Abstract
Introduction
Neuroendocrine tumors(NETs), once thought rare, are increasing in frequency in most countries and receiving increasing-attention. NETs present two-treatment problems. A proportion is functional, hormone-excess-state(F-NET), each of which must be treated. Recently, there are many advances, well-covered in reviews/consensus papers on imaging-NETs;new, novel anti-tumor treatments and understanding their pathogenesis. However, little attention has been paid to advances in the treatment of the hormone-excess-state. These advances are usually reported in case-series, and case-reports with few large studies. In this paper these advances are reviewed.
Areas covered
Advances in the last 5-years are concentrated on, but a review up to 10-years was performed. PubMed and other databases(Cochrane, etc) were searched for F-NET-syndromes including carcinoid-syndrome, as well as meeting-abstracts on NETs. All advances that controlled hormone-excess-states or facilitated-control were covered. These include new medical-therapies[serotonin-synthesis inhibitors(telotristat), Pasireotide, new agents for treating ACTHomas], increased dosing with conventional therapies(octreotide-LAR, Lanreotide-Autogel), mTor inhibitors(everolimus), Tyrosine-kinase inhibitors(sunitinib), cytoreductive surgery, liver-directed therapies(embolization, chemoembolization, radioembolization, RFA), peptide radio-receptor-therapy(PRRT) and 131I-MIBG, ablation of primary F-NETs.
Expert Opinion
Although many of the newer therapies controlling the hormone-excess-states in F-NETs are reported in relatively few patients, all the approaches show promise. Their description also generates some controversies/unresolved areas, such as the order of these new treatments, their longterm-efficacy, and effectiveness of combinations which may require large, controlled studies.
Keywords: Acid hypersecretion, carcinoid syndrome, chemoembolization, chemotherapy, cytoreductive surgery, embolization, gastrinoma, insulinoma, Lanreotide, multiple endocrine Neoplasia type 1, neuroendocrine tumor, pancreatic endocrine tumor, peptide radio-receptor therapy, octreotide, radioembolization, radio-frequency ablation, SIRT, somatostatin analogues, telotristat, Zollinger-Ellison syndrome
1. Introduction
Almost 70% of neuroendocrine tumors(NETs) occur in the gastrointestinal tract(GI) and they have long fascinated clinicians, because they can produce florid, specific clinical syndromes, secondary to their unique secretory products[1, 2]. All gastrointestinal tract NETs have many similarities in both their cytochemical properties, with characteristic expression of specific proteins(neuron specific enolase, syntaptophysin, chromogranin A[CgA]), ultrastructural features with electron dense granules, their ability to produce multiple amine/peptides and their charcterististic histological appearance with generally uniform nuclei and cytoplasm[2]. The gastrointestinal NETs include both pancreatic neuroendocrine tumors(pNETs) as well as NETs from other gastrointestinal sites(GI-NETs)(carcinoids) [2]
Recent studies demonstrate gastrointestinal NETS are not as infrequency as is commonly believed and in fact, both pNETs and GI-NETS(carcinoids) are increasing in frequency [3–5]. Although this very well many represent increased detection, nevertheless, clinicians will be seeing an increasing number of these patients. Patients with NETs can have two distinct therapeutic problems that both need to be dealt with. Numerous studies demonstrate that a significant proportion of NETs can have aggressive growth with the development of metastatic disease and in addition, up to 30% of patients with pNETs and 3–13% with GI-NETs(carcinoids) of the small intestine, a specific hormone-excess state is present[2]. Although curative resection would cure both problems, in many cases, because of the extent of disease, this is not possible and therefore treatment must be directed at each of these two problems [2, 6, ••8]. Historically, primarily for control of aggressive growth, both chemotherapy and interferon teatment has been used, however, particularly with chemotherapy, it generally has a slow duration of action( weeks) and thus has not been generallly useful for control of the hormone excess state, especially acutely. Recently, much attention has been directed at establishing newer treament approaches directed at the growth of the NETs with a number of studies involving large groups of patients as well as placebo controlled, randomized, double-blind studies demonstrating the ability to extent progression-free survival with the use of somatostatin analogue [9, 10], the mTor inhibitor-everolimus[11, 12], and tyrosine kinase inhibitor, sunitinib[13], as well as the usefulness of peptide-directed radiotherapy(PRRT) with 177Lu-labeled somatostatin analogues[••14, 15, 16]. Each of these antitumor treatments, as well as other antitumor treatment approaches directed at the growth and malignant nature of the NET such as the role of aggressive surgery[17, 18, •19] or liver-directed therapies(embolization, chemoembolization, radiofrequency ablation, radioembolization)[17, 20, 21, •22] have been well covered in recent reviews. Furthermore, recent overall guidelines for emphasizing the management NET per se of various gastrointestinal NETs including GI-NETs(carcinoids) in different GI locations[1, ••8,, 23, •24, 25, 26] as well as pNETs[1, ••8, 23, 25, 27, 28] have been published.
In these recent reviews and consensus statements, what has not been specifically covered, is the recent changes including advances and controversies, in the management of the hormone hypersecretory states. These are not specifically covered because in many cases the antitumor management of the NET itself may not specifically control the hormone-excess state, for example, in the patient who is not surgically cured or in a patient with s advanced disease present from the beginning, which occurs in 30–50% of most functional pNET(F-pNETs)(except insulinomas) and in up to 50–70% of patients with GI-NETs in some locations[2].
This article will concentrate on recent advances as well as controversies in the management of the functional hormone-excess states and will only deal with advances in the management of the NET per se(antitumor treatment, etc) when it impacts on the management of the hormone-excess state. It will concentrate primarily on changes within the last 5 years. Unfortunately, for a number of reasons a systematic analysis or critical analysis of the data available on controlling the hormone excess states is limited. Most of the reports on efficacy of the advances in control of the hormone excess state are reported in case-series, case-reports, or retrospective studies. In addition, symptom control is frequently not the primary endpoint, and the studies are usually single centered. Nevertheless, the review of this data provides some important insights in promising therapeutic approaches, which should be examined in the future by larger, controlled studies.
Before the specific advances/controversies are considered it is important to understand a number of background issues involved in the management of these syndromes
2. Functional neuroendocrine tumor syndromes(F-NETS)
2.1. F-NETS: General
F-NET syndromes can be classified into those generally associated with pNETs and those generally associated with GI-NETs(carcinoids)[29]. Whereas, a number of the pNET syndromes are caused almost entirely by NETs in the pancreas(insulinoma, glucagonoma), other syndromes that are classified as pNET syndromes, in fact, can occur in NETs in extrapancreatic sites. Examples of this include duodenal gastrinomas causing Zollinger-Ellison syndrome(ZES), duodenal somatoatinomas(SSomas) associated rarely with the somatostatinoma syndrome, VIPomas occurring in neural tissues(especially in children), and GRFomas occurring in the small intestine[1, 28, 29]. Similarly, while carcinoid syndrome is characteristically usually seen in patients with liver metastases from GI-NETs from the jejunum/ileum, it can also occur with GI-NETs(carcinoids) from other GI locations, as well as rarely due to pNETs secreting serotonin[1, 29].
There are ten well-established pNET syndromes of which 9 are associated with a functional syndrome(F-pNETs) and one is not associated with a specific clinical syndrome(nonfunctional-pNET, NF-pNET). NF-pNETs, in the strict sense are not nonfunctional, in that they usually secretes a number of different products (CgA, neuron specific enolase, pancreatic polypeptide, ghrelin, neurotensin, etc.), however, these do not result in a specific clinical syndrome [1, 2, 30]. The nine F-pNETs include gastrinomas releasing gastrin to cause ZES; insulinomas; VIPomas secreting vasoactive intestine peptide; glucagonomas; SSomas; GRFomas secreting growth hormone-releasing factor; ACTHomas; pNETs secreting serotonin causing carcinoid syndrome and pNETs causing hypercalcemia by secreting PTH-RP[1, 2]. In addition to these well-established pNET syndromes, there are number of other very rare pNETs with functional syndromes(1–5 cases) which include pNETs secreting luteinizing hormone, renin, GLP-1, IGF-2, erythropoietin, enteroglucagon and cholecystokinin[1, 31], which will not be further discussed because there are too few cases studied.
2.2. F-NETS: Why treatment of hormone-excess state needed throughout care of many F-Net patients
Numerous older studies demonstrate the functional hormone-excess state needs to be controlled at all times in these patients or there is a marked increase in both morbidity and mortality[2, 27, 32–34]. This includes both acutely when the patient is initially seen as in the pre-operative period, immediate postoperative period and during long-term follow-up[2, 32, •33].
Almost all of the patients with F-pNETs, and the majority of patients with carcinoid syndrome due to a GI-NET(carcinoid), present with symptoms due to the hormone-excess state[1, 2, 35]. Therefore, in addition to suspecting the diagnosis and establishing the diagnosis, one of earliest steps in the management of these patients is to control the hormone-excess state. This need exists throughout the management of most F-NET patients because it also has to be done during any surgical operation, as well as during the follow-up if they are not cured. The need to treat the hormone-excess state is a life-long problem in >60% of all F-NET patients because cure is possible in only the minority of patients. Cure is possible in 60% of patients with gastrinomas, less than 50% of patients with all the other F-pNETS(except insulinomas which can be surgical cured in >90%), and in <5% of patients with carcinoid syndrome due to its most frequent cause(i.e. a jejunal-ileal GI-NET(carcinoid)[6, ••8, 27, 36, 37].
In the following sections the newer modalities for treating the hormone-excess state will be briefly discussed followed in a separate section by their specific use in the various F-NET syndromes. In this latter part existing therapies of the hormone-excess state will be briefly covered also. This will be followed by a discussion of the controversies/unresolved issues.
3. Summary of recent advances/controversies in management of hormone-excess states of functional NETs(Tables 1 and 2)
Table 1.
Advances in management of hormone-excess states of functional gastrointestinal NETs (F-NETs)
In refractory cases, control of symptoms of F-NET hormone-excess state by:
|
Table 2.
Controversies/unresolved issues in current management of hormone-excess states of functional gastrointestinal NETs(F-NETs)
In refractory cases, control of symptoms of F-NET hormone-excess state:
|
3.1. Use of mTor inhibitors to control symptoms of F-NETs hormone-excess states in refractory patients
The mTor inhibitor, everolimus is now approved in the United States for its antigrowth effects on both pNETs and GI-NETs(carcinoids), as well as for pulmonary NETs, in patients with advanced progressive disease. Two placebo-controlled, prospective, randomized phase 3 studies supported this result with everolimus significantly prolonging progression-free survival in each of these patient groups[11, 12].
While its principal use in patients with NETs is for its antigrowth effects, recent studies involving small series[•38, •39, 40, 41] as well as case reports[42–46] provide support for the conclusion that everolimus also is effective at controlling the hormone-excess state in patients with refractory F-NET syndromes, including both F-pNETs and with carcinoid syndrome due to GI-NETs(carcinoids). One study shows a similar benefit with the mTor inhibitor, rapamycin[47]. |This will be discussed in more detail on the specific syndromes in the following section.
3.2. Use of the tyrosine kinase inhibitor, sunitinib, to control symptoms of F-NETs hormones-excess states in refractory patients
The tyrosine kinase inhibitor, sunitinib, inhibits the tyrosine kinase activity of PDGFRs, VEGFR-1, VEGFR-2, c-KIT and FLT3[48]. Sunitinib is approved in the United States for its antigrowth effects of pNETs in patients with progressive advanced disease. This approval was based on the results of a double-blind, prospective, placebo controlled phase 3 study which demonstrated sunitinib significantly extended progression-free survival in these patients as well and increased overall survival[13].
Sunitinib’s principal use is in patients with pNETs for its antigrowth effect in patients with advanced disease, however small series[49] and case reports[•50, 51], support the conclusion that it is effective at controlling hormone-excess states in patients with refractory, symptomatic pNET syndromes.
3.3. Use of Peptide radio-receptor therapy(PRRT) with 177Lu-labeled somatostatin analogues, to control symptoms of F-NETs hormone-excess states in refractory patients
Gastrointestinal NETs including both pNETs and GI-NETs(carcinoids) resemble each other and NETs in other locations, by overexpressing somatostatin receptors(sst2, 5) in 80–100% of well-differentiated tumors[17, 52, 53]. This overexpression of somatostatin receptors has been successfully used to image both the primary tumor and the tumor extent, using primarily 111In-labeled-somatostatin analogues or 68Ga labeled somatostatin analogues[••8, 17, 52, 53]. At present, the use of 68Ga-labeled somatostatin analogues with positron emission tomography and computed tomographic scanning(PET/CT) is the most sensitive imaging modality available for these tumors and is approved for this use both in the United States and Europe[••8, 52, 53].
Numerous studies have extended the observation of overexpression of somatostatin receptors by NETs to develop radiolabeled (111In, 177Lu, 90Y) somatostatin analogues to deliver cytotoxic doses to malignant NETs[15, 16, 52]. These studies provided strong support that peptide radioreceptor therapy [PRRT] using radiolabeled somatostatin analogues had promise as an effective cytotoxic therapy for patients with advance unresectible NETs[15, 16, 52]. Recently, a double-blind, prospective, randomized trial has supported the efficacy and safety of this approach in patients with advanced inoperable, progressive midgut GI-NETs(carcinoids)[••14]. In this trial of 230 patients with grade ½ midgut carcinoids, a marked increased in progressive-free survival (p<0.0001) was seen with PRRT treatment with a 177Lu-labeled-somatostatin-analog, with an acceptable safety profile(low rate of grade 3/4 toxcity) and with a suggestion of an improved survival, although the analysis is not yet complete[••14, ].
At present PRRT with 177Lu labeled somatosatin analogues is not approved by the FDA in the United States for its antitumor treatment in patients with advanced unresectible NETs, but it is widely used on a compassionate basis, especially in Europe[15, 16, 55]. Recent studies involving small series[•56, 57] as well as case reports[58–60, •61, 62. •63, 64–66] provide support for the conclusion that PRRT with radiolabeled somatostatin analogues also is effective at controlling hormone-excess states in patients with refractory F-NET syndromes, including both F-pNETs and with carcinoid syndrome due to GI-NETs(carcinoids.
3.4. Use of liver-directed therapies[TACE, TAE, Radioembolization (SIRTs), RFA] to reduce tumor mass/hormone release and increase response to medical therapy to control symptoms of F-NETs hormone-excess states in refractory patients
Approximately 30–50% of patients with different F-PNETs syndromes (except insulinoma=where 90% benign) and 98–100% of patients with carcinoid syndrome due to a malignant GI-NET(carcinoid), have liver metastases at presentation[2, ••8, ••24, 27, 28, 67]. These patients are rarely cured surgically and thus are candidates for various forms of liver-directed therapies, particularly when the primary tumor is resected[1, 20, 21, •22, 68, •69, 70]. Liver-directed therapies include trans-arterial embolization (TAE), trans-arterial chemo-embolization(TACE), radioembolization or selective internal radiation therapy(SIRT), as well as radio-frequency ablation(RFA) and cytoreductive surgery. The later two will be considered in separate sections below.
TAE and TACE can be safely performed because NET metastases in the liver are highly vascular and derive their blood supply primarily from tributaries of the hepatic artery, whereas normal liver parenchyma is supplied primarily by the portal vein[17, 20, 21, 22, 68, 71]. Consequently, interruption of the tributaries of the hepatic artery affects the tumor deposits much more than the normal liver. This interruption can be performed at surgery, but is usually performed percutaneously and can be either occlusion only (TAE) or with the co-administration of chemotherapeutic agents(TACE) (usually doxorubicin, mitomycin C, streptozotocin, fluorouracil, cisplatin)[17, 20, 21, •22, 68, 71].
TAE or TACE are usually performed for palliative treatment of liver-predominant disease which is not surgically resectable to reduce the hepatic tumor mass and 25–85% of patients have an objective tumor response, with a mean duration of response of 6–45 months[17, 20, 21, •22, 68, 71]. This approach has been particularly considered in patients with hepatic symptoms or refractory malignant F-NET syndromes[17, 20, 21, •22, 68, 71]. In general TACE/TAE result in a symptomatic response in 50–100% of patients and numerous series as well as case reports have documented their control of symptoms in patients with both carcinoid syndrome and F-pNET syndromes[17, 20, 21, •22, 68, 71, 72, •73, 74, 75, •76, 77, •78].
Radio-frequency ablation or other forms of ablation of metastatic NETs can also be performed at the time of surgery or percutaneously[17]. Of these different ablative surgeries, RFA is the most frequently performed, often in combination with surgery, especially to remove isolated metastases in other hepatic locations[17, 20, 21, •22, ••24, •69, 79–81]. Numerous criteria for when this should be considered have been proposed depending on the number of lesions, size, and proximity to vital structures[17, 20, 21, •22, ••24, •69, 79–82]. Besides the use of RFA as an anti-tumor treatment, a number of studies have reported enhanced symptomatic control of functional NETs after its use[17, 20, 21, •22-, ••24, •69, 79–83].
Radioembolization or SIRTs primarily involves primarily the use of 90Yttrium(90Y) microspheres (either 90Y resin microspheres[SIR-spheres) or 90Y glass microspheres(Therespheres))(17, 20, 21, •22, •69, 70). These are administered intra-arterially primarily as an anti-tumor treatment, however a number of studies have reported enhanced symptomatic control of F-NETs after their use17, 20, 21, •22, •69, 70).
These approaches will be discussed in more detail on the specific syndromes in a latter section.
3.5. Use of percutaneous/endoscopic ablative therapies(ethanol, etc.) for primary or metastases(F-pNETs) to reduce tumor mass/hormone release and increase response to medical therapy or to control symptoms of F-NETs hormone-excess states in refractory patients
As pointed out in #3.4. above a proportion of patients with F-NET syndromes have extensive disease and are not surgical candidates. In addition a portion of patients, primarily with F-pNETS have increasingly refractory symptomatic disease with only a primary or adjacent localized disease, who are not candidates for surgery, because of accompanying medical problems, risks from age, etc. Recent studies, including small series as well as case reports, demonstrate, particularly the use of ethanol injections, administered percutaneously or by endoscopic ultrasound guidance, can be highly successful in this situation in controlling the hormone-excess-state in these patients and greatly enhancing the ability to control the hormonal symptoms[84–88, •89, 90–92, •93]. This approach will be discussed in more detail on the specific syndromes in a latter section.
3.6. Use of a supra-maximal doses of conventional therapies(carcinoid syndrome, F-pNETs) to increase response to medical therapy and to control symptoms of F-NETs hormone-excess states in refractory patients
Somatostatin analogues are the initial treatment to control the hormone-excess state in most F-NET syndromes, including the carcinoid syndrome, as well as number of F-pNET syndromes(GRFomas, VIPomas, SSomas, glucagonomas, and the very rare F-pNET syndromes)[1, 2, ••8]. However, in insulinomas, gastrinomas and ACTHomas, other drugs are usually initially used, as will be discussed in the separate sections on these syndromes below.
A proportions of patients with these F-NET syndromes do not have successfully control of the hormone-excess state with conventional doses of these drugs initially (20–30% with carcinoid syndrome). Furthermore, a proportion of patients with time, especially patients with F-NETs with nongastrinoma, insulinoma syndromes, treated with somatostatin analogues, develop increasing resistance with time, when used at conventional doses[94–98]. Numerous studies, especially in patients with the carcinoid syndrome or VIPomas, have reported that increasing the doses of the somatostatin analogues by either using a higher dose, or using a conventional dose, at increased frequency than is conventionally used (more frequent than 1x/month) can help control the hormone-excess symptoms[94, 95, •96, 97–102].
3.7. Use of cytoreductives surgery to reduce tumor mass/hormone release and increase response to medical therapy or to control symptoms of F-NETs hormone-excess states in refractory patients
Unfortunately, only 5–15% of patients with F-NET syndromes with metastatic disease to the liver have potentially, completely resectable disease with the possibility of surgical cure, because of the presence of diffuse, bilobar hepatic metastases[••8, 17, •19, ••24, 28].
However, a number of authorizes recommend that cytoreductive surgery or debunking surgery be considered in patients in which 70–90% of the visible disease is resectable[••8, 17, •19, ••24, 28, 80]. This is recommended is to primarily control refractory hormone-excess symptoms, to possibly facilitate medical control of the hormone-excess state, or to control local symptoms due to the tumor bulk, such as pain[••8, 17, •19, ••24, 28, 80, 100, 103]. Even though numerous studies report this also contributes to increased survival, there are no controlled studies, which establish this[17, •19, ••24, 28, •104]. Despite lack of controlled studies, the ENETs guidelines recommend surgery for curative intent whenever possible [9].
Numerous studies, as well as case reports, have demonstrated that cytoreductive surgery can facilitate the management of the hormone-excess state symptoms and therefore should be considered in any patients with increasing refractory symptoms[••8, 17, •19, ••24, 28, 103]. However, it is becoming increasingly confusing for many practitioners the exact role of cytoreductive surgery, with the increasingly effective numbers of medical therapies available, as well as PRRT, and increased numbers of liver directed therapies.
Liver transplantation is the ultimate hepatic debulking procedure and is occasionally used in patients with refractory hormone excess states with F-NETs confined to the liver[17, 18, 19, 24, 103].
3.8. Increased use of 131I-MIBG to reduce tumor mass/hormone release and increase response to medical therapy or to control symptoms of F-NETs hormone-excess states in refractory patients
Meta-iodobenzylguanidine(MIBG) is a biogenic amine precursor analogue, which is taken up by the norepinephrine transporter and stored in primarily the cytoplasm of number of neurosecretory cells[105, 105, 106].
MIBG radiolabeled with 131I(131I-MIBG) has been used for more than 20 years in the management of chromaffin tumors(neuroblastomas, pheochromocytomas, paragangliomas), but less frequently in NET tumors[105–110]. Up to 70% of GI-NETs(carcinoids) and 9–25% of pNETs demonstrate positive uptake with 131I-MIBG and a number of studies have reported palliative effects with treatment of palliative with advanced carcinoid tumors[105–110]. These palliative effects included not only effects on tumor growth/size but also symptomatic improvement in patients with carcinoid syndrome[105–109]. 131I-MIBG treatments have also been used in occasional patients with refractory F-pNET syndromes[•73, 110–112]. Therapy with 131I-MIBG is gernally well tolerated but can include pancytopenia, thrombocytopenia, nausea and emesis.
3.9. Use of a new specific syndrome therapies to reduce tumor mass/hormone release and/or increase response to medical therapy or to control symptoms of F-NETs hormone-excess states in refractory patients
3.9.1 Use of Telostristat, a tryptophan hydroxylase inhibitor, to inhibit serotonin synthesis and control symptoms of the carcinoid syndrome
The carcinoid syndrome is characterized by the presence of diarrhea, bronchospasm, episodic flushing of the skin and in its later stages, by the development of valvular heat disease[2]. It has long been known that serotonin is one of the key mediators of the carcinoid syndrome, and is especially involved in the development of carcinoid heart disease, as well as the diarrhea, which is seen in 60–100% of patients with the carcinoid syndrome[2]. Another important feature is the requirement that the serotonin enters the general circulation and not be inactivated by the liver, which occurs in most cases(>95%) by the presence of liver metastases[2].
Recently a tryptophan hydroxylase inhibitor, telotristat etiprate, has been developed, which, by inhibiting this enzyme, blocks the conversion of tryptophan to serotonin[2, 113, 114, •115]. Telostristat is an orally active, small molecule which was designed not to cross the blood brain barrier and only act peripherally, thus avoiding the CNS side-effects that were seen with older serotonin inhibitors such as parachlorophenylanine, which limited their use[2, 113, 114, •115].
In a number of recent studies[1,••14, ••116, •117] including a Phase 3 prospective randomized trial(TELESTAR)[•115, ••116], telotristat has been shown to decrease the occurrence of diarrhea, as well as markedly decrease or normalize urinary serotonin/5-HIAA levels in patients with the carcinoid syndrome. The telotristat decrease in the diarrhea had a large impact on the emotional, social and physical aspects of the patient’s lives[113, •117, 118]. In these studies telotristat was well tolerated and adverse side effects were mild and generally of a GI nature[1,••14, •115, •117]. One of the key questions is whether the use of telotristat will alter the course of the valvular heart disease. The only information available so far is a recent abstract based on two patients in the TELESTAR study with carcinoid heart disease reported who demonstrated no further progression in the valvular disease occurred while taking telotristat and the authors proposed the use of the drug may prevent the need for valve surgery in some patients[119].
3.9.2. Use of Pasireotide, a broad spectrum sst receptor agonist, to reduce tumor mass/hormone release and/or increase response to medical therapy, to control symptoms of F-NETs hormone-excess states in refractory patients
Well-differentiated gastrointestinal NETs overexpress at least one of the 5 subclasses of somatostatin receptors(sst1–5) in almost all cases, with the most frequent being sst2(80–100%) and sst5(50–60%)[2]. Somatostatin has high affinity for all 5 sst subtypes, whereas the related synthetic analogues, octreotide, octreotate(used for imaging) and Lanreotide interact with very high affinity only with sst2 and intermediate affinity with sst3/sst5[2, •120].
As stated in the previous section, in a proportion of gastrointestinal F-NETs, octreotide/Lanreotide do not initially control the hormone-excess syndromes and furthermore, with time, in some patients that initially responded, they develop increasing resistance. Many of these NETs express the other classes of somatostatin receptors(sst1, 3, 4) which interact only with low affinity with these drugs[121, •122, •123], raising the possibility that a broad acting somatostatin analog might be more effective[121, •122, •123, 124].
Pasireotide interacts with high affinity with all SST subtypes except sst3 and recent studies demonstrate it may have increased utility in some of the gastrointestinal NETs and F-NET syndromes, in that it controlled the hormone-excess state, in some cases in patients with resistance to octreotide/lanreotide[121, •122, •123, 124, 125]. In an animal model of MEN1 patients with insulinomas, pasireotide treatment was able to control the hormone-excess state as well as the insulinoma growth[126]. While a once monthly injection of Pasireotide-LAR has been generally well-tolerated[127], hyperglycemia occurs at a higher frequency that seen with octreotide-LAR(28 vs 5%), as well as fatigue(11 vs 3.5%) and nausea(9.4 vs 0%)[124]. In two other studies hyperglycemia was seen in 16% and 79% of patients[121, •122]. In a study of normal volunteers[128], the pasireotide-associated hyperglycemia was related to a decrease in insulin secretion and incretin hormone response, without changes in hepatic/peripheral insulin sensitivity[128].
3.9.3. Use of newer agents to treat Cushing’s syndrome due to pancreatic ACTHomas or other causes of ectopic Cushing’s syndrome, to control symptoms
In various studies 4–16% of ectopic Cushing’s syndrome (EAS) is due to pancreatic ACTHomas. Although uncommon in pNETs[2, 129], Cushing’s syndrome can be the only sign of a F-pNET and it occurs in 4–5% of patients with sporadic ZES in which its development is associated with a poor prognosis, because the tumors are usually aggressive[2, 30, 130].
Until relatively recently the treatment of the EAS involved the use of bilateral adrenalectomy(preferably after period of control of the EAS), and medical treatment of the hypercortisolemia using mitotane, metapyrone and ketoconazole[131, •132, 133, 134]. Recently other classes of drugs have been used and reported to be helpful including glucocorticoid receptor antagonists(mifepristone[•135], etc.); adrenal specific therapy(etomidate, osilodrostat and fluconazole=11B-hydroxylase inhibitors); somatostatin analogues including pasireotide; and dopamine receptor agonists(cabergoline, etc.)[ 131, •132, 133, 134]. In addition PRRT has been reported to be helpful in controlling the hypercortisolism with EAS and will be discussed under the specific syndromes in a later section.
4. Summary of recent advances in management of specific hormone-excess states of functional NETs(Tables 1)
4.1. General
For each of the F-NETs the hormone-excess state needs to be controlled as quickly as possible while the diagnosis, extent of manifestations of the hormone-excess state established (carcinoid heart disease, etc.) and treatment strategy are being formulated. The latter includes establishing the location of the primary tumor and extent of disease; with F-pNETs, the possible presence of an inherited syndrome such as MEN1[2, 136, 137]; whether surgical resection is possible, and the treatment of advanced disease, if present[2, ••8, 17, 27, 28, 136, 138, 139].
Control of the hormone state is needed during tumor localization, which is particularly important to formulating management steps. The tumor localization can be time-consuming and can be difficult with F-NET syndromes. The primary tumors can be small and therefore not localized by conventional cross-sectional imaging(CT, ultrasound, MRI)[2, ••8, 28, 140, 141] and frequently requires specified studies such as somatostatin receptor scintigraphy(SRS)(using 68Ga or 111In-labeled somatostatin analogues[••8, 52, 142], endoscopic ultrasound[2, ••8, 28, 142], and rarely, specific hormonal gradient sampling[2, 28, 143, 144]).
4.2. Carcinoid syndrome
4.2.1. Current standard treatment of carcinoid syndrome
Long acting somatostatin analogues(Octreotide-LAR, Lanreotide-autogel) supplemented during break-through with subcutaneous octreotide, are the standard therapies for controlling the principal carcinoid syndrome symptoms of diarrhea, flushing, and asthma[2, •96, 98, •120, 145, •146]. With time, refractory cases may develop and therefore, there is a need for new treatments[2, •96, 98, •120, 145, •146]. Treatment of the carcinoid heart disease involves standard treatment for heart failure/arrhymias and in advanced cases, heart valve replacement[147]. Whether long-term somatostatin treatment prevents the development or progression of the carcinoid heart disease is not established[147].
4.2.2. Newer treatments of carcinoid syndrome(Table 1)
Almost all of the new therapies for F-NETs listed in Table 1 have been used successfully patients with carcinoid syndrome, some with refractory disease, although in many cases only small numbers of patients have been treated.
Both studies and case reports have reported the successful use in patients with carcinoid syndrome of the control of the symptoms of the hormone-excess state by using: everolimus[•38]; PRRT[•56, 57, 66, 95, •96, •148]; liver-directed therapies including TACE/TAE[70, 72, •73, 77, 95, 98, 149]; radioembolization[70, 150, 151]; RFA[80, 81, 83, 95, 152]; by increasing the dosage of octreotide/Lanreotide in refractory cases[94, 97, 153–155]; use of cytoreductive surgery[23, 95, 156–160]; use of 131I-MIBG[95, 105, 106, 108, 110, •161, 162–164]; with the use of the tryptophan hydroxylase inhibitor, Telotristat[108, 113, 114, ••116, •117, 118, 119] and with pasireotide in octreotide resistant cases[•122].
In one series[•56] of 35 patients with refractory carcinoid syndrome symptoms, PRRT with 177Lu-DOTA-Octreotate resulted in 63% of patients having a significant improvement in flushing and/or diarrhea. The TELESTAR study[•115, ••116] is ongoing and is a phase 3 randomized trial of the efficacy/safety of the 11B-hydroxylase inhibitor required for serotonin synthesis, in patients with carcinoid syndrome, Telotristat resulted in a 29%(250 mg) and 35%(500 mg) decrease in bowel movement frequency and was well tolerated[•115, ••116], suggesting it may be a valuable addition to control some aspects of the carcinoid syndrome. In a Phase 2, multicenter study[•122] involving 45 patients with carcinoid syndrome with refractory symptoms to octreotide-LAR, pasireotide controlled the diarrhea and/or flushing in 27%.
The carcinoid crisis is an emergency which can occur in patients with carcinoid syndrome [2, 24}. It is usually precipitated by procedures such as surgery and is characterized by extreme changes in blood pressure, flushing, diarrhea, CNS symptoms and cardiac arrhythmias[2]. High doses of somatostatin anlogues are used prophylactically prior to these procedures, as well as intravenously during the procedure, but are not always successful [2]. Whether telotristat[••116, •117, 118] will prevent the development of carcinoid crises is unclear at present.
4.3. Insulinoma
4.3.1. Current standard treatment of Insulinoma
In >90% of patients with insulinoma the treatment is curative surgical resection[2, • ••8, 28, 136]. This is the exception with the F-pNETs and occurs because insulinomas are usually solitary, almost all intrapancreatic in location, this is the only pNET syndrome in which almost of the tumors are benign, and in <3% of all patient, s insulinomas occur in patients with MEN1 in which they can be recurrent and multiple[2, ••8, 28, 136]. Therefore, control of the hormone-excess state in patients with insulinomas is usually only needed during the pre-operative period and in the minority of patients with malignant insulinomas. Currently, the primary treatment is frequent small feedings and the use of diazoxide, which can control symptoms in 50–60%. Somatostatin analogues are also used and are effective in 50%[99, 165, 166, •167] however, in some patients they can exacerbate symptoms, by inhibiting counter-regulatory mechanisms such as glucagon/growth-growth hormone release[2, 168, 169].
4.3.2. Newer treatments of insulinoma hormone-excess state(Table 1)
Many of the new therapies for F-NETs listed in Table 1 have been used successfully in patients with insulinoma, some with refractory disease, although in many cases only small numbers of patients have been treated.
Both studies and case reports have reported the successful use in patients with insulinomas, particularly in patients with malignant insulinoma which comprise<10% of all cases, but in whom the hormone-excess state can be difficult to control[•167]. Included in the recent literature are reports of the control of the symptoms of the insulinoma hormone-excess state by using: everolimus[•39, 40–46, 170, 171]; tyrosine kinase inhibitor sunitinib[•50]; PRRT[44, 55, 58–60, •61, •76, 170, 172]; liver directed therapies including TACE/TAE[44, 70, 72, •73, 74.75, •76, 172–174]; radioembolization[44, 70, 172]; RFA and use of ablative treatment(ethanol, microwave of primary/metastases[81, 83–87, •89, 90, 92, •93, 175, •176, 177, 178]; by increasing the dosage of octreotide/Lanreotide in refractory cases[153, 155]; use of cytoreductive surgery[157, 158, 160, 172, 174]; use of 131I-MIBG[•111] and with pasireotide in octreotide resistant cases[125].
In one report[55] involving 5 patients with metastatic insulinomas, with poorly controlled hypoglycemia on standard therapies, treatment with 177Lu-DOTA-octreotate or •111In-octreotide resulted in stable disease for a mean follow-up of 27 months with no hypoglycemic episodes. In three reports of 8[•89], 4[90] and 4[92] patients with insulinomas which were not operable in a number of cases, ethanol injection of the primary insulinoma controlled the symptoms of the insulinoma in all cases.
4.4. Zollinger-Ellison syndrome
4.4.1. Current standard treatment of Zollinger-Ellison syndrome(ZES)
At present approximately 25–40% of all patients with ZES are cured long-term[2, 6, ••8, 27], thus the majority require life-long treatment of the gastric acid hypersecretion. This occurs because 70–85% of patients with sporadic ZES (i.e.no MEN1) have small, duodenal tumors which are frequently missed at surgery, 25–35% have non-resectable liver metastases at diagnosis and 25% have MEN1 with multiple, small gastrinomas frequently with lymph node metastases[2, 6, ••8, 27, 37, 141, 179].
The medical control of the gastric acid hypersecretory state is highly effective with proton pump inhibitors(PPIs) and histamine H2 receptor antagonists are also effective in most patients if high, frequent dosing is used[6, 27, •33]. Somatostatin analogues are also effective but rarely used because they require parental administration compared to the orally active PPIs and histamine H2 receptor antagonists[27, •33]. Because tachypylaxis is not a problem with PPIs, these drugs remain effective and if sufficient drug is given, except for low vitamin B12 levels in some patients, there are very rarely any side-effects limiting their use, and acid secretion can be controlled in all patients with ZES long-term.[6, 27, •33, 180]. Therefore, these newer therapies to control the hormone-excess state have been rarely used in ZES patients.
4.4.2. Newer treatments to control Zollinger-Ellison syndrome(ZES) symptoms(Table 1)
Only a few of the new therapies for F-NETs listed in Table 1 have been used successfully patients with ZES, some with refractory disease, and only small numbers of patients have been treated.
Both studies and case reports have reported the successful use in patients with ZES of the control of the symptoms of the hormone-excess state by using: PRRT[65]; liver directed therapies including TACE/TAE[70, 173, 174, 181]; radioembolization[70], RFA[81, 83, •176, 182]; by increasing the dosage of octreotide/Lanreotide in refractory cases[155, 183]; use of cytoreductive surgery[156, 160, 174]; use of 131I-MIBG[110, 164] and with pasireotide in octreotide resistant cases[•122].
4.5. VIPoma syndrome
4.5.1. Current standard treatment of VIPoma symptoms
Patients with VIPoma syndrome are uncommonly cured(<20–30%) because most adults present with metastatic disease, therefore life-long treatment of the hormone-excess state is usually required[2, ••8, 28, 102]. The main stay of treatment is the use of somatostatin analogues(octreotide/lanreotide), which control the large-volume diarrhea in 70–90%, and although some patients become refractory, in most, with dose adjustments, these drugs continue to have some efficacy[2, ••8, 28, 101, 102, •120]
4.5.2. Newer treatments of VIPoma symptoms(Table 1)
A number of the new therapies for F-NETs listed in Table 1 have been used successfully patients with VIPomas, many with refractory disease, although in many reports only small numbers of patients have been treated.
Both studies and case reports have reported the successful use in patients with VIPomas of the control of the symptoms of the hormone-excess state by using: tyrosine kinase inhibitor sunitinib[47, 49]; liver directed therapies including TACE/TAE[70, 100, 173, 174, 184, 185]; radioembolization[70, 150], RFA [81, 83, •186, 187, 188]; by increasing the dosage of octreotide/Lanreotide in refractory cases[185] and the use of cytoreductive surgery[100, 102, 134, 158, 160, 174, •186, 189].
4.6. Other less frequent F-pNET syndromes(glucagonomas, pancreatic ACTHomas, etc)
4.6.1. Current standard treatment of other F-pNET-syndrome symptoms
Conventional therapies include the use of long-acting somatostatin analogues and various therapies directed at Cushing’s syndrome(see.section.3.9.3 above)
4.6.2. Newer treatments of other F-pNET symptoms(Table 1)
A number of the new therapies for F-NETs listed in Table 1 have been used successfully patients with other, less frequent F-NETs, some with refractory disease, although in many cases only small numbers of patients have been treated.
Both studies and case reports have reported the successful use in patients with a number of the other F-pNET syndromes for of the control of the symptoms of the hormone-excess state by using: PRRT [glucagonoma[•63], reninoma[64], ACTHoma[62, 65]] ; liver-directed therapies including TACE/TAE [70] [glucagonoma[174], ACTHoma[181], SIADH[•78]]; radioembolization[70, 174][glucagonoma, SSoma[150]]; RFA [glucagonoma[80, 81], ACTHoma[80], SSoma[83], PTHrPoma[81]]; by increasing the dosage of octreotide/lanreotide in refractory cases [glucagonoma[183]]; use of cytoreductive surgery [glucagonoma[•63, 156–158, 160, 174], ACTHoma[62, 190], PTHrPoma[160]] ; and use of 131I-MIBG [SSoma[107]].
5. Controversies/unresolved issues in current management of hormone-excess states of functional gastrointestinal NETs(F-NETs)(Table 2)
5.1. General
Despite the numerous advances in treatment of the functional states of F-NETs discussed above, as well as advances in treatments directed at the tumor itself, a number of areas of controversy and/or unresolved issues remain in the management of the hormone-excess states which are summarized in Table 2. These will be briefly discussed.
5.2. Controversies/unresolved issues pertain to most F-NETs
For the new therapies there are no studies, which have established the order with which they should be applied, whether drug combinations are more helpful if one fails, or even if therapy with one new medical treatment such as everolimus or sunitinib fails, is the other efficacious for controlling the hormone-excess state.
Although at present in most cases different medical therapies including the newer drugs, such as sunitinib or everolimus, are used before PRRT or liver-directed therapies, especially with the excellent results with PRRT with some F-NET syndromes, it may be appropriate to use it earlier. Furthermore, in patients that are requiring higher doses with medical management, PRRT or liver-directed therapies may be used earlier to facilitate control of the hormone-excess state.
The present place of cytoreductive surgery is really not well defined because its use is often not for the hormone-excess-state but for its effect on tumor itself. It is unclear in most studies how often it is required in patients with F-NETs for symptom control of the hormone-excess state, how effective it is for this, and whether liver-directed therapies or PRRT would not be just as effective.
5.3. Controversies/unresolved issues pertain to a specific F-NET syndrome
While the TELESTAR study established that telotristat, by inhibiting the synthesis of serotonin[•115, ••116], markedly ameliorated the diarrhea in patients with carcinoid syndrome, its effect on a number of other important manifestations of carcinoid syndrome will be very important, and are at present unclear. Part of the uncertainty is due to the release of numerous other biologically active amines/peptides by the carcinoid tumor, which are reported to contribute to the symptoms of the hormone-excess state[2]. These include various tachykinin(substance P, neurokinin A, neuropeptide K), prostaglandins, bradykinin, and kallikreins[2, 191, 192]. Whether the flushing and asthmatic symptoms in patients also can be ameliorated by telotristat, as well as the development and/or reversal of the carcinoid heart disease, is unclear. Also the pathogenesis of the fibrosis associated with the carcinoid syndrome is not clear[191], and it is unclear whether telotristat will reverse this or stops its progression, both in the heart and in the mesentery. The availability of telotristat should help to answer these questions and define the role of serotonin excess in mediating these manifestations of the carcinoid syndrome.
The role of the long-term use of pasireotide in F-pNETs at present is unclear. There is only one study[•122] on its effectiveness in small number of patients with carcinoid syndrome whose symptoms were octreotide/Lanreotide resistant. Because of the small size of this study, it remains unclear in a larger group of patients with different types of F-NET syndromes who develop resistance for on octreotide/Lanreotide, in what percentage will pasireotide be effective. It also is unclear if initially effective, whether it will remain effective, or similar to octreotide/Lanreotide, in many cases increasing resistance will develop. There is also the question of the long-term complications of the use of pasireotide. In a number of studies hyperglycemia developed in 16% to 79% of patients in different studies[121, •122]. Whether this is going to be a major problem and limit its usefulness in these patients, especially if combined with previous pancreatic resections, is unclear.
The majority of patients with pancreatic ACTHomas have malignant disease[30, 130, 193], in most cases with diffuse liver metastases, and the Cushing’s syndrome can be rapidly developing, fulminant, difficult to control and has been shown to be an independent variable for poor prognosis[30, 130, 193]. Whether medical therapy[194], even with the newer drugs, will be successful in the majority of these patients is unclear, as is the role of PRRT, liver-directed therapies or cytoreductive surgery.
6. Conclusions
With widespread use of somatostatin analogs and the development of potent gastric acid anti-secretory agents (PPIs, H2 receptor antagonists) the medical treatment of the hormone-excess states of F-NETs has become possible in most cases. However, in the case of the somatostatin analogues, resistance can develop over time, initially resistant cases exist, and the long-term treatment may not prevent or reverse all aspects of the hormone-excess state such as fibrosis in carcinoid syndrome or carcinoid heart disease. Control of the hormone-excess state can be particularly difficult in some patients with carcinoid syndrome, insulinomas and VIPomas. In this paper newer options for treatment of the hormone-excess state are reviewed with a number showing much promise. Despite these advances, a number of unanswered questions still exist as well as controversies on management of the hormone-excess states. Each of these are reviewed in depth in this paper.
7. Expert opinion
In the last few years there have been considerable attention paid to advances in anti-tumor treatments of patients with neuroendocrine tumors. This has resulted in a large number of reviews and consensus papers incorporating these advances. This is appropriate because the incidence of NETs is increasing in almost all countries, and increasingly the natural history of the NET per se is becoming the primary determinant of long-term survival. However, a proportion of these patients with F-NETs has two problems requiring management: antitumor treatment and treatment of the hormone-excess state. The antitumor treatments, for example chemotherapy, while effective against tumor growth, may not be useful to control the hormone-excess state, and therefore these two problems; controlling the hormone-excess state and the tumor growth may not be adequately dealt with by one approach.
Treatments directed at controlling malignant NET growth have received increased attention, in part because of the development of effective treatments for the hormone-excess state in patients with F-NETs. Particularly important has been medical treatment with somatostatin analogues with F-NETs, and for patients with gastrinoma, potent gastric antisecretory drugs, which control the hormone-excess state initial in most patients. The result of this increasingly effective contriol of the hormone-excess state is that, it is no longer a leading cause of early death, with the result that tumor growth is becoming the major factor for long term survival. While these treatments have been highly successful, the long-term treatment of the hormone-excess states remain a problem in many patients, because of drug-resistance developing, resistance existing at the initial treatment or because all manifestations of the hormone-excess states are not controlled. Particularly, in patients with insulinomas not controlled surgically, carcinoid syndrome or some cases of VIPomas and pancreatic ACTHomas, these later problems arise and new treatments are needed. These newer treatments for the hormone-excess states, in general, have received little attention. This paper reviews the evidence that a number of new treatments(medical, surgical, radiological) are effective in controlling the hormone-excess states and their increased use may markedly facilitate management. Because of the rarity of this some of these syndromes, there are few large perspective trials, but nevertheless, in many cases of the different new treatments, there are sufficient number of cases to support the usefulness of the different approaches. However, there are a number of unresolved issues and controversies that will likely require additional studies, which in some cases will need to be controlled studies to completely resolve the issues.
Article Highlights.
Patients with functional neuroendocrine tumors(F-NETs) have two treatment problems: treatment of the hormone-excess state and directed at tumor growth/malignancy
Recently there are many advances in antitumor therapies, which are well covered in reviews, but little analysis of advances in treatments of hormone-excess states
In contrast to antitumor studies, most advances in treatment of hormone-excess states are in small series, case series, case reports.
Numerous new treatments of the hormone-excess states are reported including: medical therapies [mTor-inhibitors (everolimus), tyrosine-kinase inhibitors (sunitinib), increases dosages of convention agents (somatostatin-analogs)]; peptide-radioreceptor therapy and increased use of 131I-MIBG; cytoreductive surgery; ablation of F-NET primaries; liver-directed therapies (embolization, chemoembolization, radioembolization, RFA); therapies directed at specific syndromes [serotonin-synthesis inhibitors(telotristat), pasireotide, new agents for Cushing’s syndrome with ACTHomas]
The evidence for the use of each approach is reviewed and the unresolved areas and controversies discussed
Abbreviations
- pancreatic ACTHoma
adrenocorticotropic hormone-secreting neuroendocrine tumor of pancreas
- CgA
chromogranin A
- CT
computed tomography
- F-NET
NET with a specific functional syndrome
- GRFoma
growth-hormone releasing factor secreting NET
- NET
neuroendocrine tumor
- NF-pNET
pNET not associated with a functional syndrome
- GI-NET
neuroendocrine tumor of the gastrointestinal tract not including pNETs
- MIBG
meta-iodobenzylguanidine
- MEN-1
Multiple Endocrine Neoplasia-type 1
- MRI
magnetic resonance imaging
- NF-PNET
nonfunctional pNET
- PPI
proton pump inhibitors
- VHL
von Hippel Lindau disease
- pNET
pancreatic neuroendocrine tumor
- PRRT
Peptide Radio-receptor therapy
- RFA
radiofrequency ablation
- TAE
trans-arterial embolization
- TACE
trans-arterial chemoembolization
- SIRT
selective internal radiation therapy
- SSoma
somatostatinoma
- sst1–5
somatostatin receptor subtypes
- VIPoma
vasoactive intestinal peptide secreting NET
- ZES
Zollinger-Ellison syndrome
Footnotes
Declaration of interest
Partial funding for this study was provided by intramural funds from NIDDK, NIH. The authors have no other finanicial relationships such as consultancies, employment related to this material, expert testimomy, honoraria, stock options, speakers bureaus or retainers.
References
- 1.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: 10.1016/j.bpg.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Jensen RT, Norton JA, Oberg K. Neuroendocrine Tumors. In: Feldman M, Friedman LS, Brandt LJ, editors. Sleisenger and Fordtran’s Gastrointestinal and Liver Diseases. Philadelphia: Elsevier Saunders; 16 A.D. pp. 501–541. [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: 10.1200/JCO.2007.15.4377. [DOI] [PubMed] [Google Scholar]
- 4.Fraenkel M, Kim MK, Faggiano A, et al. Epidemiology of gastroenteropancreatic neuroendocrine tumours. Best Pract Res Clin Gastroenterol. 2012;26:691–703. doi: 10.1016/j.bpg.2013.01.006. [DOI] [PubMed] [Google Scholar]
- 5.Ito T, Igarashi H, Nakamura K, et al. Epidemiological trends of pancreatic and gastrointestinal neuroendocrine tumors in Japan: a nationwide survey analysis. J Gastroenterol. 2015;50:58–64. doi: 10.1007/s00535-014-0934-2. [DOI] [PubMed] [Google Scholar]
- 6.Ito T, Igarashi H, Jensen RT. Zollinger-Ellison syndrome: Recent advances and controversies. Current Opinion in Gastroenterology. 2013;29:650–661. doi: 10.1097/MOG.0b013e328365efb1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fishbeyn VA, Norton JA, Benya RV, et al. Assessment and prediction of long-term cure in patients with Zollinger-Ellison syndrome: the best approach. Ann Intern Med. 1993;119:199–206. doi: 10.7326/0003-4819-119-3-199308010-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8••.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: 10.1159/000443171. Consensus paper dealing with all issues of NETs (F-NETs and NF-NETs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.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: 10.1200/JCO.2009.22.8510. [DOI] [PubMed] [Google Scholar]
- 10.Caplin ME, Pavel M, Cwikla Jb, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371:224–233. doi: 10.1056/NEJMoa1316158. [DOI] [PubMed] [Google Scholar]
- 11.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. doi: 10.1016/S0140-6736(15)00817-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yao JC, Shah MH, Ito T, et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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: 10.1056/NEJMoa1003825. [DOI] [PubMed] [Google Scholar]
- 14••.Strosberg JR, Wolin EM, Chasen B, Kulke M, Bushnell DL, Caplin ME, Baum RP, Kunz PL, Hobday TJ, Hendifar AE, Oberg KE, Sierra ML, Kwekkeboom DJ, Ruszniewski PH, Krenning EP. NETTER-1 phase III: Progression-free survival, radiographic response, and preliminary overall survival results in patients with midgut neuroeneodcrine tumors treated with 177-Lu-Dotatate. J Clin Oncol. 2016;34(4-supplement):194. Ref Type: Abstract Preliminary report summarizing results of NETTER-1 study of PRRT using 177Lu-Dotatate. [Google Scholar]
- 15.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: 10.1200/JCO.2007.15.2553. [DOI] [PubMed] [Google Scholar]
- 16.Kwekkeboom DJ, Krenning EP. Peptide Receptor Radionuclide Therapy in the Treatment of Neuroendocrine Tumors. Hematol Oncol Clin North Am. 2016;30:179–191. doi: 10.1016/j.hoc.2015.09.009. [DOI] [PubMed] [Google Scholar]
- 17.Ito T, Igarashi H, Jensen RT. Therapy of metastatic pancreatic neuroendocrine tumors (pNETs): recent insights and advances. J Gastroenterol. 2012;47:941–960. doi: 10.1007/s00535-012-0642-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Frilling A, Clift AK. Therapeutic strategies for neuroendocrine liver metastases. Cancer. 2015;121:1172–1186. doi: 10.1002/cncr.28760. [DOI] [PubMed] [Google Scholar]
- 19•.Frilling A, Al-Nahhas A, Clift AK. Transplantation and Debulking Procedures for Neuroendocrine Tumors. Front Horm Res. 2015;44:164–176. doi: 10.1159/000382145. Review summarizing results of debulking(cytoreductive surgery) and liver transplantation on symptoms/survival in patients with malignant NETs(F-/NF-NETs) [DOI] [PubMed] [Google Scholar]
- 20.De Baere T, Deschamps F, Tselikas L, et al. GEP-NETS update: Interventional radiology: role in the treatment of liver metastases from GEP-NETs. Eur J Endocrinol. 2015;172:R151–R166. doi: 10.1530/EJE-14-0630. [DOI] [PubMed] [Google Scholar]
- 21.Gupta S. Intra-arterial liver-directed therapies for neuroendocrine hepatic metastases. Semin Intervent Radiol. 2013;30:28–38. doi: 10.1055/s-0033-1333651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22•.Kennedy AS. Hepatic-directed Therapies in Patients with Neuroendocrine Tumors. Hematol Oncol Clin North Am. 2016;30:193–207. doi: 10.1016/j.hoc.2015.09.010. Review summarizing reults of liver-directed therapies on symptoms/survival in patients with progressive NETs(F-/NF-NETs) [DOI] [PubMed] [Google Scholar]
- 23.Kunz PL, Reidy-Lagunes D, Anthony LB, et al. Consensus Guidelines for the Management and Treatment of Neuroendocrine Tumors. Pancreas. 2013;42:557–577. doi: 10.1097/MPA.0b013e31828e34a4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24••.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. doi: 10.1159/000443167. Consensus paper dealing with all issues of malignant NETs (F-NETs and NF-NETs) [DOI] [PubMed] [Google Scholar]
- 25.Singh S, Asa SL, Dey C, et al. Diagnosis and management of gastrointestinal neuroendocrine tumors: An evidence-based Canadian consensus. Cancer Treat Rev. 2016;47:32–45. doi: 10.1016/j.ctrv.2016.05.003. [DOI] [PubMed] [Google Scholar]
- 26.Niederle B, Pape UF, Costa F, et al. ENETS Consensus Guidelines Update for Neuroendocrine Neoplasms of the Jejunum and Ileum. Neuroendocrinology. 2016;103:125–138. doi: 10.1159/000443170. [DOI] [PubMed] [Google Scholar]
- 27.Jensen RT, Niederle B, Mitry E, et al. Gastrinoma (duodenal and pancreatic) Neuroendocrinology. 2006;84:173–182. doi: 10.1159/000098009. [DOI] [PubMed] [Google Scholar]
- 28.Jensen RT, Cadiot G, Brandi ML, et al. ENETS Consensus Guidelines for the Management of Patients with Digestive Neuroendocrine Neoplasms: Functional Pancreatic Endocrine Tumor Syndromes. Neuroendocrinology. 2012;95:98–119. doi: 10.1159/000335591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Glanemann M, Shi B, Liang F, et al. Surgical strategies for treatment of malignant pancreatic tumors: extended, standard or local surgery? World J Surg Oncol. 2008;6:123. doi: 10.1186/1477-7819-6-123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Maton PN, Gardner JD, Jensen RT. Cushing’s syndrome in patients with Zollinger-Ellison syndrome. N Engl J Med. 1986;315:1–5. doi: 10.1056/NEJM198607033150101. [DOI] [PubMed] [Google Scholar]
- 31.Rehfeld JF, Federspiel B, Bardram L. A neuroendocrine tumor syndrome from cholecystokinin secretion. N Engl J Med. 2013;368:1165–1166. doi: 10.1056/NEJMc1215137. [DOI] [PubMed] [Google Scholar]
- 32.Ito T, Igarashi H, Uehara H, et al. Causes of Death and Prognostic Factors in Multiple Endocrine Neoplasia Type 1: A Prospective Study: Comparison of 106 MEN1/Zollinger-Ellison Syndrome Patients With 1613 Literature MEN1 Patients With or Without Pancreatic Endocrine Tumors. Medicine (Baltimore) 2013;92:135–181. doi: 10.1097/MD.0b013e3182954af1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33•.Ito T, Igarashi H, Uehara H, et al. Pharmacotherapy of Zollinger-Ellison syndrome. Expert Opin Pharmacotherapy. 2013;14:307–321. doi: 10.1517/14656566.2013.767332. Review summarizing current pharmacotherapy for control of acid hypersecretion in ZES patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ellison EH, Wilson SD. The Zollinger-Ellison syndrome: Re-appraisal and evaluation of 260 registered cases. Ann Surg. 1964;160:512–530. doi: 10.1097/00000658-196409000-00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Roy PK, Venzon DJ, Shojamanesh H, et al. Zollinger-Ellison syndrome: clinical presentation in 261 patients. Medicine (Baltimore) 2000;79:379–411. doi: 10.1097/00005792-200011000-00004. [DOI] [PubMed] [Google Scholar]
- 36.Horsch D, Ezziddin S, Haug A, et al. Effectiveness and side-effects of peptide receptor radionuclide therapy for neuroendocrine neoplasms in Germany: A multi-institutional registry study with prospective follow-up. Eur J Cancer. 2016;58:41–51. doi: 10.1016/j.ejca.2016.01.009. [DOI] [PubMed] [Google Scholar]
- 37.Norton JA, Alexander HR, Fraker DL, et al. Does the use of routine duodenotomy (DUODX) affect rate of cure, development of liver metastases or survival in patients with Zollinger-Ellison syndrome (ZES)? Ann Surg. 2004;239:617–626. doi: 10.1097/01.sla.0000124290.05524.5e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38•.Bainbridge HE, Larbi E, Middleton G. Symptomatic Control of Neuroendocrine Tumours with Everolimus. Horm Cancer. 2015;6:254–259. doi: 10.1007/s12672-015-0233-2. Study evaluating everolimus’s effectiveness to control symptoms of F-NETs. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39•.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: 10.1530/EJE-12-1101. Study demonstrating everolimus’s effectiveness to control symptoms of insulinomas. [DOI] [PubMed] [Google Scholar]
- 40.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: 10.1634/theoncologist.2010-0222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360:195–197. doi: 10.1056/NEJMc0806740. [DOI] [PubMed] [Google Scholar]
- 42.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: 10.1093/jjco/hyt193. [DOI] [PubMed] [Google Scholar]
- 43.Baratelli C, Brizzi MP, Tampellini M, et al. Intermittent everolimus administration for malignant insulinoma. Endocrinol Diabetes Metab Case Rep. 2014;2014:140047. doi: 10.1530/EDM-14-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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: 10.1155/2013/636175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cuesta Hernandez M, Gomez Hoyos E, Marcuello Foncillas C, et al. Advanced malignant insulinoma. Everolimus response and toxicity. Endocrinol Nutr. 2014;61:e1–e3. doi: 10.1016/j.endonu.2013.06.002. [DOI] [PubMed] [Google Scholar]
- 46.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: 10.1111/cen.12059. [DOI] [PubMed] [Google Scholar]
- 47.Bourcier ME, Sherrod A, DiGuardo M, et al. Successful control of intractable hypoglycemia using rapamycin in an 86-year-old man with a pancreatic insulin-secreting islet cell tumor and metastases. J Clin Endocrinol Metab. 2009;94:3157–3162. doi: 10.1210/jc.2009-0788. [DOI] [PubMed] [Google Scholar]
- 48.Raymond E, Hobday T, Castellano D, et al. Therapy innovations: tyrosine kinase inhibitors for the treatment of pancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2011;30(Suppl 1):19–26. doi: 10.1007/s10555-011-9291-2. [DOI] [PubMed] [Google Scholar]
- 49.de Mestier L, Walter T, Brixi H, et al. Sunitinib achieved fast and sustained control of VIPoma symptoms. Eur J Endocrinol. 2015;172:K1–K3. doi: 10.1530/EJE-14-0682. [DOI] [PubMed] [Google Scholar]
- 50•.Chen J, Wang C, Han J, et al. Therapeutic effect of sunitinib malate and its influence on blood glucose concentrations in a patient with metastatic insulinoma. Expert Rev Anticancer Ther. 2013;13:737–743. doi: 10.1586/era.13.45. Study demonstrating sunitinib’s effectiveness to control symptoms of insulinoma. [DOI] [PubMed] [Google Scholar]
- 51.Bourcier ME, Vinik AI. Sunitinib for the Treatment of Metastatic Paraganglioma and Vasoactive Intestinal Polypeptide-Producing Tumor (VIPoma) Pancreas. 2013;42:348–352. doi: 10.1097/MPA.0b013e31825c53fa. [DOI] [PubMed] [Google Scholar]
- 52.Kwekkeboom DJ, Kam BL, Van Essen M, et al. Somatostatin-receptor-based imaging and therapy of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer. 2010;17:R53–R73. doi: 10.1677/ERC-09-0078. [DOI] [PubMed] [Google Scholar]
- 53.Baumann T, Rottenburger C, Nicolas G, et al. Gastroenteropancreatic neuroendocrine tumours (GEP-NET) - Imaging and staging. Best Pract Res Clin Endocrinol Metab. 2016;30:45–57. doi: 10.1016/j.beem.2016.01.003. [DOI] [PubMed] [Google Scholar]
- 54••.Strosberg J, Wolin E, Chasen B, Kulke M, Bushnell DL, Caplin M, Baum RP, Mittra E, Hobday T, Kendifar A, Oberg K, Sierra ML, Kwekkeboom D, Ruszniewski P, Krenning E. 177-Lu-Dotatate significantly improves progression-free survival in patients with midgut neuroendocrine tumors: results of the Phase III NETTER-1 Trial. Abstracts of 2015 NANETs Meeting (C39); 2015. Ref Type: Abstract Preliminary report summarizing results of NETTER-1 study of PRRT using 177Lu-Dotatate. [Google Scholar]
- 55.van Schaik E, van Vliet EI, Feelders RA, et al. Improved control of severe hypoglycemia in patients with malignant insulinomas by Peptide receptor radionuclide therapy. J Clin Endocrinol Metab. 2011;96:3381–3389. doi: 10.1210/jc.2011-1563. [DOI] [PubMed] [Google Scholar]
- 56••.Koffas A, Popat R, Dimitriou N, Quigley AM, Navalkissoor S, Caplin M, Toumpanakis C. Efficacy of Lutetium-177 DOTA octreotate peptide receptor radionuclide therapy in patients with advanced neruoendocrine tumors and carcinoid syndrome refractory to somatostatin analogues. Abstract book of 13th Annual ENET Conference; 9–11 March 2016; Barcelona, Spain. 2016. p. 228. Ref Type: Abstract Study reporting ability of a PRRT to control symptoms of refractory carcinoid syndrome. [Google Scholar]
- 57.Seregni E, Maccauro M, Coliva A, et al. Treatment with tandem [(90)Y]DOTA-TATE and [(177)Lu] DOTA-TATE of neuroendocrine tumors refractory to conventional therapy: preliminary results. Q J Nucl Med Mol Imaging. 2010;54:84–91. [PubMed] [Google Scholar]
- 58.Chandra P, Yarandi SS, Khazai N, et al. Management of intractable hypoglycemia with Yttirum-90 radioembolization in a patient with malignant insulinoma. Am J Med Sci. 2010;340:414–417. doi: 10.1097/MAJ.0b013e3181ee7be2. [DOI] [PubMed] [Google Scholar]
- 59.Costa R, Costa R, Bacchi CE, et al. Metastatic insulinoma managed with radiolabeled somatostatin analog. Case Rep Endocrinol. 2013;2013:252159. doi: 10.1155/2013/252159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Fischbach J, Gut P, Matysiak-Grzes M, et al. Combined octreotide and peptide receptor radionuclide therapy ((90)Y-DOTA-TATE) in case of malignant insulinoma. Neuro Endocrinol Lett. 2012;33:273–278. [PubMed] [Google Scholar]
- 61•.Makis W, McCann K, McEwan AJ. Metastatic Insulinoma Pancreatic Neuroendocrine Tumor Treated With 177Lu-DOTATATE Induction and Maintenance Peptide Receptor Radionuclide Therapy: A Suggested Protocol. Clin Nucl Med. 2016;41:53–54. doi: 10.1097/RLU.0000000000001023. Study reporting ability of a PRRT to control symptoms of refractory insulinoma. [DOI] [PubMed] [Google Scholar]
- 62.Makis W, McCann K, Riauka TA, et al. Ectopic Corticotropin-Producing Neuroendocrine Tumor of the Pancreas Treated With 177Lu DOTATATE Induction and Maintenance Peptide Receptor Radionuclide Therapy. Clin Nucl Med. 2016;41:50–52. doi: 10.1097/RLU.0000000000000971. [DOI] [PubMed] [Google Scholar]
- 63•.Makis W, McCann K, Riauka TA, et al. Glucagonoma Pancreatic Neuroendocrine Tumor Treated With 177Lu DOTATATE Induction and Maintenance Peptide Receptor Radionuclide Therapy. Clin Nucl Med. 2015;40:877–879. doi: 10.1097/RLU.0000000000000891. Study reporting ability of a PRRT to control glucagonoma. [DOI] [PubMed] [Google Scholar]
- 64.Makis W, McCann K, Riauka TA, et al. Resolution of Hyperreninemia, Secondary Hyperaldosteronism, and Hypokalemia With 177Lu-DOTATATE Induction and Maintenance Peptide Receptor Radionuclide Therapy in a Patient With Pancreatic Neuroendocrine Tumor. Clin Nucl Med. 2015;40:880–884. doi: 10.1097/RLU.0000000000000962. [DOI] [PubMed] [Google Scholar]
- 65.Davi MV, Bodei L, Ferdeghini M, et al. Multidisciplinary approach including receptor radionuclide therapy with 90Y-DOTATOC ([90Y-DOTA0, Tyr3]-octreotide) and 177Lu-DOTATATE ([177Lu-DOTA0, Tyr3]-octreotate) in ectopic cushing syndrome from a metastatic gastrinoma: a promising proposal. Endocr Pract. 2008;14:213–218. doi: 10.4158/EP.14.2.213. [DOI] [PubMed] [Google Scholar]
- 66.Sowa-Staszczak A, Pach D, Kunikowska J, et al. Efficacy and safety of 90Y-DOTATATE therapy in neuroendocrine tumours. Endokrynol Pol. 2011;62:392–400. [PubMed] [Google Scholar]
- 67.Nieman LK. Cushing’s syndrome: update on signs, symptoms and biochemical screening. Eur J Endocrinol. 2015;173:M33–M38. doi: 10.1530/EJE-15-0464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Del Prete M, Fiore F, Modica R, et al. Hepatic arterial embolization in patients with neuroendocrine tumors. J Exp Clin Cancer Res. 2014;33:43. doi: 10.1186/1756-9966-33-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69•.Kennedy A, Bester L, Salem R, et al. Role of hepatic intra-arterial therapies in metastatic neuroendocrine tumours (NET): guidelines from the NET-Liver-Metastases Consensus Conference. HPB (Oxford) 2015;17:29–37. doi: 10.1111/hpb.12326. Review summarizing the results with liver-directed therapies for control symptoms/survival in malignant NETs(f-/NF-NETs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yang TX, Chua TC, Morris DL. Radioembolization and chemoembolization for unresectable neuroendocrine liver metastases - a systematic review. Surg Oncol. 2012;21:299–308. doi: 10.1016/j.suronc.2012.07.001. [DOI] [PubMed] [Google Scholar]
- 71.Fiore F, Del Prete M, Franco R, et al. Transarterial embolization (TAE) is equally effective and slightly safer than transarterial chemoembolization (TACE) to manage liver metastases in neuroendocrine tumors. Endocrine. 2014;47:177–182. doi: 10.1007/s12020-013-0130-9. [DOI] [PubMed] [Google Scholar]
- 72.Bloomston M, Al-Saif O, Klemanski D, et al. Hepatic artery chemoembolization in 122 patients with metastatic carcinoid tumor: lessons learned. J Gastrointest Surg. 2007;11:264–271. doi: 10.1007/s11605-007-0089-z. [DOI] [PubMed] [Google Scholar]
- 73•.Kyriacou A, Mansoor W, Lawrance J, et al. Intractable hypoglycaemia in a patient with advanced carcinoid syndrome successfully treated with hepatic embolization. Hormones (Athens ) 2015;15:118–121. doi: 10.14310/horm.2002.1607. Study reporting ability of a PRRT to control symptoms of refractory carcinid syndrome. [DOI] [PubMed] [Google Scholar]
- 74.Maiza JC, Vezzosi D, Grunenwald S, et al. Treatment with somatostatin analogs and chemoembolization of liver metastases for severe hypoglycemia in malignant insulinomas. J Endocrinol Invest. 2011;34:e253–e258. doi: 10.3275/7762. [DOI] [PubMed] [Google Scholar]
- 75.Moscetti L, Saltarelli R, Giuliani R, et al. Intra-arterial liver chemotherapy and hormone therapy in malignant insulinoma: case report and review of the literature. Tumori. 2000;86:475–479. doi: 10.1177/030089160008600609. [DOI] [PubMed] [Google Scholar]
- 76•.Muro S, Nasu J, Harada R, et al. Prompt resolution of hypoglycemia by hepatic transarterial embolization for malignant insulinoma with multiple liver metastases. Acta Med Okayama. 2014;68:303–306. doi: 10.18926/AMO/52900. Study reporting ability of a embolization to control symptoms of refractory insulinoma. [DOI] [PubMed] [Google Scholar]
- 77.Pericleous M, Caplin ME, Tsochatzis E, et al. Hepatic artery embolization in advanced neuroendocrine tumors: Efficacy and long-term outcomes. Asia Pac J Clin Oncol. 2016;12:61–69. doi: 10.1111/ajco.12438. [DOI] [PubMed] [Google Scholar]
- 78•.Yarmohammadi H, Erinjeri JP, Brown KT. Embolization of metastatic neuroendocrine tumor resulting in clinical manifestations of syndrome of inappropriate secretion of antidiuretic hormone. J Vasc Interv Radiol. 2015;26:533–537. doi: 10.1016/j.jvir.2014.11.032. Study reporting ability of a embolization to control symptoms of SIADH by F-NET. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Harring TR, Nguyen NT, Goss JA, et al. Treatment of liver metastases in patients with neuroendocrine tumors: a comprehensive review. Int J Hepatol. 2011;2011:1–11. doi: 10.4061/2011/154541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Eriksson J, Stalberg P, Nilsson A, et al. Surgery and radiofrequency ablation for treatment of liver metastases from midgut and foregut carcinoids and endocrine pancreatic tumors. World J Surg. 2008;32:930–938. doi: 10.1007/s00268-008-9510-3. [DOI] [PubMed] [Google Scholar]
- 81.Mazzaglia PJ, Berber E, Milas M, et al. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases: a 10-year experience evaluating predictors of survival. Surgery. 2007;142:10–19. doi: 10.1016/j.surg.2007.01.036. [DOI] [PubMed] [Google Scholar]
- 82.Karabulut K, Akyildiz HY, Lance C, et al. Multimodality treatment of neuroendocrine liver metastases. Surgery. 2011;150:316–325. doi: 10.1016/j.surg.2011.05.008. [DOI] [PubMed] [Google Scholar]
- 83.Gillams A, Cassoni A, Conway G, et al. Radiofrequency ablation of neuroendocrine liver metastases: the Middlesex experience. Abdom Imaging. 2005;30:435–441. doi: 10.1007/s00261-004-0258-4. [DOI] [PubMed] [Google Scholar]
- 84.Armellini E, Crino SF, Ballare M, et al. Endoscopic ultrasound-guided ethanol ablation of pancreatic neuroendocrine tumours: A case study and literature review. World J Gastrointest Endosc. 2016;8:192–197. doi: 10.4253/wjge.v8.i3.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Deprez PH, Claessens A, Borbath I, et al. Successful endoscopic ultrasound-guided ethanol ablation of a sporadic insulinoma. Acta Gastroenterol Belg. 2008;71:333–337. [PubMed] [Google Scholar]
- 86.Jurgensen C, Schuppan D, Neser F, et al. EUS-guided alcohol ablation of an insulinoma. Gastrointest Endosc. 2006;63:1059–1062. doi: 10.1016/j.gie.2005.10.034. [DOI] [PubMed] [Google Scholar]
- 87.Lakhtakia S, Ramchandani M, Galasso D, et al. EUS-guided radiofrequency ablation for management of pancreatic insulinoma by using a novel needle electrode (with videos) Gastrointest Endosc. 2016;83:234–239. doi: 10.1016/j.gie.2015.08.085. [DOI] [PubMed] [Google Scholar]
- 88.Lee MJ, Jung CH, Jang JE, et al. Successful endoscopic ultrasound-guided ethanol ablation of multiple insulinomas accompanied with multiple endocrine neoplasia type 1. Intern Med J. 2013;43:948–950. doi: 10.1111/imj.12208. [DOI] [PubMed] [Google Scholar]
- 89•.Levy MJ, Thompson GB, Topazian MD, et al. US-guided ethanol ablation of insulinomas: a new treatment option. Gastrointest Endosc. 2012;75:200–206. doi: 10.1016/j.gie.2011.09.019. Study reporting series showing ability of a US-guided ethanol injection to control symptoms of insulinoma. [DOI] [PubMed] [Google Scholar]
- 90.Qin SY, Lu XP, Jiang HX. EUS-guided ethanol ablation of insulinomas: case series and literature review. Medicine (Baltimore) 2014;93:e85. doi: 10.1097/MD.0000000000000085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Muscatiello N, Salcuni A, Macarini L, et al. Treatment of a pancreatic endocrine tumor by ethanol injection guided by endoscopic ultrasound. Endoscopy. 2008;40(Suppl 2):E258–E259. doi: 10.1055/s-2007-966962. [DOI] [PubMed] [Google Scholar]
- 92.Park dH, Choi JH, Oh D, et al. Endoscopic ultrasonography-guided ethanol ablation for small pancreatic neuroendocrine tumors: results of a pilot study. Clin Endosc. 2015;48:158–164. doi: 10.5946/ce.2015.48.2.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93•.Waung JA, Todd JF, Keane MG, et al. Successful management of a sporadic pancreatic insulinoma by endoscopic ultrasound-guided radiofrequency ablation. Endoscopy. 2016;48(Suppl 1):E144–E145. doi: 10.1055/s-0042-104650. Study reporting series showing ability of a US-guided ethanol injection to control symptoms of insulinoma. [DOI] [PubMed] [Google Scholar]
- 94.Broder MS, Beenhouwer D, Strosberg JR, et al. Gastrointestinal neuroendocrine tumors treated with high dose octreotide-LAR: a systematic literature review. World J Gastroenterol. 2015;21:1945–1955. doi: 10.3748/wjg.v21.i6.1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Khan MS, El-Khouly F, Davies P, et al. Long-term results of treatment of malignant carcinoid syndrome with prolonged release Lanreotide (Somatuline Autogel) Aliment Pharmacol Ther. 2011;34:235–242. doi: 10.1111/j.1365-2036.2011.04693.x. [DOI] [PubMed] [Google Scholar]
- 96•.Ruszniewski P, Valle JW, Lombard-Bohas C, et al. Patient-reported outcomes with lanreotide Autogel/Depot for carcinoid syndrome: An international observational study. Dig Liver Dis. 2016;48:552–558. doi: 10.1016/j.dld.2015.12.013. Study reporting effectiveness of lanreotide autogel to control symptoms of carcinoid syndrome, with dose-escalation. [DOI] [PubMed] [Google Scholar]
- 97.Strosberg JR, Benson AB, Huynh L, et al. Clinical benefits of above-standard dose of octreotide LAR in patients with neuroendocrine tumors for control of carcinoid syndrome symptoms: a multicenter retrospective chart review study. Oncologist. 2014;19:930–936. doi: 10.1634/theoncologist.2014-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Vinik AI, Wolin EM, Liyanage N, et al. Evaluation ofLanreotide Depot/Autogel efficacy and safety as a carcinoid syndrome treatment (elect): A randomized, double-blind placebo-controlled study. Endocr Pract. 2016 doi: 10.4158/EP151172.OR. In press. [DOI] [PubMed] [Google Scholar]
- 99.Vezzosi D, Bennet A, Courbon F, et al. Short- and long-term somatostatin analogue treatment in patients with hypoglycaemia related to endogenous hyperinsulinism. Clin Endocrinol (Oxf) 2008;68:904–911. doi: 10.1111/j.1365-2265.2007.03136.x. [DOI] [PubMed] [Google Scholar]
- 100.Koelz A, Kraenzlin M, Gyr K, et al. Escape of the response to a long-acting somatostatin analogue (SMS 201-995) in patients with VIPoma. Gastroenterology. 1987;92(2):527–531. doi: 10.1016/0016-5085(87)90153-3. [DOI] [PubMed] [Google Scholar]
- 101.Maton PN, Gardner JD, Jensen RT. Use of the long-acting somatostatin analog, SMS 201–995 in patients with pancreatic islet cell tumors. Dig Dis Sci. 1989;34:28S–39S. doi: 10.1007/BF01536043. [DOI] [PubMed] [Google Scholar]
- 102.Peng SY, Li JT, Liu YB, et al. Diagnosis and treatment of VIPoma in China: (case report and 31 cases review) diagnosis and treatment of VIPoma. Pancreas. 2004;28:93–97. doi: 10.1097/00006676-200401000-00015. [DOI] [PubMed] [Google Scholar]
- 103.Farley HA, Pommier RF. Treatment of Neuroendocrine Liver Metastases. Surg Oncol Clin N Am. 2016;25:217–225. doi: 10.1016/j.soc.2015.08.010. [DOI] [PubMed] [Google Scholar]
- 104•.Lesurtel M, Nagorney DM, Mazzaferro V, et al. When should a liver resection be performed in patients with liver metastases from neuroendocrine tumours? A systematic review with practice recommendations. HPB (Oxford) 2015;17:17–22. doi: 10.1111/hpb.12225. Consensus statement analyzing existing studies on cytoreductive surgery in progressing NETs(F-/NF-NETs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Khan MU, Morse M, Coleman RE. Radioiodinated metaiodobenzylguanidine in the diagnosis and therapy of carcinoid tumors. Q J Nucl Med Mol Imaging. 2008;52:441–454. [PubMed] [Google Scholar]
- 106.Taal BG, Hoefnagel CA, Valdes Olmos RA, et al. Palliative effect of metaiodobenzylguanidine in metastatic carcinoid tumors. J Clin Oncol. 1996;14:1829–1838. doi: 10.1200/JCO.1996.14.6.1829. [DOI] [PubMed] [Google Scholar]
- 107.Bomanji JB, Wong W, Gaze MN, et al. Treatment of neuroendocrine tumours in adults with 131I-MIBG therapy. Clin Oncol (R Coll Radiol ) 2003;15:193–198. doi: 10.1016/s0936-6555(02)00273-x. [DOI] [PubMed] [Google Scholar]
- 108.Pathirana AA, Vinjamuri S, Byrne C, et al. (131)I-MIBG radionuclide therapy is safe and cost-effective in the control of symptoms of the carcinoid syndrome. Eur J Surg Oncol. 2001;27:404–408. doi: 10.1053/ejso.2001.1132. [DOI] [PubMed] [Google Scholar]
- 109.Safford SD, Coleman RE, Gockerman JP, et al. Iodine-131 metaiodobenzylguanidine treatment for metastatic carcinoid. Results in 98 patients. Cancer. 2004;101:1987–1993. doi: 10.1002/cncr.20592. [DOI] [PubMed] [Google Scholar]
- 110.Kaltsas G, Korbonits M, Heintz E, et al. Comparison of somatostatin analog and meta-iodobenzylguanidine radionuclides in the diagnosis and localization of advanced neuroendocrine tumors. J Clin Endocrinol Metab. 2001;86:895–902. doi: 10.1210/jcem.86.2.7194. [DOI] [PubMed] [Google Scholar]
- 111•.Feitosa AC, Castro Junior DO, Rocha Filho J, et al. Spinal cord compression after radiolabeled metaiodobenzylguanidine analogue therapy in advanced malignant insulinoma. Arch Endocrinol Metab. 2015;59:186–189. doi: 10.1590/2359-3997000000033. Study reporting effectiveness of 131I-MIBG in a patient with malignant insulinoma. [DOI] [PubMed] [Google Scholar]
- 112.Pasieka JL, McEwan AJ, Rorstad O. The palliative role of 131I-MIBG and 111In-octreotide therapy in patients with metastatic progressive neuroendocrine neoplasms. Surgery. 2004;136:1218–1226. doi: 10.1016/j.surg.2004.06.050. [DOI] [PubMed] [Google Scholar]
- 113.Gelhorn HL, Kulke MH, O’Dorisio T, et al. Patient-reported Symptom Experiences in Patients With Carcinoid Syndrome After Participation in a Study of Telotristat Etiprate: A Qualitative Interview Approach. Clin Ther. 2016;38:759–768. doi: 10.1016/j.clinthera.2016.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Pavel M, Wiedenmann B, Caplin M, Horsch D, Freiman J, Law L, Banks P, Frazier K, Jackson J, Lapuerta P, Sands A, Zambrowicz B. Telotristat etiprate produces clinical and biochemical responses in patients with carcinoid syndrome: results of a phase 2, multicenter, opern-lable, serial-ascending study. ENETs 2013 Meeting abstracts(M4); 2013. p. 153. Ref Type: Abstract. [Google Scholar]
- 115•.Molina-Cerrillo J, Alonso-Gordoa T, Martinez-Saez O, et al. Inhibition of Peripheral Synthesis of Serotonin as a New Target in Neuroendocrine Tumors. Oncologist. 2016;21:701–707. doi: 10.1634/theoncologist.2015-0455. Review of the effectiveness ot Telotristat in controlling diarrhea in patients with carcinoid syndrome. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116••.Horsch D, Kulke M, Caplin M, Anthony L, Bergsland E, Oberg K, Welin S, Warner R, Lombard-Bohas C, Kunz P, Valle J, Fleming D, Lapuerta P, Banks P, Pavel M. Efficacy and safety of Telostristat Etiprate in patients with carcinoid syndrome not adequately controlled by somatostatin analog therapy: analysis of ongoing TELESTAR extension period. Abstract book of 13th Annual ENET Conference; 9–11 March 2016; Barcelona, Spain. 2016. p. 245. Ref Type: Abstract Study reporting effectiveness ot Telotristat in controlling diarrhea in patients with carcinoid syndrome. [Google Scholar]
- 117•.Pavel M, Horsch D, Caplin M, et al. Telotristat etiprate for carcinoid syndrome: a single-arm, multicenter trial. J Clin Endocrinol Metab. 2015;100:1511–1519. doi: 10.1210/jc.2014-2247. Study reporting effectiveness ot Telotristat in controlling diarrhea in patients with carcinoid syndrome. [DOI] [PubMed] [Google Scholar]
- 118.Pavel M, Horsch D, Anthony L, Ervin C, Kulke M, Bergsland E, Caplin M, Oberg K, Warner R, Kunz P, Metz DC, Pasieka J, Pavlakis N, DiBenedetti D, Lapuerta P. Patient interviews in TELESTAR, a Phase 3 study of Telotristat etiprate, report meaningful improvement in carcnoid syndrome. Abstract book of 13th Annual ENET Conference; 9–11 March 2016; Barcelona, Spain. 2016. p. 247. Ref Type: Abstract. [Google Scholar]
- 119.Zacks J, Lavine R, Ratner L, Warner R. Telostristat etiprate appears to halt carcinoid heart disease. Abstract book of 13th Annual ENET Conference; 9–11 March 2016; Barcelona, Spain. 2016. p. 248. Ref Type: Abstract. [Google Scholar]
- 120•.Baldelli R, Barnabei A, Rizza L, et al. Somatostatin Analogs Therapy in Gastroenteropancreatic Neuroendocrine Tumors: Current Aspects and New Perspectives. Front Endocrinol (Lausanne) 2014;5:7. doi: 10.3389/fendo.2014.00007. Review of various aspects of use of somatostatin anlogues for control of symptoms and growth in patients with NETs(F-/NF-NETs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cives M, Kunz P, Morse B, et al. Phase II clinical trial of pasireotide LAR in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer. 2014 doi: 10.1530/ERC-14-0360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122•.Kvols LK, Oberg KE, O’Dorisio TM, et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer. 2012;19:657–666. doi: 10.1530/ERC-11-0367. Study reporting effectiveness ot Pasireotide in controlling symptoms in a subset of patients with refractory carcinoid syndrome. [DOI] [PubMed] [Google Scholar]
- 123•.Wemeau JL. Therapeutic innovations in endocrine diseases - Part 4 : Pasireotide: Long-acting release somatostatin analogue. Presse Med. 2016;45:e217–e220. doi: 10.1016/j.lpm.2016.05.009. Review of the efficacy and safety of pasireotide in various hormone-excess states. [DOI] [PubMed] [Google Scholar]
- 124.Wolin EM, Jarzab B, Eriksson B, et al. Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Des Devel Ther. 2015;9:5075–5086. doi: 10.2147/DDDT.S84177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125•.Tirosh A, Stemmer SM, Solomonov E, et al. Pasireotide for malignant insulinoma. Hormones (Athens ) 2016;15:271–276. doi: 10.14310/horm.2002.1639. Study reporting effectiveness ot Pasireotide in controlling symptoms in a patient with malignanat insulinoma. [DOI] [PubMed] [Google Scholar]
- 126.Quinn TJ, Yuan Z, Adem A, et al. Pasireotide (SOM230) is effective for the treatment of pancreatic neuroendocrine tumors (PNETs) in a multiple endocrine neoplasia type 1 (MEN1) conditional knockout mouse model. Surgery. 2012;152:1068–1077. doi: 10.1016/j.surg.2012.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wolin EM, Hu K, Hughes G, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of a long-acting release (LAR) formulation of pasireotide (SOM230) in patients with gastroenteropancreatic neuroendocrine tumors: results from a randomized, multicenter, open-label, phase I study. Cancer Chemother Pharmacol. 2013;72:387–395. doi: 10.1007/s00280-013-2202-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Henry RR, Ciaraldi TP, Armstrong D, et al. Hyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteers. J Clin Endocrinol Metab. 2013;98:3446–3453. doi: 10.1210/jc.2013-1771. [DOI] [PubMed] [Google Scholar]
- 129.Kamp K, Alwani RA, Korpershoek E, et al. Prevalence and clinical features of the ectopic ACTH syndrome in patients with gastroenteropancreatic and thoracic neuroendocrine tumors. Eur J Endocrinol. 2016;174:271–280. doi: 10.1530/EJE-15-0968. [DOI] [PubMed] [Google Scholar]
- 130.Yu F, Venzon DJ, Serrano J, et al. Prospective study of the clinical course, prognostic factors and survival in patients with longstanding Zollinger-Ellison syndrome. J Clin Oncol. 1999;17:615–630. doi: 10.1200/JCO.1999.17.2.615. [DOI] [PubMed] [Google Scholar]
- 131.Ambrogio AG, Cavagnini F. Role of “old” pharmacological agents in the treatment of Cushing’s syndrome. J Endocrinol Invest. 2016 doi: 10.1007/s40618-016-0462-4. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132•.Creemers SG, Hofland LJ, Lamberts SW, et al. Cushing’s syndrome: an update on current pharmacotherapy and future directions. Expert Opin Pharmacother. 2015;16:1829–1844. doi: 10.1517/14656566.2015.1061995. Review of old and newer treatments of Cushing’s syndrome including ectopic ACTHomas. [DOI] [PubMed] [Google Scholar]
- 133.Lacroix A, Feelders RA, Stratakis CA, et al. Cushing’s syndrome. Lancet. 2015;386:913–927. doi: 10.1016/S0140-6736(14)61375-1. [DOI] [PubMed] [Google Scholar]
- 134.Nieman LK, Biller BM, Findling JW, et al. Treatment of Cushing’s Syndrome: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2015;100:2807–2831. doi: 10.1210/jc.2015-1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135•.Moraitis AG, Auchus RJ. Mifepristone Improves Octreotide Efficacy in Resistant Ectopic Cushing’s Syndrome. Case Rep Endocrinol. 2016;2016:8453801. doi: 10.1155/2016/8453801. Use of mifepristone to control Cushing’s syndrome in a patient with pancreatic ACTHomas. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Jensen RT, Berna MJ, Bingham MD, et al. Inherited pancreatic endocrine tumor syndromes: advances in molecular pathogenesis, diagnosis, management and controversies. Cancer. 2008;113(7 suppl):1807–1843. doi: 10.1002/cncr.23648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Benya RV, Metz DC, Venzon DJ, et al. Zollinger-Ellison syndrome can be the initial endocrine manifestation in patients with multiple endocrine neoplasia-type 1. Am J Med. 1994;97:436–444. doi: 10.1016/0002-9343(94)90323-9. [DOI] [PubMed] [Google Scholar]
- 138.Gibril F, Doppman JL, Reynolds JC, et al. Bone metastases in patients with gastrinomas: a prospective study of bone scanning, somatostatin receptor scanning, and MRI in their detection, their frequency, location and effect of their detection on management. J Clin Oncol. 1998;16:1040–1053. doi: 10.1200/JCO.1998.16.3.1040. [DOI] [PubMed] [Google Scholar]
- 139.von Schrenck T, Howard JM, Doppman JL, et al. Prospective study of chemotherapy in patients with metastatic gastrinoma. Gastroenterology. 1988;94:1326–1334. doi: 10.1016/0016-5085(88)90670-1. [DOI] [PubMed] [Google Scholar]
- 140.Krudy AG, Doppman JL, Jensen RT, et al. Localization of islet cell tumors by dynamic CT: Comparison with plain CT, arteriography, sonography and venous sampling. Am J Roentgenol. 1984;143:585–589. doi: 10.2214/ajr.143.3.585. [DOI] [PubMed] [Google Scholar]
- 141.Thom AK, Norton JA, Axiotis CA, et al. Location, incidence and malignant potential of duodenal gastrinomas. Surgery. 1991;110:1086–1093. [PubMed] [Google Scholar]
- 142.Ito T, Jensen RT. Imaging in multiple endocrine neoplasia type 1: recent studies show enhanced sensitivities but increased controversies. Int J Endocr Oncol. 2016;3:53–66. doi: 10.2217/ije.15.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Doppman JL, Miller DL, Chang R, et al. Gastrinomas: localization by means of selective intraarterial injection of secretin. Radiology. 1990;174:25–29. doi: 10.1148/radiology.174.1.2294556. [DOI] [PubMed] [Google Scholar]
- 144.Cherner JA, Doppman JL, Norton JA, et al. Selective venous sampling for gastrin to localize gastrinomas. A prospective study. Ann Intern Med. 1986;105:841–847. doi: 10.7326/0003-4819-105-6-841. [DOI] [PubMed] [Google Scholar]
- 145.Pavel M, Baudin E, Couvelard A, et al. ENETS Consensus Guidelines for the Management of Patients with Liver and Other Distant Metastases from Neuroendocrine Neoplasms of Foregut, Midgut, Hindgut, and Unknown Primary. Neuroendocrinology. 2012;95:157–176. doi: 10.1159/000335597. [DOI] [PubMed] [Google Scholar]
- 146•.Pokuri VK, Fong MK, Iyer R. Octreotide and Lanreotide in Gastroenteropancreatic Neuroendocrine Tumors. Curr Oncol Rep. 2016;18:7. doi: 10.1007/s11912-015-0492-7. Review of treatment of symptoms/tumor growth by NETs(F-/Nf-NETs) by somatostatin analogues. [DOI] [PubMed] [Google Scholar]
- 147.Patel C, Mathur M, Escarcega RO, et al. Carcinoid heart disease: current understanding and future directions. Am Heart J. 2014;167:789–795. doi: 10.1016/j.ahj.2014.03.018. [DOI] [PubMed] [Google Scholar]
- 148•.Bushnell DL, Jr, O’Dorisio TM, O’Dorisio MS, et al. 90Y-edotreotide for metastatic carcinoid refractory to octreotide. J Clin Oncol. 2010;28:1652–1659. doi: 10.1200/JCO.2009.22.8585. Study reporting ability of PRRT with 90Y-edotreotide to control of symptoms/tumor growth by malignant NETs(F-/Nf-NETs) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Gu P, Wu J, Newman E, et al. Treatment of liver metastases in patients with neuroendocrine tumors of gastroesophageal and pancreatic origin. Int J Hepatol. 2012;2012(Article ID131659):1–8. doi: 10.1155/2012/131659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.King J, Quinn R, Glenn DM, et al. Radioembolization with selective internal radiation microspheres for neuroendocrine liver metastases. Cancer. 2008;113:921–929. doi: 10.1002/cncr.23685. [DOI] [PubMed] [Google Scholar]
- 151.Paprottka PM, Hoffmann RT, Haug A, et al. Radioembolization of symptomatic, unresectable neuroendocrine hepatic metastases using yttrium-90 microspheres. Cardiovasc Intervent Radiol. 2012;35:334–342. doi: 10.1007/s00270-011-0248-1. [DOI] [PubMed] [Google Scholar]
- 152.Cazzato RL, Garnon J, Ramamurthy N, et al. 18F-FDOPA PET/CT-Guided Radiofrequency Ablation of Liver Metastases from Neuroendocrine Tumours: Technical Note on a Preliminary Experience. Cardiovasc Intervent Radiol. 2016 doi: 10.1007/s00270-016-1334-1. (In press) [DOI] [PubMed] [Google Scholar]
- 153.Chadha MK, Lombardo J, Mashtare T, et al. High-dose octreotide acetate for management of gastroenteropancreatic neuroendocrine tumors. Anticancer Res. 2009;29:4127–4130. [PubMed] [Google Scholar]
- 154.Anthony L, Vinik AI. Evaluating the characteristics and the management of patients with neuroendocrine tumors receiving octreotide LAR during a 6-year period. Pancreas. 2011;40:987–994. doi: 10.1097/MPA.0b013e31821f66b4. [DOI] [PubMed] [Google Scholar]
- 155.Ferolla P, Faggiano A, Grimaldi F, et al. Shortened interval of long-acting octreotide administration is effective in patients with well-differentiated neuroendocrine carcinomas in progression on standard doses. J Endocrinol Invest. 2012;35:326–331. doi: 10.3275/7869. [DOI] [PubMed] [Google Scholar]
- 156.McEntee GP, Nagorney DM, Kvols LK, et al. Cytoreductive hepatic surgery for neuroendocrine tumors. Surgery. 1990;108:1091–1096. [PubMed] [Google Scholar]
- 157.Que FG, Sarmiento JM, Nagorney DM. Hepatic surgery for metastatic gastrointestinal neuroendocrine tumors. Adv Exp Med Biol. 2006;574:43–56. doi: 10.1007/0-387-29512-7_7. [DOI] [PubMed] [Google Scholar]
- 158.Sarmiento JM, Heywood G, Rubin J, et al. Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. J Am Coll Surg. 2003;197:29–37. doi: 10.1016/S1072-7515(03)00230-8. [DOI] [PubMed] [Google Scholar]
- 159.Farley HA, Pommier RF. Surgical Treatment of Small Bowel Neuroendocrine Tumors. Hematol Oncol Clin North Am. 2016;30:49–61. doi: 10.1016/j.hoc.2015.09.001. [DOI] [PubMed] [Google Scholar]
- 160.Berber E, Flesher N, Siperstein AE. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases. World J Surg. 2002;26:985–990. doi: 10.1007/s00268-002-6629-5. [DOI] [PubMed] [Google Scholar]
- 161•.Ezziddin S, Sabet A, Logvinski T, et al. Long-term outcome and toxicity after dose-intensified treatment with 131I-MIBG for advanced metastatic carcinoid tumors. J Nucl Med. 2013;54:2032–2038. doi: 10.2967/jnumed.112.119313. Study of the ability of 131I-MIBG to control symptoms/tumor growth in patients with metastatic carcinoid tumors. [DOI] [PubMed] [Google Scholar]
- 162.Grunwald F, Ezziddin S. 131I-metaiodobenzylguanidine therapy of neuroblastoma and other neuroendocrine tumors. Semin Nucl Med. 2010;40:153–163. doi: 10.1053/j.semnuclmed.2009.11.004. [DOI] [PubMed] [Google Scholar]
- 163.Mulholland N, Chakravartty R, Devlin L, et al. Long-term outcomes of (131)Iodine MIBG therapy in metastatic gastrointestinal pancreatic neuroendocrine tumours: single administration predicts non-responders. Eur J Nucl Med Mol Imaging. 2015;42:2002–2012. doi: 10.1007/s00259-015-3116-4. [DOI] [PubMed] [Google Scholar]
- 164.Buscombe JR, Cwikla Jb, Caplin ME, et al. Long-term efficacy of low activity meta-[131I]iodobenzylguanidine therapy in patients with disseminated neuroendocrine tumours depends on initial response. Nucl Med Commun. 2005;26:969–976. doi: 10.1097/01.mnm.0000184941.06123.b9. [DOI] [PubMed] [Google Scholar]
- 165.Usukura M, Yoneda T, Oda N, et al. Medical treatment of benign insulinoma using octreotide LAR: a case report. Endocr J. 2007;54:95–101. doi: 10.1507/endocrj.k05-157. [DOI] [PubMed] [Google Scholar]
- 166.Vezzosi D, Bennet A, Rochaix P, et al. Octreotide in insulinoma patients: efficacy on hypoglycemia, relationships with Octreoscan scintigraphy and immunostaining with anti-sst2A and anti-sst5 antibodies. Eur J Endocrinol. 2005;152:757–767. doi: 10.1530/eje.1.01901. [DOI] [PubMed] [Google Scholar]
- 167•.Baudin E, Caron P, Lombard-Bohas C, et al. Malignant insulinoma: recommendations for characterisation and treatment. Ann Endocrinol (Paris) 2013;74:523–533. doi: 10.1016/j.ando.2013.07.001. Review of treatment of patients with malignat insulinomas. [DOI] [PubMed] [Google Scholar]
- 168.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: 10.1530/acta.0.1210034. [DOI] [PubMed] [Google Scholar]
- 169.Healy ML, Dawson SJ, Murray RM, et al. Severe hypoglycaemia after long-acting octreotide in a patient with an unrecognized malignant insulinoma. Intern Med J. 2007;37:406–409. doi: 10.1111/j.1445-5994.2007.01371.x. [DOI] [PubMed] [Google Scholar]
- 170.Ong GS, Henley DE, Hurley D, et al. Therapies for the medical management of persistent hypoglycaemia in two cases of inoperable malignant insulinoma. Eur J Endocrinol. 2010;162:1001–1008. doi: 10.1530/EJE-09-1010. [DOI] [PubMed] [Google Scholar]
- 171.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: 10.1530/EJE-12-1101. [DOI] [PubMed] [Google Scholar]
- 172.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: 10.1016/j.pan.2014.07.011. [DOI] [PubMed] [Google Scholar]
- 173.Chamberlain RS, Canes D, Brown KT, et al. Hepatic neuroendocrine metastases: does intervention alter outcomes? J Am Coll Surg. 2000;190:432–445. doi: 10.1016/s1072-7515(00)00222-2. [DOI] [PubMed] [Google Scholar]
- 174.Yao KA, Talamonti MS, Nemcek A, et al. Indications and results of liver resection and hepatic chemoembolization for metastatic gastrointestinal neuroendocrine tumors. Surgery. 2001;130:677–682. doi: 10.1067/msy.2001.117377. [DOI] [PubMed] [Google Scholar]
- 175.Chen OT, Dojki FK, Weber SM, et al. Percutaneous Microwave Ablation of an Insulinoma in a Patient with Refractory Symptomatic Hypoglycemia. J Gastrointest Surg. 2015;19:1378–1381. doi: 10.1007/s11605-015-2831-2. [DOI] [PubMed] [Google Scholar]
- 176•.Rossi S, Viera FT, Ghittoni G, et al. Radiofrequency ablation of pancreatic neuroendocrine tumors: a pilot study of feasibility, efficacy, and safety. Pancreas. 2014;43:938–945. doi: 10.1097/MPA.0000000000000133. Study reporting the effectiveness of RFA in controlling growth and hormone excess state in NETs (F-/NF-NETs) [DOI] [PubMed] [Google Scholar]
- 177.Limmer S, Huppert PE, Juette V, et al. Radiofrequency ablation of solitary pancreatic insulinoma in a patient with episodes of severe hypoglycemia. Eur J Gastroenterol Hepatol. 2009;21:1097–1101. doi: 10.1097/meg.0b013e328323d70e. [DOI] [PubMed] [Google Scholar]
- 178.Scott A, Hinwood D, Donnelly R. Radio-frequency ablation for symptom control in a patient with metastatic pancreatic insulinoma. Clin Endocrinol (Oxf) 2002;56:557–559. doi: 10.1046/j.1365-2265.2002.01506.x. [DOI] [PubMed] [Google Scholar]
- 179.MacFarlane MP, Fraker DL, Alexander HR, et al. A prospective study of surgical resection of duodenal and pancreatic gastrinomas in multiple endocrine neoplasia-Type 1. Surgery. 1995;118:973–980. doi: 10.1016/s0039-6060(05)80102-3. [DOI] [PubMed] [Google Scholar]
- 180.Termanini B, Gibril F, Sutliff VE, III, et al. Effect of long-term gastric acid suppressive therapy on serum vitamin B12 levels in patients with Zollinger-Ellison syndrome. Am J Med. 1998;104:422–430. doi: 10.1016/s0002-9343(98)00087-4. [DOI] [PubMed] [Google Scholar]
- 181.Said R, O’Reilly EM, Blumgart L, et al. Pancreatic islet cell carcinoma presenting with concurrent Cushing’s and Zollinger-Ellison syndromes: case series and literature review. Eur J Gastroenterol Hepatol. 2010;22:246–252. doi: 10.1097/MEG.0b013e3283314827. [DOI] [PubMed] [Google Scholar]
- 182.Ellison EC. Zollinger-Ellison syndrome: a personal perspective. Am Surg. 2008;74:563–571. [PubMed] [Google Scholar]
- 183.Eriksson B, Renstrup J, Imam H, et al. High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: Clinical and biological effects. Ann Oncol. 1997;8:1041–1044. doi: 10.1023/a:1008205415035. [DOI] [PubMed] [Google Scholar]
- 184.Case CC, Wirfel K, Vassilopoulou-Sellin R. Vasoactive intestinal polypeptide-secreting tumor (VIPoma) with liver metastases: dramatic and durable symptomatic benefit from hepatic artery embolization, a case report. Med Oncol. 2002;19:181–187. doi: 10.1385/MO:19:3:181. [DOI] [PubMed] [Google Scholar]
- 185.Shaib W, Mitchell K, Saif MW. Amelioration of symptoms and reduction of VIP levels after hepatic artery chemoembolization in a patient with sandostatin resistant VIPoma. Yale J Biol Med. 2010;83:27–33. [PMC free article] [PubMed] [Google Scholar]
- 186•.Abu-Zaid A, Azzam A, Abudan Z, et al. Sporadic pancreatic vasoactive intestinal peptide-producing tumor (VIPoma) in a 47-year-old male. Hematol Oncol Stem Cell Ther. 2014;7:109–115. doi: 10.1016/j.hemonc.2014.03.002. Use of cytoreductive surgery, RFA, embolization and somatostatin analogues to control symptoms in a patient with a VIPoma. [DOI] [PubMed] [Google Scholar]
- 187.Haiqing W, Jiayin Y, Jian Y, et al. Intractable and dramatic diarrhea in liver transplantation recipient with vasoactive intestinal peptide-producing tumor after split liver transplantation: a case report. Transplant Proc. 2015;47:171–173. doi: 10.1016/j.transproceed.2014.07.078. [DOI] [PubMed] [Google Scholar]
- 188.Moug SJ, Leen E, Horgan PG, et al. Radiofrequency ablation has a valuable therapeutic role in metastatic VIPoma. Pancreatology. 2006;6:155–159. doi: 10.1159/000090257. [DOI] [PubMed] [Google Scholar]
- 189.Zhang X, Zhou L, Liu Y, et al. Surgical resection of vasoactive intestinal peptideoma with hepatic metastasis aids symptom palliation: A case report. Exp Ther Med. 2016;11:783–787. doi: 10.3892/etm.2016.3019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Amikura K, Alexander HR, Norton JA, et al. The role of surgery in the management of ACTH-producing islet cell tumors of the pancreas. Surgery. 1995;118:1125–1130. doi: 10.1016/s0039-6060(05)80123-0. [DOI] [PubMed] [Google Scholar]
- 191.Druce M, Rockall A, Grossman AB. Fibrosis and carcinoid syndrome: from causation to future therapy. Nat Rev Endocrinol. 2009;5:276–283. doi: 10.1038/nrendo.2009.51. [DOI] [PubMed] [Google Scholar]
- 192.Cunningham JL, Janson ET, Agarwal S, et al. Tachykinins in endocrine tumors and the carcinoid syndrome. Eur J Endocrinol. 2008;159:275–282. doi: 10.1530/EJE-08-0196. [DOI] [PubMed] [Google Scholar]
- 193.Doppman JL, Nieman L, Miller DL, et al. Ectopic adrenocorticotropic hormone syndrome: localization studies in 28 patients [published erratum appears in Radiology 1989 Oct;173(1):286] Radiology. 1989;172:115–124. doi: 10.1148/radiology.172.1.2544919. [DOI] [PubMed] [Google Scholar]
- 194.Alexandraki KI, Grossman AB. Therapeutic Strategies for the Treatment of Severe Cushing’s Syndrome. Drugs. 2016;76:447–458. doi: 10.1007/s40265-016-0539-6. [DOI] [PubMed] [Google Scholar]
