC, Hassan M, Phan A , 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;26:3063–3072 [DOI] [PubMed] [Google Scholar]
  • 4. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer 2003;97:934–959 [DOI] [PubMed] [Google Scholar]
  • 5. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012;149:274–293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Wolin EM. PI3K/Akt/mTOR Pathway Inhibitors in the Therapy of Pancreatic Neuroendocrine Tumors. Cancer Lett 2013;335:1–8 [DOI] [PubMed] [Google Scholar]
  • 7. Shida T, Kishimoto T, Furuya M , et al. Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemother Pharmacol 2010;65:889–893 [DOI] [PubMed] [Google Scholar]
  • 8. Righi L, Volante M, Rapa I , et al. Mammalian target of rapamycin signaling activation patterns in neuroendocrine tumors of the lung. Endocr Relat Cancer 2010;17:977–987 [DOI] [PubMed] [Google Scholar]
  • 9. Qian ZR, Ter-Minassian M, Chan JA , et al. Prognostic significance of MTOR pathway component expression in neuroendocrine tumors. J Clin Oncol 2013;31:3418–3425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Kasajima A, Pavel M, Darb-Esfahani S , et al. mTOR expression and activity patterns in gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer 2011;18:181–192 [DOI] [PubMed] [Google Scholar]
  • 11. Komori Y, Yada K, Ohta M , et al. Mammalian target of rapamycin signaling activation patterns in pancreatic neuroendocrine tumors. J Hepatobiliary Pancreat Sci 2014;21:288–295 [DOI] [PubMed] [Google Scholar]
  • 12. Missiaglia E, Dalai I, Barbi S , et al. Pancreatic Endocrine Tumors: Expression Profiling Evidences a Role for AKT-mTOR Pathway. J Clin Oncol 2010;28:245–255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Casciano R, Chulikavit M, Perrin A , et al. Cost-effectiveness of everolimus vs sunitinib in treating patients with advanced, progressive pancreatic neuroendocrine tumors in the United States. J Med Econ 2012;15 Suppl 1:55–64 [DOI] [PubMed] [Google Scholar]
  • 14. Perren A, Komminoth P, Saremaslani P , et al. Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am J Pathol 2000;157:1097–1103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Jiao Y, Shi C, Edil BH , et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 2011;331:1199–1203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Scarpa A, Chang DK, Nones K , et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 2017;543:65–71 [DOI] [PubMed] [Google Scholar]
  • 17. Grozinsky-Glasberg S, Franchi G, Teng M , et al. Octreotide and the mTOR inhibitor RAD001 (everolimus) block proliferation and interact with the Akt-mTOR-p70S6K pathway in a neuro-endocrine tumour cell Line. Neuroendocrinology 2008;87:168–181 [DOI] [PubMed] [Google Scholar]
  • 18. Chan J, Kulke M. Targeting the mTOR Signaling Pathway in Neuroendocrine Tumors. Curr Treat Options Oncol 2014;15:365–379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Yang H, Rudge DG, Koos JD , et al. mTOR kinase structure, mechanism and regulation. Nature 2013;497:217–223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Yao JC, Phan AT, Chang DZ , et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol 2008;26:4311–4318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Pavel ME, Hainsworth JD, Baudin E , et al. 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;378:2005–2012 [DOI] [PubMed] [Google Scholar]
  • 22. Yao JC, Shah MH, Ito T , et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med 2011;364:514–523•• RADIANT3 study reporting Phase 3 study of Everolimus in pNETs which led to FDA approval
  • 23. Yao JC, Fazio N, Singh S , et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet 2016;387:968–977•• Latest RADIANT study reporting Phase 3 study of Everolimus in lung/GI NETs(RADIANT-4)
  • 24. Raymond E, Dahan L, Raoul JL , et al. Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N Engl J Med 2011;364:501–513 [DOI] [PubMed] [Google Scholar]
  • 25. Caplin ME, Pavel M, Cwikla Jb , et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014;371:224–233• Recent Phase 3 study leading to approval of Lanreotide for treatment of patients with advanced NETs
  • 26. Strosberg J, El-Haddad G, Wolin E , et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med 2017;376:125–135•• Phase 3 trial that led to recent approval of PRRT for treatment of metastatic NETs
  • 27. Kwekkeboom DJ, de Herder WW, Kam BL , et al. Treatment with the radiolabeled somatostatin analog [177 Lu-DOTA 0,Tyr3]octreotate: toxicity, efficacy, and survival. J Clin Oncol 2008;26:2124–2130 [DOI] [PubMed] [Google Scholar]
  • 28. Fidelman N, Kerlan RK, Hawkins RA Jr. , et al. Radioembolization with (90)Y glass microspheres for the treatment of unresectable metastatic liver disease from chemotherapy-refractory gastrointestinal cancers: final report of a prospective pilot study. J Gastrointest Oncol 2016;7:860–874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Benson AB III, Geschwind JF, Mulcahy MF , et al. Radioembolisation for liver metastases: results from a prospective 151 patient multi-institutional phase II study. Eur J Cancer 2013;49:3122–3130 [DOI] [PubMed] [Google Scholar]
  • 30. Kulke MH, Horsch D, Caplin ME , et al. Telotristat Ethyl, a Tryptophan Hydroxylase Inhibitor for the Treatment of Carcinoid Syndrome. J Clin Oncol 2017;35:14–23 [DOI] [PubMed] [Google Scholar]
  • 31. Vinik AI, Wolin EM, Liyanage N , et al. Evaluation of Lanreotide Depot/Autogel efficacy and safety as a carcinoid syndrome treatment(ELECT): A randomized, double-blind, placebo-controlled trial. Endocr Pract 2016;22:1068–1080 [DOI] [PubMed] [Google Scholar]
  • 32. Bosman FT, Carneiro F, Hruban RH et al. WHO World Health Organization Classification of Tumors and Genetics of the Digestive System. Lyon, France: IARC press; 2010. [Google Scholar]
  • 33. WHO classification of tumours of the lung, pleura, thymus and heart. Lyon, France: International Agency for Research on Cancer; 2015. [Google Scholar]
  • 34. Rindi G, Falconi M, Klersy C , et al. TNM Staging of Neoplasms of the Endocrine Pancreas: Results From a Large International Cohort Study. J Natl Cancer Inst 2012;104:764–777 [DOI] [PubMed] [Google Scholar]
  • 35. Luo G, Javed A, Strosberg JR , et al. Modified Staging Classification for Pancreatic Neuroendocrine Tumors on the Basis of the American Joint Committee on Cancer and European Neuroendocrine Tumor Society Systems. J Clin Oncol 2017;35:274–280 [DOI] [PubMed] [Google Scholar]
  • 36. Morin E, Cheng S, Mete O , et al. Hormone profiling, WHO 2010 grading, and AJCC/UICC staging in pancreatic neuroendocrine tumor behavior. Cancer Med 2013;2:701–711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Sorbye H, Welin S, Langer SW , et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): The NORDIC NEC study. Ann Oncol 2012; [DOI] [PubMed] [Google Scholar]
  • 38. Milione M, Maisonneuve P, Spada F , et al. The Clinicopathologic Heterogeneity of Grade 3 Gastroenteropancreatic Neuroendocrine Neoplasms: Morphological Differentiation and Proliferation Identify Different Prognostic Categories. Neuroendocrinology 2016; [DOI] [PubMed] [Google Scholar]
  • 39. Heetfeld M, Chougnet CN, Olsen IH , et al. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer 2015;22:657–664 [DOI] [PubMed] [Google Scholar]
  • 40. Hijioka S, Hosoda W, Matsuo K , et al. Rb Loss and KRAS Mutation Are Predictors of the Response to Platinum-Based Chemotherapy in Pancreatic Neuroendocrine Neoplasm with Grade 3: A Japanese Multicenter Pancreatic NEN-G3 Study. Clin Cancer Res 2017;23:4625–4632 [DOI] [PubMed] [Google Scholar]
  • 41. Cho MY, Kim JM, Sohn JH , et al. Current Trends of the Incidence and Pathological Diagnosis of Gastroenteropancreatic Neuroendocrine Tumors (GEP-NETs) in Korea 2000–2009: Multicenter Study. Cancer Res Treat 2012;44:157–165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Hijioka S, Hosoda W, Mizuno N , et al. Does the WHO 2010 classification of pancreatic neuroendocrine neoplasms accurately characterize pancreatic neuroendocrine carcinomas? J Gastroenterol 2015;50:564–572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Tang LH, Basturk O, Sue JJ , et al. A Practical Approach to the Classification of WHO Grade 3 (G3) Well-differentiated Neuroendocrine Tumor (WD-NET) and Poorly Differentiated Neuroendocrine Carcinoma (PD-NEC) of the Pancreas. Am J Surg Pathol 2016;40:1192–1202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Tang LH, Untch BR, Reidy DL , et al. Well-Differentiated Neuroendocrine Tumors with a Morphologically Apparent High-Grade Component: A Pathway Distinct from Poorly Differentiated Neuroendocrine Carcinomas. Clin Cancer Res 2016;22:1011–1017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Basturk O, Yang Z, Tang LH , et al. The high-grade (WHO G3) pancreatic neuroendocrine tumor category is morphologically and biologically heterogenous and includes both well differentiated and poorly differentiated neoplasms. Am J Surg Pathol 2015;39:683–690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. WHO classification of tumours of endocrine organs. Lyon, France: gency for Research on Cancer; 2017. [Google Scholar]
  • 47. Haug AR, Cindea-Drimus R, Auernhammer CJ , et al. The role of 68Ga-DOTATATE PET/CT in suspected neuroendocrine tumors. J Nucl Med 2012;53:1686–1692 [DOI] [PubMed] [Google Scholar]
  • 48. Haug AR, Cindea-Drimus R, Auernhammer CJ , et al. Neuroendocrine tumor recurrence: diagnosis with 68Ga-DOTATATE PET/CT. Radiology 2014;270:517–525 [DOI] [PubMed] [Google Scholar]
  • 49. Deppen SA, Liu E, Blume JD , et al. Safety and Efficacy of 68Ga-DOTATATE PET/CT for Diagnosis, Staging, and Treatment Management of Neuroendocrine Tumors. J Nucl Med 2016;57:708–714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Has Simsek D, Kuyumcu S, Turkmen C , et al. Can complementary 68Ga-DOTATATE and 18F-FDG PET/CT establish the missing link between histopathology and therapeutic approach in gastroenteropancreatic neuroendocrine tumors? J Nucl Med 2014;55:1811–1817 [DOI] [PubMed] [Google Scholar]
  • 51. Bahri H, Laurence L, Edeline J , et al. High prognostic value of 18F-FDG PET for metastatic gastroenteropancreatic neuroendocrine tumors: a long-term evaluation. J Nucl Med 2014;55:1786–1790 [DOI] [PubMed] [Google Scholar]
  • 52. Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol 2009;10:307–318 [DOI] [PubMed] [Google Scholar]
  • 53. Motzer RJ, Escudier B, Oudard S , et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008;372:449–456 [DOI] [PubMed] [Google Scholar]
  • 54. Krueger DA, Care MM, Holland K , et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med 2010;363:1801–1811 [DOI] [PubMed] [Google Scholar]
  • 55. Dantal J Everolimus: preventing organ rejection in adult kidney transplant recipients. Expert Opin Pharmacother 2012;13:767–778 [DOI] [PubMed] [Google Scholar]
  • 56. Baselga J, Campone M, Piccart M , et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med 2012;366:520–529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet 2004;43:83–95 [DOI] [PubMed] [Google Scholar]
  • 58. O’Donnell A, Faivre S, Burris HA, III , et al. Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J Clin Oncol 2008;26:1588–1595 [DOI] [PubMed] [Google Scholar]
  • 59. Pavel ME, Baudin E, Oberg KE , et al. Efficacy of everolimus plus octreotide LAR in patients with advanced neuroendocrine tumor and carcinoid syndrome: final overall survival from the randomized, placebo-controlled phase 3 RADIANT-2 study. Ann Oncol 2017; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Fazio N, Granberg D, Grossman A , et al. Everolimus plus octreotide long-acting repeatable in patients with advanced lung neuroendocrine tumors: analysis of the phase 3, randomized, placebo-controlled RADIANT-2 study. Chest 2013;143:955–962 [DOI] [PubMed] [Google Scholar]
  • 61. Castellano D, Bajetta E, Panneerselvam A , et al. Everolimus plus octreotide long-acting repeatable in patients with colorectal neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-2 study. Oncologist 2013;18:46–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Anthony LB, Pavel ME, Hainsworth JD , et al. Impact of Previous Somatostatin Analogue Use on the Activity of Everolimus in Patients with Advanced Neuroendocrine Tumors: Analysis from the Phase III RADIANT-2 Trial. Neuroendocrinology 2015;102:18–25 [DOI] [PubMed] [Google Scholar]
  • 63. Strosberg JR, Yao JC, Bajetta E , et al. Efficacy of octreotide long-acting repeatable in neuroendocrine tumors: RADIANT-2 placebo arm post hoc analysis. Endocr Relat Cancer 2015;22:933–940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Pavel ME, Becerra C, Grosch K , et al. Effect of everolimus on the pharmacokinetics of octreotide long-acting repeatable in patients with advanced neuroendocrine tumors: An analysis of the randomized phase III RADIANT-2 trial. Clin Pharmacol Ther 2017;101:462–468 [DOI] [PubMed] [Google Scholar]
  • 65. Yao JC, Pavel M, Lombard-Bohas C , et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers From the Randomized, Phase III RADIANT-3 Study. J Clin Oncol 2016;34:3906–3913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Ito T, Okusaka T, Ikeda M , et al. Everolimus for advanced pancreatic neuroendocrine tumours: a subgroup analysis evaluating Japanese patients in the RADIANT-3 trial. Jpn J Clin Oncol 2012;42:903–911 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Lombard-Bohas C, Yao JC, Hobday T , et al. Impact of prior chemotherapy use on the efficacy of everolimus in patients with advanced pancreatic neuroendocrine tumors: a subgroup analysis of the phase III RADIANT-3 trial. Pancreas 2015;44:181–189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Singh S, Carnaghi C, Buzzoni R , et al. Everolimus in Neuroendocrine Tumors of the Gastrointestinal Tract and Unknown Primary. Neuroendocrinology 2017; [DOI] [PubMed] [Google Scholar]
  • 69. Fazio N, Buzzoni R, Delle Fave G , et al. Everolimus in advanced, progressive, well-differentiated, non-functional neuroendocrine tumors: RADIANT-4 lung subgroup analysis. Cancer Sci 2018;109:174–181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Buzzoni R, Carnaghi C, Strosberg J , et al. Impact of prior therapies on everolimus activity: an exploratory analysis of RADIANT-4. Onco Targets Ther 2017;10:5013–5030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Pavel ME, Singh S, Strosberg JR , et al. Health-related quality of life for everolimus versus placebo in patients with advanced, non-functional, well-differentiated gastrointestinal or lung neuroendocrine tumours (RADIANT-4): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2017;18:1411–1422• Recent report of quality of life changes in patients from RADIANT-4 study
  • 72. Yao JC, Lombard-Bohas C, Baudin E , et al. Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. J Clin Oncol 2010;28:69–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Bajetta E, Catena L, Fazio N , et al. Everolimus in combination with octreotide long-acting repeatable in a first-line setting for patients with neuroendocrine tumors: an ITMO group study. Cancer 2014;120:2457–2463 [DOI] [PubMed] [Google Scholar]
  • 74. Bajetta E, Catena L, Pusceddu S , et al. Everolimus in Combination with Octreotide LAR in First Line Setting for Patients with Neuroendocrine Tumors: A 5- Years Update. Neuroendocrinology 2017; [DOI] [PubMed] [Google Scholar]
  • 75. Kulke MH, Ruszniewski P, Van Cutsem E , et al. A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann Oncol 2017;28:1309–1315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ferolla P, Brizzi MP, Meyer T , et al. Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 2017;18:1652–1664 [DOI] [PubMed] [Google Scholar]
  • 77. Chan JA, Ryan DP, Zhu AX , et al. Phase I study of pasireotide (SOM 230) and everolimus (RAD001) in advanced neuroendocrine tumors. Endocr Relat Cancer 2012;19:615–623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Dasari A, Phan A, Gupta S , et al. Phase I study of the anti-IGF1R antibody cixutumumab with everolimus and octreotide in advanced well-differentiated neuroendocrine tumors. Endocr Relat Cancer 2015;22:431–441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kim HS, Shaib WL, Zhang C , et al. Phase 1b study of pasireotide, everolimus, and selective internal radioembolization therapy for unresectable neuroendocrine tumors with hepatic metastases. Cancer 2018; [DOI] [PubMed] [Google Scholar]
  • 80. O’Reilly T, McSheehy PM. Biomarker Development for the Clinical Activity of the mTOR Inhibitor Everolimus (RAD001): Processes, Limitations, and Further Proposals. Transl Oncol 2010;3:65–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. von Wichert G, Jehle PM, Hoeflich A , et al. Insulin-like growth factor-I is an autocrine regulator of chromogranin A secretion and growth in human neuroendocrine tumor cells. Cancer Res 2000;60:4573–4581 [PubMed] [Google Scholar]
  • 82. Weckbecker G, Briner U, Lewis I , et al. SOM230: a new somatostatin peptidomimetic with potent inhibitory effects on the growth hormone/insulin-like growth factor-I axis in rats, primates, and dogs. Endocrinology 2002;143:4123–4130 [DOI] [PubMed] [Google Scholar]
  • 83. Chan JA, Blaszkowsky L, Stuart K , et al. A prospective, phase 1/2 study of everolimus and temozolomide in patients with advanced pancreatic neuroendocrine tumor. Cancer 2013; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Chan JA, Mayer RJ, Jackson N , et al. Phase I study of sorafenib in combination with everolimus (RAD001) in patients with advanced neuroendocrine tumors. Cancer Chemother Pharmacol 2013;71:1241–1246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Claringbold PG, Turner JH. NeuroEndocrine Tumor Therapy with Lutetium-177-octreotate and Everolimus (NETTLE): A Phase I Study. Cancer Biother Radiopharm 2015;30:261–269 [DOI] [PubMed] [Google Scholar]
  • 86. Salazar R, Garcia-Carbonero R, Libutti SK , et al. Phase II Study of BEZ235 versus Everolimus in Patients with Mammalian Target of Rapamycin Inhibitor-Naive Advanced Pancreatic Neuroendocrine Tumors. Oncologist 2017; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Oh DY, Kim TW, Park YS , et al. Phase 2 study of everolimus monotherapy in patients with nonfunctioning neuroendocrine tumors or pheochromocytomas/paragangliomas. Cancer 2012;118:6162–6170 [DOI] [PubMed] [Google Scholar]
  • 88. Hobday TJ, Qin R, Reidy-Lagunes D , et al. Multicenter Phase II Trial of Temsirolimus and Bevacizumab in Pancreatic Neuroendocrine Tumors. J Clin Oncol 2015;33:1551–1556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Berruti A, Fazio N, Ferrero A , et al. Bevacizumab plus octreotide and metronomic capecitabine in patients with metastatic well-to-moderately differentiated neuroendocrine tumors: the XELBEVOCT study. BMC Cancer 2014;14:184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Chan JA, Stuart K, Earle CC , et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J Clin Oncol 2012;30:2963–2968 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Panzuto F, Rinzivillo M, Fazio N , et al. Real-world study of everolimus in advanced progressive neuroendocrine tumors. Oncologist 2014;19:966–974• Recent real world study of everolimus in patients with advanced panNETs
  • 92. Berardi R, Torniai M, Pusceddu S , et al. Prognostic impact of the cumulative dose and dose intensity of everolimus in patients with pancreatic neuroendocrine tumors. Cancer Med 2017;6:1493–1499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Liu CT, Chen MH, Chen JS , et al. The efficacy and safety of everolimus for the treatment of progressive gastroenteropancreatic neuroendocrine tumors: A multi-institution observational study in Taiwan . Asia Pac J Clin Oncol 2016;12:396–402 [DOI] [PubMed] [Google Scholar]
  • 94. Lee L, Ito T, Igarashi H , et al. Impact of everolimus on Japanese patients with advanced pancreatic neuroendocrine neoplasms. J Hepatobiliary Pancreat Sci 2017;24:95–102 [DOI] [PubMed] [Google Scholar]
  • 95. Lee KJ, Cho JH, Lee SH , et al. Clinical outcomes of everolimus in patients with advanced, nonfunctioning pancreatic neuroendocrine tumors: a multicenter study in Korea. Cancer Chemother Pharmacol 2017;80:799–805 [DOI] [PubMed] [Google Scholar]
  • 96. Yao JC, Phan AT, Hess K , et al. Perfusion computed tomography as functional biomarker in randomized run-in study of bevacizumab and everolimus in well-differentiated neuroendocrine tumors. Pancreas 2015;44:190–197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Kamp K, Gumz B, Feelders RA , et al. Safety and efficacy of everolimus in gastrointestinal and pancreatic neuroendocrine tumors after (177)Lu-octreotate. Endocr Relat Cancer 2013;20:825–831 [DOI] [PubMed] [Google Scholar]
  • 98. Angelousi A, Kamp K, Kaltsatou M , et al. Sequential Everolimus and Sunitinib Treatment in Pancreatic Metastatic Well-Differentiated Neuroendocrine Tumours Resistant to Prior Treatments. Neuroendocrinology 2017;105:394–402 [DOI] [PubMed] [Google Scholar]
  • 99. Yoo C, Cho H, Song MJ , et al. Efficacy and safety of everolimus and sunitinib in patients with gastroenteropancreatic neuroendocrine tumor. Cancer Chemother Pharmacol 2017;79:139–146 [DOI] [PubMed] [Google Scholar]
  • 100. Capdevila J, Sevilla I, Alonso V , et al. Evaluation of the efficacy and safety of lanreotide in combination with targeted therapies in patients with neuroendocrine tumours in clinical practice: a retrospective cross-sectional analysis. BMC Cancer 2015;15:495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Wolin EM. Long-term everolimus treatment of patients with pancreatic neuroendocrine tumors. Chemotherapy 2014;60:143–150 [DOI] [PubMed] [Google Scholar]
  • 102. Ter-Minassian M, Zhang S, Brooks NV , et al. Association Between Tumor Progression Endpoints and Overall Survival in Patients with Advanced Neuroendocrine Tumors. Oncologist 2017;22:165–172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Ito T, Lee L, Jensen RT. Treatment of symptomatic neuroendocrine tumor syndromes: recent advances and controversies. Expert Opin Pharmacother 2016;17:2191–2205• Summary of the ability of everolimus and other agents to control functional NET syndromes
  • 104. Ito T, Lee L, Jensen RT. Carcinoid-syndrome: recent advances, current status and controversies. Curr Opin Endocrinol Diabetes Obes 2018;25:22–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Falconi M, Eriksson B, Kaltsas G , et al. ENETS Consensus Guidelines Update for the Management of Patients with Functional Pancreatic Neuroendocrine Tumors and Non-Functional Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2016;103:153–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Strosberg JR, Halfdanarson TR, Bellizzi AM , et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Midgut Neuroendocrine Tumors. Pancreas 2017;46:707–714• Recent guidelines for treatment of midgut carcinoids including use of everolimus
  • 107. Roy PK, Venzon DJ, Feigenbaum KM , et al. Gastric secretion in Zollinger-Ellison syndrome: correlation with clinical expression, tumor extent and role in diagnosis - A prospective NIH study of 235 patients and review of the literature in 984 cases. Medicine(Baltimore) 2001;80:189–222 [DOI] [PubMed] [Google Scholar]
  • 108. Berna MJ, Hoffmann KM, Serrano J , et al. Serum gastrin in Zollinger-Ellison syndrome: I. Prospective study of fasting serum gastrin in 309 patients from the National Institutes of Health and comparison with 2229 cases from the literature. Medicine (Baltimore) 2006;85:295–330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Berna MJ, Hoffmann KM, Long SH , et al. Serum gastrin in Zollinger-Ellison syndrome: II. Prospective study of gastrin provocative testing in 293 patients from the National Institutes of Health and comparison with 537 cases from the literature. evaluation of diagnostic criteria, proposal of new criteria, and correlations with clinical and tumoral features. Medicine (Baltimore) 2006;85:331–364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Ito T, Igarashi H, Jensen RT. Zollinger-Ellison syndrome: Recent advances and controversies. Current Opinion in Gastroenterology 2013;29:650–661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Metz DC, Jensen RT. Gastrointestinal neuroendocrine tumors:; Pancreatic endocrine tumors. Gastroenterology 2008;135:1469–1492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Collen MJ, Howard JM, McArthur KE , et al. Comparison of ranitidine and cimetidine in the treatment of gastric hypersecretion. Ann Intern Med 1984;100:52–58 [DOI] [PubMed] [Google Scholar]
  • 113. Ito T, Igarashi H, Uehara H , et al. Pharmacotherapy of Zollinger-Ellison syndrome. Expert Opin Pharmacotherapy 2013;14:307–321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: Advances . Best Pract Res Clin Gastroenterol 2012;26:737–753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Ito T, Igarashi H, Jensen RT. Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances. J Gastroenterol 2012;47:941–960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Stehouwer CD, Lems WF, Fischer HR , et al. Aggravation of hypoglycemia in insulinoma patients by the long-acting somatostatin analogue octreotide (Sandostatin). Acta Endocrinol (Copenh) 1989;121:34–40 [DOI] [PubMed] [Google Scholar]
  • 117. Pavel ME, Chen D, He W , et al. Everolimus Effect on Gastrin and Glucagon in Pancreatic Neuroendocrine Tumors. Pancreas 2017;46:751–757 [DOI] [PubMed] [Google Scholar]
  • 118. Capdevila J, Diez Miranda I, Obiols G , et al. Control of carcinoid syndrome with everolimus. Ann Oncol 2011;22:237–239 [DOI] [PubMed] [Google Scholar]
  • 119. Bainbridge HE, Larbi E, Middleton G. Symptomatic Control of Neuroendocrine Tumours with Everolimus . Horm Cancer 2015;6:254–259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Houde VP, Brule S, Festuccia WT , et al. Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue. Diabetes 2010;59:1338–1348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Bernard V, Lombard-Bohas C, Taquet MC , et al. Efficacy of Everolimus in Patients with Metastatic Insulinoma and Refractory Hypoglycemia. Eur J Endocrinol 2013;168:665–674 [DOI] [PubMed] [Google Scholar]
  • 122. Bozkirli E, Bakiner O, Abali H , et al. A case of inoperable malignant insulinoma with resistant hypoglycemia who experienced the most significant clinical improvement with everolimus. Case Rep Endocrinol 2013;2013:636175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Asayama M, Yamada-Murano T, Hara H , et al. Everolimus dramatically improves glycemic control in unresectable metastatic insulinoma: a case report. Jpn J Clin Oncol 2014;44:186–190 [DOI] [PubMed] [Google Scholar]
  • 124. Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med 2009;360:195–197 [DOI] [PubMed] [Google Scholar]
  • 125. Fiebrich HB, Siemerink EJ, Brouwers AH , et al. Everolimus induces rapid plasma glucose normalization in insulinoma patients by effects on tumor as well as normal tissues. Oncologist 2011;16:783–787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Thomas NJ, Brooke AM, Besser GM. Long-term maintenance of normoglycaemia using everolimus in a patient with disseminated insulinoma and severe hypoglycaemia. Clin Endocrinol (Oxf) 2013;78:799–800 [DOI] [PubMed] [Google Scholar]
  • 127. Scharf M, Mueller D, Koenig U , et al. Management of a metastasized high grade insulinoma (G3) with refractory hypoglycemia: case report and review of the literature. Pancreatology 2014;14:542–545 [DOI] [PubMed] [Google Scholar]
  • 128. Garcia-Carbonero R, Sorbye H, Baudin E , et al. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology 2016;103:186–194 [DOI] [PubMed] [Google Scholar]
  • 129. Panzuto F, Rinzivillo M, Spada F , et al. Everolimus in Pancreatic Neuroendocrine Carcinomas G3. Pancreas 2017;46:302–305• Small series of G3NETs well differentiated panNETs successfully treated with everolimus
  • 130. Genc CG, Klumpen HJ, Denecke T , et al. Successful treatment of high-grade pancreatic neuroendocrine neoplasms with everolimus. Acta Oncol 2017;1–3 [DOI] [PubMed] [Google Scholar]
  • 131. Fonseca PJ, Uriol E, Galvan JA , et al. Prolonged clinical benefit of everolimus therapy in the management of high-grade pancreatic neuroendocrine carcinoma. Case Rep Oncol 2013;6:441–449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Tanaka H, Matsusaki S, Baba Y , et al. Neuroendocrine tumor G3: a pancreatic well-differentiated neuroendocrine tumor with a high proliferative rate. Clin J Gastroenterol 2015;8:414–420 [DOI] [PubMed] [Google Scholar]
  • 133. Chung DC, Brown SB, Graeme-Cook F , et al. Localization of putative tumor suppressor loci by genome-wide allelotyping in human pancreatic endocrine tumors. Cancer Res 1998;58:3706–3711 [PubMed] [Google Scholar]
  • 134. Bollard J, Couderc C, Blanc M , et al. Antitumor effect of everolimus in preclinical models of high-grade gastroenteropancreatic neuroendocrine carcinomas. Neuroendocrinology 2013;97:331–340 [DOI] [PubMed] [Google Scholar]
  • 135. Gilabert M, Rho YS, Kavan P. Targeted Therapies Provide Treatment Options for Poorly Differentiated Pancreatic Neuroendocrine Carcinomas. Oncology 2017;92:170–172 [DOI] [PubMed] [Google Scholar]
  • 136. Tarhini A, Kotsakis A, Gooding W , et al. Phase II study of everolimus (RAD001) in previously treated small cell lung cancer. Clin Cancer Res 2010;16:5900–5907 [DOI] [PubMed] [Google Scholar]
  • 137.Sun JM, Kim JR, Do IG , et al. A phase-1b study of everolimus plus paclitaxel in patients with small-cell lung cancer. Br J Cancer 2013;109:1482–1487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Christopoulos P, Engel-Riedel W, Grohe C , et al. Everolimus with paclitaxel and carboplatin as first-line treatment for metastatic large-cell neuroendocrine lung carcinoma: a multicenter phase II trial. Ann Oncol 2017;28:1898–1902 [DOI] [PubMed] [Google Scholar]
  • 139. Dasari A, Mehta K, Byers LA , et al. Comparative study of lung and extrapulmonary poorly differentiated neuroendocrine carcinomas: A SEER database analysis of 162,983 cases. Cancer 2018;124:807–815 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Martins D, Spada F, Lambrescu I , et al. Predictive Markers of Response to Everolimus and Sunitinib in Neuroendocrine Tumors. Target Oncol 2017;12:611–622• Summary of the predictive markers of response to everolimus and sunitinib
  • 141. Yao JC, Pavel M, Phan AT , et al. Chromogranin A and Neuron-Specific Enolase as Prognostic Markers in Patients with Advanced pNET Treated with Everolimus. J Clin Endocrinol Metab 2011;96:3741–3749 [DOI] [PubMed] [Google Scholar]
  • 142. Serra S, Zheng L, Hassan M , et al. The FGFR4-G388R single-nucleotide polymorphism alters pancreatic neuroendocrine tumor progression and response to mTOR inhibition therapy. Cancer Res 2012;72:5683–5691 [DOI] [PubMed] [Google Scholar]
  • 143. Cros J, Moati E, Raffenne J , et al. Gly388Arg FGFR4 Polymorphism Is Not Predictive of Everolimus Efficacy in Well-Differentiated Digestive Neuroendocrine Tumors. Neuroendocrinology 2016;103:495–499 [DOI] [PubMed] [Google Scholar]
  • 144. Meric-Bernstam F, Akcakanat A, Chen H , et al. PIK3CA/PTEN Mutations and Akt Activation As Markers of Sensitivity to Allosteric mTOR Inhibitors. Clin Cancer Res 2012;18:1777–1789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Gagliano T, Bellio M, Gentilin E , et al. mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr Relat Cancer 2013;20:463–475 [DOI] [PubMed] [Google Scholar]
  • 146. Benslama N, Bollard J, Vercherat C , et al. Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Invest New Drugs 2016;34:654–662 [DOI] [PubMed] [Google Scholar]
  • 147. Boulay A, Zumstein-Mecker S, Stephan C , et al. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res 2004;64:252–261 [DOI] [PubMed] [Google Scholar]
  • 148. Tanaka C, O’Reilly T, Kovarik JM , et al. Identifying optimal biologic doses of everolimus (RAD001) in patients with cancer based on the modeling of preclinical and clinical pharmacokinetic and pharmacodynamic data. J Clin Oncol 2008;26:1596–1602 [DOI] [PubMed] [Google Scholar]
  • 149. van Asselt SJ, Oosting SF, Brouwers AH , et al. Everolimus Reduces (89)Zr-Bevacizumab Tumor Uptake in Patients with Neuroendocrine Tumors. J Nucl Med 2014;55:1087–1092 [DOI] [PubMed] [Google Scholar]
  • 150. Rugo HS, Hortobagyi GN, Yao J , et al. Meta-analysis of stomatitis in clinical studies of everolimus: incidence and relationship with efficacy. Ann Oncol 2016;27:519–525 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Abdel-Rahman O, Fouad M. Risk of mucocutaneous toxicities in patients with solid tumors treated with everolimus; a systematic review and meta-analysis. Expert Rev Anticancer Ther 2014;14:1529–1536 [DOI] [PubMed] [Google Scholar]
  • 152. Ravaud A, Urva SR, Grosch K , et al. Relationship between everolimus exposure and safety and efficacy: meta-analysis of clinical trials in oncology. Eur J Cancer 2014;50:486–495• Meta-analysis of safety efficacy of everolimus in all tumors related to exposure time
  • 153. Shameem R, Lacouture M, Wu S. Incidence and risk of rash to mTOR inhibitors in cancer patients--a meta-analysis of randomized controlled trials. Acta Oncol 2015;54:124–132 [DOI] [PubMed] [Google Scholar]
  • 154. Ramirez-Fort MK, Case EC, Rosen AC , et al. Rash to the mTOR inhibitor everolimus: systematic review and meta-analysis. Am J Clin Oncol 2014;37:266–271 [DOI] [PubMed] [Google Scholar]
  • 155. Abdel-Rahman O, Fouad M. Risk of fatigue and hepatic and metabolic toxicities in patients with solid tumors treated with everolimus: a meta-analysis. Future Oncol 2015;11:79–90 [DOI] [PubMed] [Google Scholar]
  • 156. Garcia CA, Wu S. Attributable Risk of Infection to mTOR Inhibitors Everolimus and Temsirolimus in the Treatment of Cancer . Cancer Invest 2016;34:521–530 [DOI] [PubMed] [Google Scholar]
  • 157. Iacovelli R, Palazzo A, Mezi S , et al. Incidence and risk of pulmonary toxicity in patients treated with mTOR inhibitors for malignancy. A meta-analysis of published trials. Acta Oncol 2012;51:873–879 [DOI] [PubMed] [Google Scholar]
  • 158. Noguchi S, Shinohara N, Ito T , et al. Relationship between Pulmonary Adverse Events and Everolimus Exposure in Japanese and Non-Japanese Patients: A Meta-Analysis of Oncology Trials. Oncology 2017;92:243–254 [DOI] [PubMed] [Google Scholar]
  • 159. Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: A meta-analysis. Acta Oncol 2016;55:1196–1203 [DOI] [PubMed] [Google Scholar]
  • 160. Xu J, Tian D. Hematologic toxicities associated with mTOR inhibitors temsirolimus and everolimus in cancer patients: a systematic review and meta-analysis. Curr Med Res Opin 2014;30:67–74 [DOI] [PubMed] [Google Scholar]
  • 161. Shameem R, Hamid MS, Wu S. Risk of anemia attributable to everolimus in patients with cancer: a meta-analysis of randomized controlled trials. Anticancer Res 2015;35:2333–2340 [PubMed] [Google Scholar]
  • 162. Qi WX, Huang YJ, Yao Y , et al. Incidence and risk of treatment-related mortality with mTOR inhibitors everolimus and temsirolimus in cancer patients: a meta-analysis. PLoS ONE 2013;8:e65166.• Meta-analysis of mortality due to mTOR inhibitors in cancer patients
  • 163. Rogers SC, Garcia CA, Wu S. Discontinuation of Everolimus Due to Related and Unrelated Adverse Events in Cancer Patients: A Meta-Analysis . Cancer Invest 2017;35:552–561• Meta-analysis of discontinuation of therapy in patients with cancer treated with everolimus
  • 164. Choueiri TK, Je Y, Sonpavde G , et al. Incidence and risk of treatment-related mortality in cancer patients treated with the mammalian target of rapamycin inhibitors. Ann Oncol 2013;24:2092–2097 [DOI] [PubMed] [Google Scholar]
  • 165. Plante A, Baudin E, Do Cao C , et al. Patient-reported tolerance in treatments approved in neuroendocrine tumors: A national survey from the French Group of Endocrine Tumors. Clin Res Hepatol Gastroenterol 2018. (In press)0029• Recent study assessing tolerances to various antitumor treatments in patients with advanced NETs. including everolimus
  • 166. Pavel M, Unger N, Borbath I , et al. Safety and QOL in Patients with Advanced NET in a Phase 3b Expanded Access Study of Everolimus. Target Oncol 2016;11:667–675 [DOI] [PubMed] [Google Scholar]
  • 167. Deppenweiler M, Falkowski S, Saint-Marcoux F , et al. Towards therapeutic drug monitoring of everolimus in cancer? Results of an exploratory study of exposure-effect relationship. Pharmacol Res 2017;121:138–144 [DOI] [PubMed] [Google Scholar]
  • 168. Aapro M, Andre F, Blackwell K , et al. Adverse event management in patients with advanced cancer receiving oral everolimus: focus on breast cancer. Ann Oncol 2014;25:763–773 [DOI] [PubMed] [Google Scholar]
  • 169. Hsieh AC, Costa M, Zollo O , et al. Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E. Cancer Cell 2010;17:249–261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell 2006;124:471–484 [DOI] [PubMed] [Google Scholar]
  • 171. Houghton PJ. Everolimus. Clin Cancer Res 2010;16:1368–1372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Tamburini J, Chapuis N, Bardet V , et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood 2008;111:379–382 [DOI] [PubMed] [Google Scholar]
  • 173. Kuo SH, Hsu CH, Chen LT , et al. Lack of compensatory pAKT activation and eIF4E phosphorylation of lymphoma cells towards mTOR inhibitor, RAD001. Eur J Cancer 2011;47:1244–1257 [DOI] [PubMed] [Google Scholar]
  • 174. Svejda B, Kidd M, Kazberouk A , et al. Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer 2011;117:4141–4154 [DOI] [PubMed] [Google Scholar]
  • 175. Wang X, Yue P, Kim YA , et al. Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/raptor inhibition-initiated, mTOR/rictor-independent Akt activation. Cancer Res 2008;68:7409–7418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Choo AY, Yoon SO, Kim SG , et al. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A 2008;105:17414–17419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Kang SA, Pacold ME, Cervantes CL , et al. mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science 2013;341:1236566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Gupta M, Ansell SM, Novak AJ , et al. Inhibition of histone deacetylase overcomes rapamycin-mediated resistance in diffuse large B-cell lymphoma by inhibiting Akt signaling through mTORC2. Blood 2009;114:2926–2935 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Sarbassov DD, Guertin DA, Ali SM , et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307:1098–1101 [DOI] [PubMed] [Google Scholar]
  • 180. Stan R, McLaughlin MM, Cafferkey R , et al. Interaction between FKBP12-rapamycin and TOR involves a conserved serine residue. J Biol Chem 1994;269:32027–32030 [PubMed] [Google Scholar]
  • 181. Carracedo A, Ma L, Teruya-Feldstein J , et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest 2008;118:3065–3074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Di Nicolantonio F, Arena S, Tabernero J , et al. Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus. J Clin Invest 2010;120:2858–2866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Vandamme T, Beyens M, de Beeck KO , et al. Long-term acquired everolimus resistance in pancreatic neuroendocrine tumours can be overcome with novel PI3K-AKT-mTOR inhibitors. Br J Cancer 2016;114:650–658• Study of tumor resistance to everolimus
  • 184. Antonuzzo L, Del RM, Barucca V , et al. Critical focus on mechanisms of resistance and toxicity of m-TOR inhibitors in pancreatic neuroendocrine tumors. Cancer Treat Rev 2017;57:28–35 [DOI] [PubMed] [Google Scholar]
  • 185. Freitag H, Christen F, Lewens F , et al. Inhibition of mTOR’s Catalytic Site by PKI-587 Is a Promising Therapeutic Option for Gastroenteropancreatic Neuroendocrine Tumor Disease. Neuroendocrinology 2017;105:90–104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Passacantilli I, Capurso G, Archibugi L , et al. Combined therapy with RAD001 e BEZ235 overcomes resistance of PET immortalized cell lines to mTOR inhibition. Oncotarget 2014;5:5381–5391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Zitzmann K, Ruden J, Brand S , et al. Compensatory activation of Akt in response to mTOR and Raf inhibitors - a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease. Cancer Lett 2010;295:100–109 [DOI] [PubMed] [Google Scholar]
  • 188. Bendell JC, Kurkjian C, Infante JR , et al. A phase 1 study of the sachet formulation of the oral dual PI3K/mTOR inhibitor BEZ235 given twice daily (BID) in patients with advanced solid tumors. Invest New Drugs 2015;33:463–471 [DOI] [PubMed] [Google Scholar]
  • 189. Fazio N, Buzzoni R, Baudin E , et al. A Phase II Study of BEZ235 in Patients with Everolimus-resistant, Advanced Pancreatic Neuroendocrine Tumours. Anticancer Res 2016;36:713–719• Phase 2 study of the dual inhibitor BEZ235 in Everolimus resistant patients
  • 190. Rinke A, Muller HH, Schade-Brittinger C , et al. 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;27:4656–4663 [DOI] [PubMed] [Google Scholar]
  • 191. Faivre S, Niccoli P, Castellano D , et al. Sunitinib in Pancreatic Neuroendocrine Tumors: Updated Progression-Free Survival and Final Overall Survival From a Phase III Randomized Study. Ann Oncol 2016; [DOI] [PubMed] [Google Scholar]
  • 192. Dilz LM, Denecke T, Steffen IG , et al. Streptozocin/5-fluorouracil chemotherapy is associated with durable response in patients with advanced pancreatic neuroendocrine tumours. Eur J Cancer 2015;51:1253–1262 [DOI] [PubMed] [Google Scholar]
  • 193. Clewemar Antonodimitrakis P, Sundin A, Wassberg C , et al. Streptozocin and 5-Fluorouracil for the Treatment of Pancreatic Neuroendocrine Tumors: Efficacy, Prognostic Factors and Toxicity. Neuroendocrinology 2016;103:345–353 [DOI] [PubMed] [Google Scholar]
  • 194. Cives M, Ghayouri M, Morse B , et al. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer 2016;23:759–767 [DOI] [PubMed] [Google Scholar]
  • 195. von Schrenck T, Howard JM, Doppman JL , et al. Prospective study of chemotherapy in patients with metastatic gastrinoma. Gastroenterology 1988;94:1326–1334 [DOI] [PubMed] [Google Scholar]
  • 196. Crona J, Fanola I, Lindholm DP , et al. Effect of temozolomide in patients with metastatic bronchial carcinoids. Neuroendocrinology 2013;98:151–155 [DOI] [PubMed] [Google Scholar]
  • 197. Crona J, Bjorklund P, Welin S , et al. Treatment, prognostic markers and survival in thymic neuroendocrine tumours. a study from a single tertiary referral centre. Lung Cancer 2013;79:289–293 [DOI] [PubMed] [Google Scholar]
  • 198. Kulke MH, Stuart K, Enzinger PC , et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. J Clin Oncol 2006;24:401–406 [DOI] [PubMed] [Google Scholar]
  • 199. Krug S, Gress TM, Michl P , et al. The Role of Cytotoxic Chemotherapy in Advanced Pancreatic Neuroendocrine Tumors. Digestion 2017;96:67–75 [DOI] [PubMed] [Google Scholar]
  • 200. Partelli S, Bartsch DK, Capdevila J , et al. ENETS Consensus Guidelines for Standard of Care in Neuroendocrine Tumours: Surgery for Small Intestinal and Pancreatic Neuroendocrine Tumours. Neuroendocrinology 2017;105:255–265 [DOI] [PubMed] [Google Scholar]
  • 201. Howe JR, Cardona K, Fraker DL , et al. The Surgical Management of Small Bowel Neuroendocrine Tumors: Consensus Guidelines of the North American Neuroendocrine Tumor Society. Pancreas 2017;46:715–731 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Singh S, Dey C, Kennecke H , et al. Consensus Recommendations for the Diagnosis and Management of Pancreatic Neuroendocrine Tumors: Guidelines from a Canadian National Expert Group. Ann Surg Oncol 2015;22:2685–2699 [DOI] [PubMed] [Google Scholar]
  • 203. Oberg K, Knigge U, Kwekkeboom D , et al. Neuroendocrine gastro-entero-pancreatic tumors: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2012;23 Suppl 7:vii124-vii130 [DOI] [PubMed] [Google Scholar]
  • 204. Caplin ME, Baudin E, Ferolla P , et al. Pulmonary neuroendocrine (carcinoid) tumors: European Neuroendocrine Tumor Society expert consensus and recommendations for best practice for typical and atypical pulmonary carcinoids. Ann Oncol 2015;26:1604–1620 [DOI] [PubMed] [Google Scholar]
  • 205. Vinik A, Bottomley A, Korytowsky B , et al. Patient-Reported Outcomes and Quality of Life with Sunitinib Versus Placebo for Pancreatic Neuroendocrine Tumors: Results From an International Phase III Trial. Target Oncol 2016;11:815–824 [DOI] [PubMed] [Google Scholar]
  • 206. Marinova M, Mucke M, Mahlberg L , et al. Improving quality of life in patients with pancreatic neuroendocrine tumor following peptide receptor radionuclide therapy assessed by EORTC QLQ-C30. Eur J Nucl Med Mol Imaging 2018;45:38–46 [DOI] [PubMed] [Google Scholar]
  • 207. Pavel M, O’Toole D, Costa F , et al. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology 2016;103:172–185•• Recent ENETs guidelines for the treatment of advanced disease in NET patients including use of everolimus
  • 208. Kulke MH, Shah MH, Benson AB , et al. Neuroendocrine Tumors: version 3.2017: NCCN Clinical Practice Guidelines in Oncology. NCCN Clinical Practice Guidelines in Oncology 2017;1–116• Latest NCCN guidelines for treatment of advanced NETs including use of everolimus
  • 209. Janson ET, Sorbye H, Welin S , et al. Nordic guidelines 2014 for diagnosis and treatment of gastroenteropancreatic neuroendocrine neoplasms. Acta Oncol 2014;53:1284–1297 [DOI] [PubMed] [Google Scholar]
  • 210. Kulke MH, Anthony LB, Bushnell DL , et al. NANETS Treatment Guidelines: Well-Differentiated Neuroendocrine Tumors of the Stomach and Pancreas. Pancreas 2010;39:735–752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Strosberg JR, Coppola D, Klimstra DS , et al. The NANETS Consensus Guidelines for the Diagnosis and Management of Poorly Differentiated (High-Grade) Extrapulmonary Neuroendocrine Carcinomas . Pancreas 2010;39:799–800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Hicks RJ, Kwekkeboom DJ, Krenning E , et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology 2017;105:295–309 [DOI] [PubMed] [Google Scholar]
  • 213. Bodei L, Kidd M, Paganelli G , et al. Long-term tolerability of PRRT in 807 patients with neuroendocrine tumours: the value and limitations of clinical factors. Eur J Nucl Med Mol Imaging 2015;42:5–19 [DOI] [PubMed] [Google Scholar]
  • 214. Shipkova M, Hesselink DA, Holt DW , et al. Therapeutic Drug Monitoring of Everolimus: A Consensus Report. Ther Drug Monit 2016;38:143–169 [DOI] [PubMed] [Google Scholar]

RESOURCES