Pasireotide in the treatment of neuroendocrine tumors: a review of the literature

in Endocrine-Related Cancer
Authors:
Giovanni VitaleDepartment of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano IRCCS, Milan, Italy

Search for other papers by Giovanni Vitale in
Current site
Google Scholar
PubMed
Close
,
Alessandra DicitoreLaboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano IRCCS, Milan, Italy

Search for other papers by Alessandra Dicitore in
Current site
Google Scholar
PubMed
Close
,
Concetta SciammarellaDepartment of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy

Search for other papers by Concetta Sciammarella in
Current site
Google Scholar
PubMed
Close
,
Sergio Di MolfettaDepartment of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy

Search for other papers by Sergio Di Molfetta in
Current site
Google Scholar
PubMed
Close
,
Manila RubinoUnit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, Milan, Italy

Search for other papers by Manila Rubino in
Current site
Google Scholar
PubMed
Close
,
Antongiulio FaggianoDepartment of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy

Search for other papers by Antongiulio Faggiano in
Current site
Google Scholar
PubMed
Close
, and
Annamaria ColaoDepartment of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy

Search for other papers by Annamaria Colao in
Current site
Google Scholar
PubMed
Close
View More View Less

Free access

Somatostatin analogs have an important role in the medical therapy of neuroendocrine tumors (NETs). Octreotide and lanreotide, both somatostatin analogs binding with high affinity for the somatostatin receptor (SSTR)2, can control symptoms in functional NETs. In addition, these compounds, because of their antiproliferative effects, can stabilize growth of well-differentiated NETs. Pasireotide is a novel multireceptor-targeted somatostatin analog with high affinity for SSTR1, 2, 3, and 5. This review provides an overview of the state of the art of pasireotide in the treatment of NETs, with the aim of addressing clinical relevance and future perspectives for this molecule in the management of NETs.

Abstract

Somatostatin analogs have an important role in the medical therapy of neuroendocrine tumors (NETs). Octreotide and lanreotide, both somatostatin analogs binding with high affinity for the somatostatin receptor (SSTR)2, can control symptoms in functional NETs. In addition, these compounds, because of their antiproliferative effects, can stabilize growth of well-differentiated NETs. Pasireotide is a novel multireceptor-targeted somatostatin analog with high affinity for SSTR1, 2, 3, and 5. This review provides an overview of the state of the art of pasireotide in the treatment of NETs, with the aim of addressing clinical relevance and future perspectives for this molecule in the management of NETs.

Introduction

Neuroendocrine tumors (NETs) comprise a heterogeneous group of neoplasms in most cases with a low rate of growth and sometimes able to produce and secrete several hormones that drive forward specific clinical syndromes, leading to significant disability and a negative impact on quality of life. Several therapeutic options for NETs are available and a multidisciplinary approach is needed for a proper management of this disease. The current first-line therapy of these neoplasms is represented by surgery followed by medical management, such as somatostatin analogs (SSAs), mTOR inhibitors, tyrosine kinase inhibitors, chemotherapy and peptide receptor radionuclide therapy. However, new treatment options to further improve survival outcomes are needed (Walenkamp et al. 2014). Somatostatin receptors (SSTRs) are expressed in NETs and SSAs are widely used as medical therapy both in functioning and non-functioning well-differentiated NETs. Octreotide long-acting release and lanreotide autogel are the two first-generation long-acting SSAs currently approved for the treatment of well-differentiated NETs. These SSAs, characterized by a restricted affinity profile for SSTR2 and at lesser extent SSTR5, inhibit hormonal secretion and provide an effective control of hypersecretion symptoms in about 40–60% of patients with functioning NETs (Pavel et al 2017, Mazziotti et al. 2017). In addition, two prospective multicenter randomized clinical trials (PROMID and CLARINET) demonstrated that both SSAs have antiproliferative activity in the management of advanced NETs. Indeed, although SSAs have limited activity in terms of tumor shrinkage, they are able to stabilize tumor proliferation, leading to a significant delay in disease progression (Rinke et al. 2009, Caplin et al. 2014).

Pasireotide is a second-generation multireceptor-targeted SSA with a broader spectrum of affinity with respect to octreotide and lanreotide for different receptor subtypes (SSTR1, 2, 3, 5). Pasireotide is currently approved for the treatment of acromegaly and Cushing’s disease. This paper provides a state-of-the-art overview of pasireotide in the treatment of NETs, with the aim of addressing clinical relevance and future perspectives for this molecule in the management of NETs.

Expression of SSTR subtypes in NETs

Several studies have identified the expression of SSTRs in NET cell lines, primary NETs cell cultures and tissues through a variety of techniques including reverse transcriptase polymerase chain reaction (RT-PCR), in situ hybridization (ISH), immunohistochemistry (IHC) and autoradiography (Table 1).

Table 1

Expression of somatostatin receptor (SSTR) subtypes in neuroendocrine tumors (NETs), evaluated through different methods.

Tumor type SSTR1 (%) SSTR2 (%) SSTR3 (%) SSTR4 (%) SSTR5 (%) Method Reference
Pancreatic NETs (n = 15) 100 100 73 67 RT-PCR Mohamed et al. (2014)
Pancreatic NETs (n = 33) 90.1 84.8 78.8 24.2 42.4 RT-PCR Papotti et al. (2002)
GI NETs (n = 13) 92.3 100 53.8 15.4 76.9 RT-PCR Papotti et al. (2002)
Pancreatic NETs (n = 22) 68.2 36.4 63.6 IHC Papotti et al. (2002)
GI NETs (n = 15) 80 60 80 IHC Papotti et al. (2002)
GEP NETs (n = 17) 47 65 35 ISH Reubi et al. (1994)
GI NETs (n = 25) 68 60 ISH Janson et al. (1996)
Pancreatic NETs (n = 16) 94 (2A) IHC Kimura et al. (1999)
Pancreatic NETs (n = 21) 85.7 61.9 IHC Yerci et al. (2015)
Stomach NETs (n = 8) 100 37.5 IHC Yerci et al. (2015)
Small bowel NETs (n = 10) 70 70 IHC Yerci et al. (2015)
Appendix NETs (n = 7) 85.7 71.5 IHC Yerci et al. (2015)
Rectum NETs (n = 3) 100 66.6 IHC Yerci et al. (2015)
Gastrinoma (n = 33) 30 100 (2A) 79 76 IHC Kulaksiz et al. (2002)
Insulinoma (n = 36) 31 58 (2A) 78 78 IHC Kulaksiz et al. (2002)
Carcinoid tumor (n = 35) 37 86 (2A) 71 83 IHC Kulaksiz et al. (2002)
NETs (n = 52) 73 (2A) 25 IHC Kasajima et al. (2017)
Insulinoma (n = 20) 50 70 20 20 70 RT-PCR Bertherat et al. (2003)
Insulinoma (n = 18) 44 72 44 28 72 Autoradiography Bertherat et al. (2003)
Ileal NETs (n = 27) 52 96 15 4 48 Autoradiography Reubi & Waser (2003)
Insulinoma (n = 27) 61 69 35 4 20 Autoradiography Reubi & Waser (2003)
Gastrinoma (n = 10) 10 100 20 0 33 Autoradiography Reubi & Waser (2003)
Glucagonoma (n = 4) 67 67 33 0 0 Autoradiography Reubi & Waser (2003)
VIPoma (n = 4) 25 100 25 0 25 Autoradiography Reubi & Waser (2003)
Lung carcinoid (n = 29) 71 68 3 0 23 Autoradiography Reubi & Waser (2003)
Typical carcinoid (n = 57) 79 96 (2A), 66 (2B) 49 5 0 IHC Tsuta et al. (2012)
Atypical carcinoid (n = 11) 77 77 (2A), 77 (2B) 33 0 0 IHC Tsuta et al. (2012)
Large cell NEC (n = 21) 60 60 (2A), 30 (2B) 40 0 15 IHC Tsuta et al. (2012)
Small-cell carcinoma (n = 57) 27 69 (2A), 24 (2B) 15 0 0 IHC Tsuta et al. (2012)
Lung carcinoid (n = 119) 63 72 (2A) 20 0 40 IHC Kanakis et al. (2015)
Lung carcinoid (n = 11) 88 88 (2A) 12.5 0 63 RT-PCR Kanakis et al. (2015)
Typical carcinoid (n = 41) 34 (2A) 66 IHC Righi et al. (2010)
Typical carcinoid with metastases (n = 24) 71 (2A) 58 IHC Righi et al. (2010)
Atypical carcinoid (n = 73) 51 (2A) 45 IHC Righi et al. (2010)
Large cell NEC (n = 60) 33 (2A) 33 IHC Righi et al. (2010)
Small-cell carcinoma (n = 61) 38 (2A) 29 IHC Righi et al. (2010)
Pheochromocytoma (n = 6) 83 100 83 83 100 RT-PCR Pasquali et al. (2008)
Pheochromocytoma (n = 39) 95 100 56 54 RT-PCR Saveanu et al. (2011)
Paraganglioma (n = 13) 92 100 31 31 RT-PCR Saveanu et al. (2011)
GEP NETs (n = 35) 100 100 63 89 RT-PCR Saveanu et al. (2011)
Pheochromocytoma (n = 52) 8 25 (2A) 90 10 15 IHC Mundschenk et al. (2003)
Benign Pheochromocytoma (n = 7) 29 29 (2A) 100 0 29 IHC Unger et al. (2008)
Malignant Pheochromocytoma (n = 8) 0 0 (2A) 75 13 38 IHC Unger et al. (2008)

–, not evaluated; GEP, Gastroenteropancreatic; GI, gastrointestinal; IHC, immunohistochemistry; ISH, in situ hybridization; NEC, neuroendocrine carcinoma; RT-PCR, reverse transcriptase polymerase chain reaction.

In most of these studies, SSTR mRNA expression was evaluated by RT-PCR. The characterization of pancreatic NET primary cultures from 15 patients showed expression of SSTR1 and SSTR2 mRNAs in all cases, with a higher SSTR2 expression level compared to SSTR1 in 80% of cases. SSTR3 and SSTR5 mRNAs were expressed in 73 and 67% of cases, respectively. A different expression of SSTR5 was observed in relation to the WHO histological grade of the tumor (100% in grade 1, only 37% in grade 2 tumors) (Mohamed et al. 2014). Also, in another series of pancreatic and gastrointestinal NETs, SSTR1 and SSTR2 mRNA were the most commonly detected subtypes (over 80%), followed by SSTR3 and SSTR5. Poorly differentiated NETs showed a reduced SSTR expression (Papotti et al. 2002). In pancreatic NET cell lines (BON-1 and QGP-1), mRNA levels of SSTRs were expressed in the following order: SSTR5 > SSTR1 > SSTR2 > SSTR3, but in QGP-1 cells, the expression of SSTR3 was not detectable (van Adrichem et al. 2016). ISH confirmed the expression of SSTR1 and SSTR2 mRNA in about 50–70% of gastroenteropancreatic (GEP) NETs (Reubi et al. 1994, Janson et al. 1996).

IHC and autoradiography confirmed that SSTRs 1, 2 and 5 were the most frequent receptors in GEP NETs, whereas the expression of SSTR3 was quite variable (Kimura et al. 1999, Kulaksiz et al. 2002, Papotti et al. 2002, Bertherat et al. 2003, Reubi & Waser 2003, Yerci et al. 2015, Kasajima et al. 2017).

In lung NETs, SSTR1 and SSTR2 were the most expressed subtypes, followed by SSTR3. The expression of SSTR5 varied from 0 to 63% of tumors and SSTR4 resulted to be rarely expressed. SSTRs showed a decreased protein expression from well to poorly differentiated pulmonary neuroendocrine carcinomas (Reubi & Waser 2003, Righi et al. 2010, Tsuta et al. 2012, Kanakis et al. 2015).

In primary pheochromocytoma, cell cultures and tissues SSTRs were expressed at mRNA level with a prevalent expression of SSTR2 and SSTR5 (Pasquali et al. 2008). Also, in another paper, SSTR2 mRNA was the most frequently detected subtype in pheochromocytoma and paraganglioma tissues (SSTR2 > SSTR1 > SSTR3 and SSTR5) (Saveanu et al. 2011). Other studies found at IHC that SSTR3 was the predominant receptor subtype in both malignant and benign pheochromocytomas and SSTR2A was expressed in less than one-third of pheochromocytoma and preferentially in benign lesions (Mundschenk et al. 2003, Unger et al. 2008).

Although the highly variable expression of SSTR subtypes reported in these studies and depending on the methodology adopted, the majority of NETs expresses several SSTRs subtypes. This has paved the way to the development of new SSTR panligands, such as pasireotide, which display a broader receptor-binding profile compared with octreotide and lanreotide.

After activation, SSTRs modulate different intracellular effectors, mediating antisecretory and antiproliferative effects of SSAs in NETs (Fig. 1). In particular, antisecretory activity is exerted via decrease of cAMP levels and calcium channel activity. The activation of phosphotyrosine phosphatases, such as SRC homology phosphatase type 1 and type 2, density-enhanced protein-tyrosine phosphatase-1, and the modulation of mitogen-activated protein kinase (MAPK) activity are mainly involved in antiproliferative effects of SSTRs. Both adenylyl cyclase inhibition and phosphotyrosine phosphatases activation are induced by all SSTRs, while MAPK activity depends on SSTR subtype since it is increased by SSTR4, decreased by SSTR3 and 5 and modulated in both directions by SSTR1 and 2. Indeed, different conformations of the receptor–SSA complex are the reason of the selective activation of different pathways and of the modulation of receptor endocytosis (Barbieri et al. 2014).

Figure 1
Figure 1

Schematic representation of the intracellular signaling pathways modulated by pasireotide and other drugs involved in combined treatments. AC: adenylyl cyclase; Akt, Protein kinase B; ASCL1, achaete-scute homolog like 1; Bax, bcl2-associated X; CAB, cabergoline; CAF, cancer-associated fibroblast; cAMP, cyclic adenosine monophosphate; Ca2+, calcium ion; COX, cyclooxygenase 2; CX, celecoxib; DR2, dopamine receptor 2; 4E-BP1, eukaryotic translation initiation factor 4E-binding protein 1; eNOS, endothelial nitric oxide synthase; ERK1/2, extracellular signal–regulated kinase 1/2; EV, everolimus; GEM, gemcitabine; IGF2, insulin-like growth factor 2; IL 6, interleukin 6; IRA, insulin receptor isoform A; MEK1/2, mitogen-activated protein kinase kinase 1/2; mTOR, mammalian target of rapamycin; PAS, pasireotide; PI3K, phosphoinositide 3 kinase; RAF; RAS; RTK, receptor tyrosine kinase; SHP-1, -2, Src homology phosphatase type 1, 2; SSTR1-5, somatostatin receptor 1-5; TER, teriflunomide; VEGF, vascular endothelial growth factor.

Citation: Endocrine-Related Cancer 25, 6; 10.1530/ERC-18-0010

Pasireotide as a single-agent treatment in NETs

Preclinical studies

In 2004, Schmid & Schoeffter showed that pasireotide inhibits forskolin-stimulated cAMP accumulation with similar efficacy of somatostatin (SRIF-14) in CCL39 cells (Chinese hamster lung fibroblasts), stably transfected with the human SSTR1 gene, and in CHOK1 cells (Chinese hamster ovary), stably transfected with the human SSTR2, SSTR3 and SSTR5 genes. This effect was more potent than that reported with octreotide (Schmid & Schoeffter 2004).

Several further studies described in vitro a significant antitumor activity of pasireotide in NET cells. Ono et al. reported a more potent antiproliferative effect and decrease in Ki-67 after incubation of NCI-H727 cells, a bronchial NET cell line, with pasireotide compared to the untreated control and the conventional octreotide (Ono et al. 2007). van Adrichem et al. showed that pasireotide statistically inhibited bioactivity of insulin receptor isoforms A (IRA) in BON-1 cells, evaluated by KIRA assay, while no effect was observed during incubation with octreotide (van Adrichem et al. 2016). In primary pheochromocytoma cell cultures, both octreotide and pasireotide induced a significant reduction in cell viability and catecholamine production and an increase in apoptotic cells. These effects were more potent with pasireotide than with octreotide (Pasquali et al. 2008). In pancreatic NET primary cell cultures, octreotide and pasireotide similarly reduced cell proliferation through induction of apoptosis, decreased chromogranin A (CgA) secretion in a dose-dependent manner and reduced cAMP levels. Interestingly, a different SSTR2 trafficking was observed during incubation of NET cells with octreotide and pasireotide. After 5 min octreotide induced the SSTR2 internalization, maintained in the cytosol up to 30 min, this trafficking was associated with phosphorylation on Ser341/343 of SSTR2. Pasireotide induced a rapid and transient internalization of SSTR2. After 5 min of incubation with pasireotide, SSTR2 was internalized, but after 15 min, it was observed at the cell surface and no phosphorylation on Ser341/343 of SSTR2 was detected. The rapid recycling of the SSTR2 and the lack of Ser phosphorylation, crucial for the SSTR2 desensitization, underlined that the level of active receptors at the cell surface was greater with pasireotide than with octreotide. These data suggest a possible benefit in the long-term treatment with pasireotide (Mohamed et al. 2014).

In a mouse model of multiple endocrine neoplasia type 1 (MEN1), where mice deleted for an allele of the Men1 gene (Men1+/−) develop pancreatic NETS (positive for insulin) and pituitary adenomas (predominantly prolactinomas but also somatotrophinomas and non-functioning adenomas), the antitumor activity of pasireotide has been evaluated. In these animals, pancreatic NETs expressed SSTR1, 2 and 5. Treatment of Men1+/− mice with pasireotide decreased proliferation in pancreatic and pituitary NETs through induction in apoptosis. Men1+/− mice treated with pasireotide showed an increased survival. In addition, pasireotide decreased development of pancreatic NETs and size of pituitary adenomas. These results suggest that pasireotide may have a chemopreventive role in the treatment of MEN1-associated NETs (Walls et al. 2016).

In another MEN1 transgenic mouse model adopting pancreas-specific promoter (Pdx1) in combination with Cre-Lox system to knockout Men1 in pancreas of mice (Pdx1-Cre:Men1 floxed/floxed) and able to develop insulinoma, pasireotide decreased insulin levels and improved serum glucose levels compared to the untreated control group. In addition, pasireotide stimulated apoptosis and reduced the tumor size of insulinoma (Quinn et al. 2012).

Clinical studies

Octreotide and lanreotide inhibit the release of bioactive hormones and significantly improve diarrhea and flushing in most of patients with the carcinoid syndrome (Ruszniewski et al. 2004, Modlin et al. 2010). However, long-term use of these agents is limited by tachyphylaxis, occurring in a considerable number of patients within 1 year after beginning of treatment (Hofland & Lamberts 2003). Pasireotide has been tested as a monotherapy for the management of diarrhea and flushing in patients with carcinoid syndrome resistant to first-generation SSAs at conventional doses. In a phase II, open-label, multicenter, prospective single-arm study (Kvols et al. 2012), 44 patients with advanced NETs of the gastrointestinal tract and poorly controlled carcinoid syndrome during therapy with octreotide LAR received s.c. pasireotide up to 1200 µg twice daily. Levels of tumor markers (urinary 5-hydroxyindole acetic acid and serum chromogranin A) rapidly decreased after pasireotide initiation and remained low. In 12 (27%) of these patients, pasireotide achieved at least partial control of symptoms when administered at doses of 600–900 mg twice daily for 15 consecutive days. A similar proportion of patients (20.9%) achieved symptom control after 6 months of pasireotide treatment in a phase III multicenter study (Wolin et al. 2015) randomizing patients with metastatic gastrointestinal NETs and inadequately controlled carcinoid symptoms under first-generation SSAs (octreotide LAR 30 mg/28 days, octreotide s.c. 600 μg/day, lanreotide autogel 120 mg/28 days or lanreotide SR 30 mg/14 days). Subjects were randomized to receive pasireotide LAR 60 mg or high-dose octreotide LAR (40 mg every 28 days). However, this study was halted before completion due to a low predictive probability of showing differences in symptom control between treatment arms (odds ratio (OR) 0.73; 95% confidence interval (CI), 0.27–1.97; P = 0.53).

Limited evidence suggests that pasireotide therapy may be beneficial in preventing recurrent hypoglycemia due to insulinoma. Indeed, pasireotide LAR 40 mg/28 days significantly reduced hypoglycemic events in a patient with a G2 stage IV insulinoma who had undergone excision of the primary tumor and of liver metastases without achieving adequate glycemic control (Tirosh et al. 2016). Since both the pancreatic lesion and the liver metastases had a negative staining for SSTR5, the anti-hypoglycemic effect may come from inhibition of insulin secretion through SSTR5 activation on normal pancreatic beta-cells. Hendren et al. also described the case of a subject with malignant insulinoma and severe hypoglycemic episodes despite the use of diazoxide and octreotide LAR. Treatment with pasireotide LAR induced a near resolution of hypoglycemic events (Hendren et al. 2018). However, further investigation and, eventually, a phase III controlled trial for interventions would be needed to validate the use of pasireotide as a treatment in patients with advanced metastatic insulinoma and symptomatic hypoglycemia.

First-generation SSAs have demonstrated antitumor activity in patients with advanced well-differentiated NETs of the gastroenteropancreatic tract with or without secretory symptoms (Rinke et al. 2009, Caplin et al. 2014). Due to its wider binding profile, pasireotide should theoretically represent a more powerful antiproliferative agent; however, data supporting its role in tumor control for patients with NETs are still scarce. In patients with metastatic NETs of the digestive tract and carcinoid symptoms refractory to available SSAs at conventional doses, Wolin et al. found an improved, though not statistically significant, tumor control rate at 6 months among patients receiving pasireotide LAR (60 mg every 4 weeks) compared to the octreotide arm (40 mg every 4 weeks) (62.7% vs 46.2%, OR 1.96; 95% CI, 0.89–4.32; P = 0.09). In a post hoc analysis, patients on pasireotide LAR had a 5-month longer progression-free survival (PFS) than patients on octreotide LAR (11.8 months vs 6.8 months), corresponding to a 54% reduction in estimated risk for disease progression or death (HR 0.46; 95% CI, 0.20–0.98; P = 0.045) (Wolin et al. 2015). Data from a small, open-label, phase II study (Cives et al. 2015) of pasireotide LAR (60 mg every 4 weeks) have also shown promising antitumor efficacy with mean PFS of 11 months (95% CI, 7.6–16) in patients with locally unresectable or metastatic NET without prior systemic therapy, including octreotide or lanreotide. A greater effect was observed in patients with low-grade midgut carcinoids, low (<10%) hepatic tumor burden, normal baseline chromogranin A and high tumoral SSTR5 expression. When best response to therapy was evaluated using the RECIST criteria, 64% (18/28) of patients had at least stable disease with 13 patients showing some degree of tumor shrinkage. A recent phase I study evaluated the maximum tolerated dose and efficacy of pasireotide of long-acting pasireotide in patients with advanced NETs and documented disease progression. A total of 29 patients were treated with 80 mg (n = 13) and 120 mg/28 days (n = 16). Two partial responses were observed in the 16 patients receiving the 120 mg dose of pasireotide. Most patients (>75%) had stable disease treated with both doses. In addition, the median PFS (95% CI) was 8.3 months and 10.8 months in the 80-mg dose group and 120-mg dose group, respectively (Yao et al. 2017).

Long-acting intramuscular formulation of pasireotide is well tolerated as single therapy in patients with advanced NETs (Wolin et al. 2013). The safety profile of pasireotide includes gastrointestinal symptoms (such as nausea, diarrhea and abdominal pain) and fatigue, which are largely consistent with first-generation SSAs. However, compared with other SSAs, pasireotide therapy is associated with a higher frequency of hyperglycemia often requiring start/adjustment of antidiabetic drugs (Wolin et al. 2013). Increased fasting blood glucose is more evident in patients with a history of impaired fasting glucose or diabetes mellitus (Kvols et al. 2012). In a phase III study, hyperglycemia was observed in 15/53 (28.3%) and 3/42 (5.3%) patients treated with pasireotide LAR (60 mg) and octreotide LAR (40 mg) every 28 days, respectively. The rate of grade 3 or 4 hyperglycemia was higher with pasireotide LAR (9.4%) than with octreotide LAR (1.8%) (Wolin et al. 2013). Pasireotide-associated hyperglycemia seems to be mainly related to a marked suppression in insulin and incretins (GIP and GLP-1) secretion without any alteration in hepatic and peripheral insulin sensitivity. All five SSTR subtypes are variably expressed in human pancreatic islets, with a preferential expression of SSTR1 and 5 in beta-cells, and of SSTR2 in alpha-cells (Kumar et al. 1999). The different expression of SSTR2 and SSTR5 on pancreatic alpha- and beta-cells is probably the cause of a greater decrease of insulin secretion and lesser suppression of glucagon with pasireotide when compared to octreotide or lanreotide (Henry et al. 2013). A recent study defined the maximum tolerated dose for pasireotide LAR at 120 mg/28 days; at this dose, a high incidence of bradycardia (31%) was observed (Yao et al. 2017).

Pasireotide as a combined treatment in NETs

The development and progression of NETs depend on the activity of complex signaling pathways which have been also described in other tumors (Walenkamp et al. 2014). Combination therapy represents a cornerstone of NET therapy, as a result of targeting several key pathways in a characteristically synergistic/additive manner. In addition, this approach potentially overcomes or delays the development of drug resistance. It has been reported that addition of SSAs to chemotherapy or biologic therapy may increase treatment efficacy (Wolin 2012). RADIANT-1 phase II trial reported a better median PFS in patients with pancreatic NETs treated with everolimus plus octreotide LAR (16.7 months) compared to that observed after everolimus alone (9.7 months) (Yao et al. 2010). On the basis of several preclinical and clinical trials, pasireotide may be effective not only as a single agent but also combined to standard or new compounds in the management of NETs (Fig. 1).

Pasireotide plus everolimus

Since several clinical trials together with in vitro and in vivo studies demonstrated the efficacy of targeting mTOR or SSTR in NET therapy, combined treatment with SSAs and everolimus has been considered as a potential therapeutic tool for NETs (Bousquet et al. 2012, Oberstein & Saif 2012). Surprisingly, a synergistic or additive in vitro inhibition of the PI3K/AKT/mTOR pathway through SSAs (octreotide and pasireotide) and everolimus combination was not always evident in NET cells, although each molecule independently suppressed this pathway. Indeed, there was not a favorable antitumor activity after incubation of NET cell lines (KRJ-I, P-STS, NCI-H727 and BON-1) with mTOR inhibitors and octreotide. This may be due to feedback mechanisms within the PI3K/AKT/mTOR pathway, such as the activation of AKT or cross-activation of tumor escape pathways (ERK/pERK) (Moreno et al. 2008, Svejda et al. 2011). Zatelli et al. showed that the combination pasireotide plus everolimus was unable to potentiate both antiproliferative and antisecretory effects of everolimus in primary cells from human bronchial carcinoid (Zatelli et al. 2010). Similar underwhelming results were reported for primary cultures of human pancreatic NETs. Everolimus plus pasireotide was not able to decrease either cell viability or chromogranin A secretion compared to single drugs. Interestingly, pasireotide alone induced apoptotic processes (through caspase activation), that were reverted by combined treatment, probably due to the activation of the survival pathways, such as AKT, induced by everolimus (Mohamed et al. 2017).

A phase I study evaluated the safety and feasibility of combining pasireotide and everolimus in 21 patients with locally unresectable or metastatic NETs of low (77%) or intermediate histological grade (23%). Patients were treated with escalating doses of pasireotide (600–1200 µg s.c. twice daily, followed by pasireotide LAR 40–60 mg i.m. monthly) and everolimus (5–10 mg daily). Hyperglycemia was one of the most commonly observed side effects (90% of cases), of grade 3 or 4 in 8 out of 21 patients. Other grade 3–4 side effects were uncommon (mucositis, diarrhea, rash, joint pain, prolonged QTc interval, hypophosphatemia, thrombocytopenia and leucopenia). In terms of efficacy, the best responses were stable disease in 19 patients (90%) and partial response in one patient (5%), evaluated according to RECIST criteria. No differences were observed according to primary tumor site (Chan et al. 2012).

COOPERATE-2 was a randomized phase II trial in which 160 patients with advanced, progressive grade 1–2 pancreatic NETs were treated with the combination of everolimus (10 mg daily) and pasireotide (60 mg/28 days) vs everolimus alone. The median PFS was similar in both arms, 16.8 months (12.1–19.6) in combination arm vs 16.6 months (11.1–19.5) in everolimus arm (HR 0.991 95% CI, 0.64–1.54), although partial response rate was significantly higher in combination arm (20%) compared to that with everolimus alone (6%). No difference in the overall survival was observed, with a median follow-up of 22.6 months. Higher incidence of hyperglycemia (all grades 76% vs 27%) and diabetes mellitus (26% vs 7%) was noted in the combination arm compared to everolimus alone, while rates of stomatitis, diarrhea, abdominal pain, pneumonitis and cardiac events were similar between both groups (Kulke et al. 2017). The combined treatment determined a higher grade of side effects, particularly for hyperglycemia, diarrhea and weight loss. The less favorable toxicity profile was responsible for a higher frequency of dose reduction and therapy discontinuation in patients treated with both compounds that may have an impact on PFS. Moreover, previous studies showed that, even though pasireotide binds with different affinity all the subtypes of SSA receptors, it exerts a different effect on SST2 internalization, as previously described (Mohamed et al. 2014). If this different mechanism exerts a role on pasireotide antitumor activity and tolerability, it should be better clarified.

A recent prospective, randomized and phase II trial study (LUNA trial) assessed the efficacy and safety of pasireotide (60 mg/28 days i.m.) and/or everolimus (10 mg daily) in 124 patients with unresectable or metastatic, well-differentiated lung (typical or atypical carcinoid) and thymus NETs. The analysis of relative change of tumor size from baseline showed 11/36 (31%) patients in the pasireotide group, 16/33 (49%) in the everolimus group and 24/33 (73%) in the combination group experienced some degree of tumor shrinkage, but only one patient in each group reached a partial response (2.4%). In the other cases, the tumor shrinkage was not a RECIST response. Median PFS was 8.5 months in the pasireotide group, 12.5 months in the everolimus group and 11.8 months in the combination group. The most common side effects reported during treatment with pasireotide (alone and in combination) were diarrhea, hyperglycemia and weight loss. Patients treated with everolimus (alone and in combination) experienced above all stomatitis and diarrhea. In combination group, there was a higher rate of adverse events, particularly for hyperglycemia, diarrhea, weight loss and asthenia (Ferolla et al. 2017).

Pasireotide plus cabergoline

Another potential target for the treatment of NETs is the dopaminergic system. SSTR and dopamine receptors type 2 (DR2) are frequently co-expressed in NETs and can heterodimerize, enhancing functional activity and exerting a synergistic inhibitory effect (Gatto & Hofland 2011). Since somatostatin and dopamine analogs could inhibit the secretion of several hormones, in particular IGF1 and IGF2 targeting indirectly IRA activation, van Adrichem et al. evaluated the possibility to combine SSAs plus cabergoline, a dopamine agonist, in human pancreatic NET cell lines (BON-1 and QGP-1). These cells highly expressed both IGF2 and IRA. IGF system has a tumor-promoting role and mitogenic effects in NETs and include IGF1, IGF2, IGF receptor (IGFR1 and 2), IGF-binding proteins, IRA and insulin receptor isoform B (IRB). In human pancreatic NET cell lines, pasireotide combined to cabergoline significantly decreased the IGF2 mRNA expression; no change on mRNA expression was observed during incubation with each compound alone or octreotide plus cabergoline. In addition, a stronger inhibition in IRA bioactivity has been observed with pasireotide plus cabergoline than each drug alone. These encouraging data showed that activation of SSTR2 and 5 together with DR2 could inhibit IRA activation induced by IGF2, and combined treatment with pasireotide and cabergoline could be an innovative approach to the treatment of NETs (van Adrichem et al. 2016). Interestingly, the heterodimerization property of these receptors has been recently used to develop several chimeric compounds that bound and synergistically activated both SSTR and DR2, showing significant antitumor effects, at least in vitro, in NETs (Culler 2011, Zitzmann et al. 2013).

Pasireotide plus teriflunomide

Among several signaling pathways involved in tumor progression, RAF1/mitogen-activated protein kinase kinase (MEK)/ERK1/2 pathway has an important role in the regulation of NET cell proliferation (Carter et al. 2013). Several in vitro studies in NET cells demonstrated that not only RAF and MEK inhibitors (Zitzmann et al. 2010, Iida et al. 2012) but also RAF1 activators (Cook et al. 2010, Fazio et al. 2014) showed antitumor effects in NETs. In addition, phosphorylation of ERK1/2 seemed to be important for the activity of SSAs, probably because of its role in the decrease of transcripts for Ki67 and in the induction of cell cycle arrest through p21 increase (Kidd et al. 2008). On the light of this evidence, Somnay et al. evaluated the antitumor activity of pasireotide in combination with teriflunomide, a Raf activator. The antiproliferative effect of pasireotide at low dose was improved by its association with teriflunomide in BON cells. Moreover, this combination was synergistic and able to induce a decrease in the expression of chromogranin A and ASCL1, both neuroendocrine biomarkers, and an increase of ERK1/2 phosphorylation. Finally, teriflunomide plus pasireotide increased the activity of proapoptotic enzymes, PARP and caspase-3, reducing at the same time the expression of some anti-apoptotic proteins (XIAP and Mcl-1) compared to either drug alone. These data demonstrated that combined therapy with these two drugs could represent a promising therapeutic option for NETs (Somnay et al. 2013).

Pasireotide plus aurora

Georgieva et al. proposed a new therapeutic approach to gastroenteropancreatic NETs using the aurora kinase B inhibitor, ZM447439, as antitumoral option in monotherapy or in combination with other standard chemotherapeutical drugs, including pasireotide. Unfortunately, pasireotide as a single agent showed a higher antiproliferative activity compared to combination with ZM447439 in BON cell line (Georgieva et al. 2010).

Ongoing clinical trials

Several other trials are currently investigating the role of pasireotide alone, or in combination with everolimus and/or loco-regional therapy in NETs. The objective of these studies is to clarify the efficacy of pasireotide in terms of safety, symptomatic and biochemical control, and effects on tumor mass and survival in NETs. We have found 8 ongoing clinical trials with this topic, listed at the www.clinicaltrial.gov website (Table 2).

Table 2

Ongoing clinical trials in neuroendocrine tumors (NETs), as listed at the www.clinicaltrial.gov website, using pasireotide as search criteria.

Study Treatment Study design Inclusion criteria Primary endpoint Study ID
Study of pasireotide LAR in patients with metastatic NETs Pasireotide Phase 2 NETs Progression-free survival Nbib1253161
An open-label, multicenter, single-arm study of pasireotide LAR in patients with rare tumors of neuroendocrine origin Pasireotide Phase 2 Pancreatic NETs, pituitary adenoma, Nelson syndrome, ectopic ACTH syndrome Percentage of responders at month 6 Nbib958841
Open-label study to test the efficacy and safety of pasireotide in patients with ectopic ACTH-producing tumors Pasireotide Phase 2 Ectopic-ACTH secreting tumors Normalization of 24 h urine free cortisol Nbib2780882
A phase I of pasireotide s.c. followed by pasireotide LAR in patients with metastatic melanoma or metastatic Merkel cell carcinoma Pasireotide Phase 1 Metastatic melanoma, Merkel cell carcinoma Safety and tolerability Nbib1652547
Extension Study to the COOPERATE-1 Study Pasireotide

Everolimus
Phase 1 NETs Safety and tolerability Nbib1590199
Pasireotide, everolimus and selective internal radioembolization therapy for unresectable hepatic metastases Pasireotide

Everolimus

Radioembolization
Phase 1 NETs Safety Nbib1469572
A 3-arm randomized phase II trial evaluating single agent and combined efficacy of pasireotide and everolimus in adult patients with radioiodine-refractory differentiated and medullary thyroid cancer Pasireotide

Everolimus
Phase 2 Radioiodine-refractory differentiated thyroid cancer, medullary thyroid cancer Response rate Nbib1270321
SOM230 alone or in combination with RAD001 in patients with medullary thyroid cancer Pasireotide

Everolimus
Phase 2 Medullary thyroid cancer Progression-free survival Nbib1625520

A multi-institutional, prospective phase II open-label trial has the goal to clarify if pasireotide LAR 60 mg/28 days, as first-line therapy, can shrink or slow the growth of locally unresectable or metastatic carcinoid or pancreatic NETs. Patients with poorly differentiated neuroendocrine carcinomas or small-cell carcinomas and patients treated with prior systemic antineoplastic drugs, including SSAs, are excluded (Nbib1253161, www.clinicaltrial.gov). Another phase II study is assessing the efficacy and safety of pasireotide in patients with rare tumors of neuroendocrine origin (Nelson syndrome, pancreatic NETs, pituitary and ectopic-ACTH secreting tumor), analyzing the number of patients attaining normalization or more than 50% reduction in primary biochemical tumor marker (Nbib958841, www.clinicaltrial.gov).

A phase II study is evaluating the effect of pasireotide on cortisol secretion in non-pituitary ectopic-ACTH secreting tumor. The purpose of this study is to investigate the efficacy of pasireotide (initial dose of 600 μg twice daily for one month, increased to 900 μg twice during the treatment period of 6 months) for normalizing 24-h urine-free cortisol in patients with ectopic ACTH-producing tumors. Salivary and plasmatic cortisol, clinical signs and symptoms and safety will also be evaluated (Nbib2780882, www.clinicaltrial.gov).

A phase I study is currently evaluating the safety, pharmacokinetics and efficacy of pasireotide s.c. and LAR in patients with Merkel cell carcinoma, an aggressive neuroendocrine neoplasm of the skin (Nbib1652547, www.clinicaltrial.gov).

The extension phase of COOPERATE-1 could provide more information about the toxicities and, as secondary endpoints, about efficacy of the combination of pasireotide and everolimus in the long term (Nbib1590199, www.clinicaltrial.gov). Another phase I study is investigating the safety and activity of pasireotide, everolimus and hepatic selective internal radioembolization therapy in patients with pancreatic and intestinal NETs, with unresectable liver metastasis, of low-intermediate grade (Nbib1469572, www.clinicaltrial.gov). Moreover, two studies are investigating for the first time the efficacy of the combination of everolimus and pasireotide in medullary thyroid cancer in terms of response rate and PFS (Nbib1270321, Nbib1625520, www.clinicaltrial.gov).

Pasireotide as a combined treatment in non-endocrine tumors

Beside NETs, SSTRs are aberrantly expressed in other tumors. Few preclinical and clinical studies evaluated the effects of pasireotide in combination with several drugs in non-endocrine tumors, suggesting a potential therapeutic use of these new combinations in NETs (Fig. 1).

Pasireotide plus chemotherapy

In several tumors, stromal cancer–associated fibroblasts (CAF) have a main role in cancer progression and in the development of chemoresistance via the secretion of growth factors and immune suppressive cytokines, mesenchymal–epithelial cell interactions and generation of specific niches within the tumor microenvironment that facilitate the acquisition of drug resistance (Wang et al. 2017). In pancreatic adenocarcinoma, pasireotide showed a significant antitumor activity through an effect on CAF (Moatassim-Billah et al. 2016). Exclusively, pasireotide, and not octreotide, was able to inhibit in vitro CAF tumorigenic features. CAF secretome, able to stimulate in vitro cancer cell survival, migration and invasive features, was abolished after incubation with pasireotide. This effect was mediated by the activation of SSTR1 and shutdown of the protein synthesis via 4E-BP1 in CAF. In mice orthotopically co-xenografted with the human pancreatic cancer MiaPaCa-2 cells and CAF, pasireotide was not able to inhibit tumor growth but abolished the formation of metastasis (Moatassim-Billah et al. 2016). In the light of these considerations and taking into account the implication of tumor microenvironment in chemoresistance, a promising pharmacological strategy has recently been suggested, combining pasireotide with gemcitabine, a chemotherapeutic drug used in the treatment of pancreatic adenocarcinoma. In nude mice xenografted with pancreatic cancer cells and CAF, gemcitabine and pasireotide decreased tumor growth with a dramatic increase in cancer cell apoptosis. The activation of SSTR1 by pasireotide on CAF inhibited the mTOR/4E-BP1 pathway and the secretion of chemoprotective and soluble growth factors, cytokines and/or chemokines, including interleukin 6 (IL 6), with a relevant role in the induction of chemoresistance (Duluc et al. 2015).

Combination of pasireotide and gemcitabine has manageable safety profile, as demonstrated in a phase I study enrolling patients with metastatic or locally advanced pancreatic adenocarcinoma. Patients were treated with gemcitabine 1000 mg/m2 weekly, for 3 weeks every months and pasireotide LAR 40 or 60 mg every 28 days. Preliminary signals of activity were reported during this trial with a disease control rate of 68% (stable disease in 9/16 patients, partial response in 2/16 patients), with a median PFS of 4.1 months (range 1–16 months) and a median overall survival of 6.9 months (range 1–25 months) (Suleiman et al. 2015). Similar results were reported in a dose-escalation, open-label, phase I trial evaluating the combination of pasireotide and standard FOLFIRI (5-fluorouracil, leucovorin and irinotecan) regimen in 16 patients with advanced gastrointestinal malignancies. Treatment was well tolerated, 2 patients achieved partial response (colon cancer and esophageal cancer) and 7 patients had stable disease. Median PFS was 3.6 months (95% CI: 2.4–5.5 months) and median survival was 7.2 months (95% CI: 4.3–11.4 months) (Mahipal et al. 2015).

In the literature, there are no studies evaluating the effects of pasireotide on CAF in NETs. However, the relevant role of tumor microenvironment and the interplay between tumor cells and fibroblasts in the development and progression of NETs and probably in the induction of drug resistance is well known. In addition, in this tumor fibrotic complication (carcinoid heart disease, mesenteric/retroperitoneal fibrosis, etc.) may lead to devastating clinical sequelae and affect prognosis (Laskaratos et al. 2017).

Pasireotide plus cyclooxygenase-2 inhibitors

The cyclooxygenase (COX) enzymes catalyze a key step in the synthesis of prostanoids, relevant mediator of inflammation and angiogenesis. COX-2 is overexpressed in cancer cells and is implicated in carcinogenesis, tumor progression and resistance to chemo- and radiotherapy. Somatostatin-14 inhibited COX-2 expression in colon cancer cells through SSTR3 and 5 activation and inhibited cell proliferation (Colucci et al. 2008). Interestingly, the combination of pasireotide and celecoxib, a selective COX-2 inhibitor, synergistically inhibited cell growth in hepatocellular carcinoma cells (HepG-2 cell line) through apoptosis induction (Xie et al. 2011). Furthermore, the combination treatment significantly prolonged survival of nude mice with HepG2 xenografts and down-regulated vascular endothelial growth factor expression (Xie et al. 2011). These results are potentially applicable in NETs. Indeed, several studies suggested a role of COX-2 in the pathogenesis and progression of NETs. COX-2 is expressed in pheochromocytoma tissue (Salmenkivi et al. 2001) and up-regulated in gastrointestinal and bronchopulmonary carcinoids (Mizuno et al. 2006). Moreover, COX-2 up-regulates both expression and bioactivity of chromogranin A in a cell line of rat pheochromocytoma (PC12) (Connolly et al. 2007). In a cohort of 247 patients with histologically confirmed primary or metastatic GEP-NET, COX-2 overexpression was found to be associated with poor differentiation, higher stage and lymphatic invasion and to be inversely correlated with SSTR2 expression. Kaplan–Meier analyses also showed COX-2 overexpression being associated with poor overall survival (Kim et al. 2011). These data suggest that COX-2 could be a therapeutic target in a selected subset of NETs, with considerable potential for future therapy combining selective COX-2 inhibitors with pasireotide.

Conclusions and future perspectives

SSAs represent the cornerstone of therapy for patients with advanced G1-G2 NETs not eligible for surgery. Although a preliminary study supported a role of pasireotide in the therapy of patients with metastatic carcinoid syndrome refractory to conventional SSAs (Kvols et al. 2012), a phase III study showed that pasireotide LAR and high-dose octreotide LAR have similar efficacy for symptom control in patients with functional NET and symptoms inadequately controlled with the recommended doses of available SSAs (Wolin et al. 2015). In addition, it is still unknown whether pasireotide has greater antiproliferative effects than octreotide/lanreotide, particularly in medically naïve patients with NETs. After 6 months of treatment, a trend toward higher tumor control rate was observed with pasireotide LAR (60 mg/every 4 weeks) in NETs, although it was statistically not significant (Wolin et al. 2015).

The efficacy of combination therapy with pasireotide plus everolimus in NETs is also controversial. Studies investigating the combination of pasireotide and everolimus showed a higher response rate, in terms of tumor stabilization and tumor regression, but this better response rate was not able to determine a statistically significant benefit in PFS compared to each drug alone.

A potential limitation in the majority of previous studies was the heterogeneous cohort of enrolled patients with unknown SSTRs profile. This information could be relevant for the selection of patients responsive to pasireotide. Indeed, Cives et al. reported that high SSTR5 expression in NETs was associated with prolonged PFS in a small series of patients treated with pasireotide LAR (Cives et al. 2015). Therefore, the antitumor activity of pasireotide LAR warrants further exploration in adequately powered prospective phase III studies with symptom control, PFS and additional tumor control metrics as predefined endpoints and after a complete characterization of SSTR subtypes in NETs. This may identify subgroups of patients with greater rates of symptomatic, biochemical and radiological objective responses to pasireotide than to the currently available SSAs. Future trials should also investigate the potential effects of high-dose treatment with pasireotide in NETs. In fact, a phase I study with pasireotide at high dose (120 mg every 4 weeks) showed encouraging antitumor activity (Yao et al. 2017). Moreover, an association between pasireotide and other targeted therapy used in NETs, such as sunitinib or lenvatinib, should be explored. This treatment had a different toxicity profile from everolimus, and the association could be better tolerated.

In conclusion, without any evidence of a more potent antitumor effect and due to the less favorable toxicity profile of pasireotide, this drug is not currently used routinely as therapy in advanced NETs. However, we cannot exclude a role of pasireotide restricted to a subset of NETs with a specific SSTR profile. Recent preclinical studies demonstrated a potent antitumor activity of pasireotide in combination with dopamine agonists, Raf activators, chemotherapy and COX-2 inhibitors in several cancer models. Future studies will need to confirm these issues to improve the treatment of advanced NETs.

Declaration of interest

A Colao and A Faggiano have received consultant fees from Novartis and Ipsen. G Vitale has received consultant fees from Novartis.

Funding

This research did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

Acknowledgements

We would like to acknowledge all the collaborators of the ‘NIKE’ project: Albertelli M, Bianchi A, Circelli L, De Cicco F, Di Dato C, Fanciulli G, Ferraù F, Gallo M, Giannetta E, Grillo F, Grossrubatscher E, Guadagno E, Guarnotta V, Kara E, Lo Calzo F, Malandrino P, Messina E, Modica R, Muscogiuri G, Pizza G, Razzore P, Rota F, Ruggeri R M and Zatelli M C.

References

  • Barbieri F, Albertelli M, Grillo F, Mohamed A, Saveanu A, Barlier A, Ferone D & Florio T 2014 Neuroendocrine tumors: insights into innovative therapeutic options and rational development of targeted therapies. Drug Discovery Today 19 458468. (https://doi.org/10.1016/j.drudis.2013.10.015)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bertherat J, Tenenbaum F, Perlemoine K, Videau C, Alberini JL, Richard B, Dousset B, Bertagna X & Epelbaum J 2003 Somatostatin receptors 2 and 5 are the major somatostatin receptors in insulinomas: an in vivo and in vitro study. Journal of Clinical Endocrinology and Metabolism 88 53535360. (https://doi.org/10.1210/jc.2002-021895)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Bousquet C, Lasfargues C, Chalabi M, Billah SM, Susini C, Vezzosi D, Caron P & Pyronnet S 2012 Clinical review: current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy. Journal of Clinical Endocrinology and Metabolism 97 727737. (https://doi.org/10.1210/jc.2011-2088)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al.2014 Lanreotide in metastatic enteropancreatic neuroendocrine tumors. New England Journal of Medicine 371 224233. (https://doi.org/10.1056/NEJMoa1316158)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Carter Y, Jaskula-Sztul R, Chen H & Mazeh H 2013 Signaling pathways as specific pharmacologic targets for neuroendocrine tumor therapy: RET, PI3K, MEK, growth factors, and Notch. Neuroendocrinology 97 5766. (https://doi.org/10.1159/000335136)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chan JA, Ryan DP, Zhu AX, Abrams TA, Wolpin BM, Malinowski P, Regan EM, Fuchs CS & Kulke MH 2012 Phase I study of pasireotide (SOM 230) and everolimus (RAD001) in advanced neuroendocrine tumors. Endocrine-Related Cancer 19 615623. (https://doi.org/10.1530/ERC-11-0382)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cives M, Kunz PL, Morse B, Coppola D, Schell MJ, Campos T, Nguyen PT, Nandoskar P, Khandelwal V & Strosberg JR 2015 Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocrine-Related Cancer 22 19. (https://doi.org/10.1530/ERC-14-0360)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Colucci R, Blandizzi C, Ghisu N, Florio T & Del Tacca M 2008 Somatostatin inhibits colon cancer cell growth through cyclooxygenase-2 downregulation. British Journal of Pharmacology 155 198209. (https://doi.org/10.1038/bjp.2008.268)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Connolly R, Gates D, Loh N, Baban D, Thakker R, Johnston B, McCance D, Ardill J, O’Connor DT, Taupenot L, et al.2007 Cox-2 promotes chromogranin A expression and bioactivity: evidence for a prostaglandin E2-dependent mechanism and the involvement of a proximal cyclic adenosine 5′-monophosphate-responsive element. Endocrinology 148 43104317. (https://doi.org/10.1210/en.2007-0167)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cook MR, Pinchot SN, Jaskula-Sztul R, Luo J, Kunnimalaiyaan M & Chen H 2010 Identification of a novel Raf-1 pathway activator that inhibits gastrointestinal carcinoid cell growth. Molecular Cancer Therapeutics 9 429437. (https://doi.org/10.1158/1535-7163.MCT-09-0718)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Culler MD 2011 Somatostatin-dopamine chimeras: a novel approach to treatment of neuroendocrine tumors. Hormone and Metabolic Research 43 854857. (https://doi.org/10.1055/s-0031-1287769)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP, et al.2015 Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Molecular Medicine 7 735753. (https://doi.org/10.15252/emmm.201404346)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fazio N, Abdel-Rahman O, Spada F, Galdy S, De Dosso S, Capdevila J & Scarpa A 2014 RAF signaling in neuroendocrine neoplasms: from bench to bedside. Cancer Treatment Reviews 40 974979. (https://doi.org/10.1016/j.ctrv.2014.06.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferolla P, Brizzi MP, Meyer T, Mansoor W, Mazieres J, Do Cao C, Léna H, Berruti A, Damiano V, Buikhuisen W, et al.2017 Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncology 18 16521664. (https://doi.org/10.1016/S1470-2045(17)30681-2)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Gatto F & Hofland LJ 2011 The role of somatostatin and dopamine D2 receptors in endocrine tumors. Endocrine-Related Cancer 18 R233R251. (https://doi.org/10.1530/ERC-10-0334)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Georgieva I, Koychev D, Wang Y, Holstein J, Hopfenmüller W, Zeitz M & Grabowski P 2010 ZM447439, a novel promising aurora kinase inhibitor, provokes antiproliferative and proapoptotic effects alone and in combination with bio- and chemotherapeutic agents in gastroenteropancreatic neuroendocrine tumor cell lines. Neuroendocrinology 91 121130. (https://doi.org/10.1159/000258705)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hendren NS, Panach K, Brown TJ, Peng L, Beg MS, Weissler J & Mirfakhraee S 2018 Pasireotide for the treatment of refractory hypoglycaemia from malignant insulinoma. Clinical Endocrinology 88 341343. (https://doi.org/10.1111/cen.13503)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henry RR, Ciaraldi TP, Armstrong D, Burke P, Ligueros-Saylan M & Mudaliar S 2013 Hyperglycemia associated with pasireotide: Results from a mechanistic study in healthy volunteers. Journal of Clinical Endocrinology and Metabolism 98 34463453. (https://doi.org/10.1210/jc.2013-1771)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Hofland LJ & Lamberts SW 2003 The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocrine Reviews 24 2847. (https://doi.org/10.1210/er.2000-0001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iida S, Miki Y, Ono K, Akahira J, Nakamura Y, Suzuki T & Sasano H 2012 Synergistic antitumour effects of RAD001 with MEK inhibitors in neuroendocrine tumours: a potential mechanism of therapeutic limitation of mTOR inhibitor. Molecular and Cellular Endocrinology 350 99106. (https://doi.org/10.1016/j.mce.2011.11.024)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Janson ET, Gobl A, Kalkner KM & Oberg K 1996 A comparison between the efficacy of somatostatin receptor scintigraphy and that of in situ hybridization for somatostatin receptor subtype 2 messenger RNA to predict therapeutic outcome in carcinoid patients. Cancer Research 56 25612565.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kanakis G, Grimelius L, Spathis A, Tringidou R, Rassidakis GZ, Oberg K, Kaltsas G & Tsolakis AV 2015 Expression of somatostatin receptors 1–5 and dopamine receptor 2 in lung carcinoids: implications for a therapeutic role. Neuroendocrinology 101 211222. (https://doi.org/10.1159/000381061)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kasajima A, Papotti M, Ito W, Brizzi MP, La Salvia A, Rapa I, Tachibana T, Yazdani S, Sasano H & Volante M 2017 High interlaboratory and interobserver agreement of somatostatin receptor immunohistochemical determination and correlation with response to somatostatin analogs. Human Pathology 72 144152. (https://doi.org/10.1016/j.humpath.2017.11.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kidd M, Drozdov I, Joseph R, Pfragner R, Culler M & Modlin I 2008 Differential cytotoxicity of novel somatostatin and dopamine chimeric compounds on bronchopulmonary and small intestinal neuroendocrine tumor cell lines. Cancer 113 690700. (https://doi.org/10.1002/cncr.23700)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim HS, Lee HS & Kim WH 2011 Clinical significance of protein expression of cyclooxygenase-2 and somatostatin receptors in gastroenteropancreatic neuroendocrine tumors. Cancer Research and Treatment 43 181188. (https://doi.org/10.4143/crt.2011.43.3.181)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Kimura N, Pilichowska M, Date F, Kimura I & Schindler M 1999 Immunohistochemical expression of somatostatin type 2A receptor in neuroendocrine tumors. Clinical Cancer Research 5 34833487.

    • Search Google Scholar
    • Export Citation
  • Kulaksiz H, Eissele R, Rossler D, Schulz S, Hollt V, Cetin Y & Arnold R 2002 Identification of somatostatin receptor subtypes 1, 2A, 3, and 5 in neuroendocrine tumours with subtype specific antibodies. Gut 50 5260. (https://doi.org/10.1136/gut.50.1.52)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kulke MH, Ruszniewski P, Van Cutsem E, Lombard-Bohas C, Valle JW, De Herder WW, Pavel M, Degtyarev E, Brase JC, Bubuteishvili-Pacaud L, et al.2017 A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial Annals of Oncology 28 13091315. (https://doi.org/10.1093/annonc/mdx078)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Kumar U, Sasi R, Suresh S, Patel A, Thangaraju M, Metrakos P, Patel SC & Patel YC 1999 Subtype-selective expression of the five somatostatin receptors (hSSTR1-5) in human pancreatic islet cells: a quantitative double-label immunohistochemical analysis. Diabetes 48 7785. (https://doi.org/10.2337/diabetes.48.1.77)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kvols LK, Oberg KE, O’Dorisio TM, Mohideen P, de Herder WW, Arnold R, Hu K, Zhang Y, Hughes G, Anthony L, et al. 2012 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. Endocrine-Related Cancer 14 657666. (https://doi.org/10.1530/ERC-11-0367)

    • Search Google Scholar
    • Export Citation
  • Laskaratos FM, Rombouts K, Caplin M, Toumpanakis C, Thirlwell C & Mandair D 2017 Neuroendocrine tumors and fibrosis: an unsolved mystery? Cancer 123 47704790. (https://doi.org/10.1002/cncr.31079)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mahipal A, Shibata D, Siegel E, Springett G, Almhanna K, Fulp W, Williams-Elson I & Kim R 2015 Phase I trial of combination of FOLFIRI and pasireotide, a somatostatin analogue, in advanced gastrointestinal malignancies. Investigational New Drugs 33 10931099. (https://doi.org/10.1007/s10637-015-0277-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mazziotti G, Mosca A, Frara S, Vitale G & Giustina A 2017 Somatostatin analogs in the treatment of neuroendocrine tumors: current and emerging aspects. Expert Opinion on Pharmacotherapy 18 16791689. (https://doi.org/10.1080/14656566.2017.1391217)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mizuno S, Kato K, Hashimoto A, Sugitani M, Sheikh A, Komuro S, Jike T, Iwasaki A, Arakawa Y & Nemoto N 2006 Expression of cyclo-oxygenase-2 in gastrointestinal carcinoid tumors. Journal of Gastroenterology and Hepatology 21 13131319. (https://doi.org/10.1111/j.1440-1746.2005.04153.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moatassim-Billah S, Duluc C, Samain R, Jean C, Perraud A, Decaup E, Cassant-Sourdy S, Bakri Y, Selves J, Schmid H, et al.2016 Anti-metastatic potential of somatostatin analog SOM230: indirect pharmacological targeting of pancreatic cancer-associated fibroblasts. Oncotarget 7 4158441598. (https://doi.org/10.18632/oncotarget.9296)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Modlin IM, Pavel M, Kidd M & Gustafsson BI 2010 Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Alimentary Pharmacology and Therapeutics 31 169188. (https://doi.org/10.1111/j.1365-2036.2009.04174.x)

    • Search Google Scholar
    • Export Citation
  • Mohamed A, Blanchard MP, Albertelli M, Barbieri F, Brue T, Niccoli P, Delpero JR, Monges G, Garcia S, Ferone D, et al.2014 Pasireotide and octreotide antiproliferative effects and sst2 trafficking in human pancreatic neuroendocrine tumor cultures. Endocrine-Related Cancer 21 691704. (https://doi.org/10.1530/ERC-14-0086)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mohamed A, Romano D, Saveanu A, Roche C, Albertelli M, Barbieri F, Brue T, Niccoli P, Delpero JR, Garcia S, et al. 2017 Anti-proliferative and anti-secretory effects of everolimus on human pancreatic neuroendocrine tumors primary cultures: is there any benefit from combination with somatostatin analogs? Oncotarget 20 4104441063.

    • Search Google Scholar
    • Export Citation
  • Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC & Meric-Bernstam F 2008 Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors Endocrine-Related Cancer 15 257266. (https://doi.org/10.1677/ERC-07-0202)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mundschenk J, Unger N, Schulz S, Hollt V, Steinke R & Lehnert H 2003 Somatostatin receptor subtypes in human pheochromocytoma: subcellular expression pattern and functional relevance for octreotide scintigraphy. Journal of Clinical Endocrinology and Metabolism 88 51505157. (https://doi.org/10.1210/jc.2003-030262)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Oberstein PE & Saif MW 2012 Safety and efficacy of everolimus in adult patients with neuroendocrine tumors. Clinical Medicine Insights: Oncology 6 4151. (https://doi.org/10.4137/CMO.S7319)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ono K, Suzuki T, Miki Y, Taniyama Y, Nakamura Y, Noda Y, Watanabe M & Sasano H 2007 Somatostatin receptor subtypes in human non-functioning neuroendocrine tumors and effects of somatostatin analogue SOM230 on cell proliferation in cell line NCI-H727. Anticancer Research 27 22312239.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Papotti M, Bongiovanni M, Volante M, Allia E, Landolfi S, Helboe L, Schindler M, Cole SL & Bussolati G 2002 Expression of somatostatin receptor types 1–5 in 81 cases of gastrointestinal and pancreatic endocrine tumors. A correlative immunohistochemical and reverse-transcriptase polymerase chain reaction analysis. Virchows Archiv 440 461475. (https://doi.org/10.1007/s00428-002-0609-x)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Pasquali D, Rossi V, Conzo G, Pannone G, Bufo P, De Bellis A, Renzullo A, Bellastella G, Colao A, Vallone G, et al. 2008 Effects of somatostatin analog SOM230 on cell proliferation, apoptosis, and catecholamine levels in cultured pheochromocytoma cells. Journal of Molecular Endocrinology 40 263271.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pavel M, Valle JW, Eriksson B, Rinke A, Caplin M, Chen J, Costa F, Falkerby J, Fazio N, Gorbounova V, et al.2017 ENETS Consensus Guidelines for the standards of care in neuroendocrine neoplasms: systemic therapy – biotherapy and novel targeted agents. Neuroendocrinology 105 266280. (https://doi.org/10.1159/000471880)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Quinn TJ, Yuan Z, Adem A, Geha R, Vrikshajanani C, Koba W, Fine E, Hughes DT, Schmid HA & Libutti SK 2012 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 152 10681077. (https://doi.org/10.1016/j.surg.2012.08.021)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Reubi JC & Waser B 2003 Concomitant expression of several peptide receptors in neuroendocrine tumours: molecular basis for in vivo multireceptor tumour targeting. European Journal of Nuclear Medicine and Molecular Imaging 30 781793. (https://doi.org/10.1007/s00259-003-1184-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Reubi JC, Schaer JC, Waser B & Mengod G 1994 Expression and localization of somatostatin receptor SSTR1, SSTR2, and SSTR3 messenger RNAs in primary human tumors using in situ hybridization. Cancer Research 54 34553459.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Righi L, Volante M, Tavaglione V, Bille A, Daniele L, Angusti T, Inzani F, Pelosi G, Rindi G & Papotti M 2010 Somatostatin receptor tissue distribution in lung neuroendocrine tumours: a clinicopathologic and immunohistochemical study of 218 ‘clinically aggressive’ cases. Annals of Oncology 21 548555. (https://doi.org/10.1093/annonc/mdp334)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, Mayer C, Aminossadati B, Pape UF, Bläker M, et al. 2009 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. Journal of Clinical Oncology 27 46564663. (https://doi.org/10.1200/JCO.2009.22.8510)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ruszniewski P, Ish-Shalom S, Wymenga M, O’Toole D, Arnold R, Tomassetti P, Bax N, Caplin M, Eriksson B, Glaser B, et al.2004 Rapid and sustained relief from the symptoms of carcinoid syndrome: results from an open 6-month study of the 28-day prolonged-release formulation of lanreotide. Neuroendocrinology 80 244251. (https://doi.org/10.1159/000082875)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salmenkivi K, Haglund C, Ristimaki A, Arola J & Heikkila P 2001 Increased expression of cyclooxygenase-2 in malignant pheochromocytomas. Journal of Clinical Endocrinology and Metabolism 86 56155619. (https://doi.org/10.1210/jcem.86.11.8052)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Saveanu A, Muresan M, De Micco C, Taieb D, Germanetti AL, Sebag F, Henry JF, Brunaud L, Enjalbert A, Weryha G, et al.2011 Expression of somatostatin receptors, dopamine D(2) receptors, noradrenaline transporters, and vesicular monoamine transporters in 52 pheochromocytomas and paragangliomas. Endocrine-Related Cancer 18 287300. (https://doi.org/10.1530/ERC-10-0175)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmid HA & Schoeffter P 2004 Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology 80 4750. (https://doi.org/10.1159/000080741)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Somnay Y, Chen H & Kunnimalaiyaan M 2013 Synergistic effect of pasireotide and teriflunomide in carcinoids in vitro. Neuroendocrinology 97 183192. (https://doi.org/10.1159/000341810)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suleiman Y, Mahipal A, Shibata D, Siegel EM, Jump H, Fulp WJ & Springett GM 2015 Phase I study of combination of pasireotide LAR + gemcitabine in locally advanced or metastatic pancreatic cancer. Cancer Chemotherapy and Pharmacology 76 481487. (https://doi.org/10.1007/s00280-015-2814-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Svejda B, Kidd M, Kazberouk A, Lawrence B, Pfragner R & Modlin IM 2011 Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer 117 41414154. (https://doi.org/10.1002/cncr.26011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tirosh A, Stemmer SM, Solomonov E, Elnekave E, Saeger W, Ravkin Y, Nir K, Talmor Y & Shimon I 2016 Pasireotide for malignant insulinoma. Hormones 15 271276.

    • Search Google Scholar
    • Export Citation
  • Tsuta K, Wistuba II & Moran CA 2012 Differential expression of somatostatin receptors 1–5 in neuroendocrine carcinoma of the lung. Pathology, Research and Practice 208 470474. (https://doi.org/10.1016/j.prp.2012.05.014)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Unger N, Serdiuk I, Sheu SY, Walz MK, Schulz S, Saeger W, Schmid KW, Mann K & Petersenn S 2008 Immunohistochemical localization of somatostatin receptor subtypes in benign and malignant adrenal tumours. Clinical Endocrinology 68 850857. (https://doi.org/10.1111/j.1365-2265.2007.03124.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • van Adrichem RC, de Herder WW, Kamp K, Brugts MP, de Krijger RR, Sprij-Mooij DM, Lamberts SW, van Koetsveld PM, Janssen JA & Hofland LJ 2016 Effects of somatostatin analogs and dopamine agonists on insulin-like growth factor 2-induced insulin receptor isoform A activation by gastroenteropancreatic neuroendocrine tumor cells. Neuroendocrinology 103 815825. (https://doi.org/10.1159/000444280)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Walenkamp A, Crespo G, Fierro Maya F, Fossmark R, Igaz P, Rinke A, Tamagno G, Vitale G, Öberg K & Meyer T 2014 Hallmarks of gastrointestinal neuroendocrine tumours: implications for treatment. Endocrine-Related Cancer 21 R445R460. (https://doi.org/10.1530/ERC-14-0106)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA & Thakker RV 2016 Pasireotide therapy of multiple endocrine neoplasia type 1-associated neuroendocrine tumors in female mice deleted for an Men1 Allele improves survival and reduces tumor progression. Endocrinology 157 17891798. (https://doi.org/10.1210/en.2015-1965)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al.2017 Role of tumor microenvironment in tumorigenesis. Journal of Cancer 8 761773. (https://doi.org/10.7150/jca.17648)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wolin EM 2012 The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointestinal Cancer Research 5 161168.

    • Search Google Scholar
    • Export Citation
  • Wolin EM, Hu K, Hughes G, Bouillaud E, Giannone V & Resendiz KH 2013 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 Chemotherapy and Pharmacology 72 387395. (https://doi.org/10.1007/s00280-013-2202-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wolin EM, Jarzab B, Eriksson B, Walter T, Toumpanakis C, Morse MA, Tomassetti P, Weber MM, Fogelman DR, Ramage J, et al.2015 Phase III study of pasireotide long-acting release in patients with metastatic neuroendocrine tumors and carcinoid symptoms refractory to available somatostatin analogues. Drug Design Development and Therapy 9 50755086. (https://doi.org/10.2147/DDDT.S84177)

    • Search Google Scholar
    • Export Citation
  • Xie Y, Chen S, Wang CH & Tang CW 2011 SOM230 combined with celecoxib prolongs the survival in nude mice with HepG-2 xenografts. Cancer Biology and Therapy 12 8692.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Yao JC, Chan JA, Mita AC, Kundu MG, Hermosillo Reséndiz K, Hu K, Ravichandran S, Strosberg JR & Wolin EM 2017 Phase I dose-escalation study of long-acting pasireotide in patients with neuroendocrine tumors. OncoTargets and Therapy 10 31773186. (https://doi.org/10.2147/OTT.S128547)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yao JC, Lombard-Bohas C, Baudin E, Kvols LK, Rougier P, Ruszniewski P, Hoosen S, St Peter J, Haas T, Lebwohl D, et al.2010 Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial. Journal of Clinical Oncology 28 6976. (https://doi.org/10.1200/JCO.2009.24.2669)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Yerci O, Sehitoglu I, Ugras N, Cubukcu E, Yuce S, Bedir R & Cure E 2015 Somatostatin receptor 2 and 5 expressions in gastroenteropancreatic neuroendocrine tumors in Turkey. Asian Pacific Journal of Cancer Prevention 16 43774381. (https://doi.org/10.7314/APJCP.2015.16.10.4377)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Zatelli MC, Minoia M, Martini C, Tagliati F, Ambrosio MR, Schiavon M, Buratto M, Calabrese F, Gentilin E, Cavallesco G, et al.2010 Everolimus as a new potential antiproliferative agent in aggressive human bronchial carcinoids. Endocrine-Related Cancer 28 719729. (https://doi.org/10.1677/ERC-10-0097)

    • Search Google Scholar
    • Export Citation
  • Zitzmann K, Andersen S, Vlotides G, Spöttl G, Zhang S, Datta R, Culler M, Göke B & Auernhammer CJ 2013 The novel somatostatin receptor 2/dopamine type 2 receptor chimeric compound BIM-23A758 decreases the viability of human GOT1 midgut carcinoid cells. Neuroendocrinology 98 128136. (https://doi.org/10.1159/000353784)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zitzmann K, Ruden J, Brand S, Goke B, Lichtl J, Spottl G & Auernhammer CJ 2010 Compensatory activation of Akt in response to mTOR and Raf inhibitors – a rationale for dual-targeted therapy approaches in neuroendocrine tumour disease. Cancer Letters 295 100109. (https://doi.org/10.1016/j.canlet.2010.02.018)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • View in gallery

    Schematic representation of the intracellular signaling pathways modulated by pasireotide and other drugs involved in combined treatments. AC: adenylyl cyclase; Akt, Protein kinase B; ASCL1, achaete-scute homolog like 1; Bax, bcl2-associated X; CAB, cabergoline; CAF, cancer-associated fibroblast; cAMP, cyclic adenosine monophosphate; Ca2+, calcium ion; COX, cyclooxygenase 2; CX, celecoxib; DR2, dopamine receptor 2; 4E-BP1, eukaryotic translation initiation factor 4E-binding protein 1; eNOS, endothelial nitric oxide synthase; ERK1/2, extracellular signal–regulated kinase 1/2; EV, everolimus; GEM, gemcitabine; IGF2, insulin-like growth factor 2; IL 6, interleukin 6; IRA, insulin receptor isoform A; MEK1/2, mitogen-activated protein kinase kinase 1/2; mTOR, mammalian target of rapamycin; PAS, pasireotide; PI3K, phosphoinositide 3 kinase; RAF; RAS; RTK, receptor tyrosine kinase; SHP-1, -2, Src homology phosphatase type 1, 2; SSTR1-5, somatostatin receptor 1-5; TER, teriflunomide; VEGF, vascular endothelial growth factor.

  • Barbieri F, Albertelli M, Grillo F, Mohamed A, Saveanu A, Barlier A, Ferone D & Florio T 2014 Neuroendocrine tumors: insights into innovative therapeutic options and rational development of targeted therapies. Drug Discovery Today 19 458468. (https://doi.org/10.1016/j.drudis.2013.10.015)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bertherat J, Tenenbaum F, Perlemoine K, Videau C, Alberini JL, Richard B, Dousset B, Bertagna X & Epelbaum J 2003 Somatostatin receptors 2 and 5 are the major somatostatin receptors in insulinomas: an in vivo and in vitro study. Journal of Clinical Endocrinology and Metabolism 88 53535360. (https://doi.org/10.1210/jc.2002-021895)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Bousquet C, Lasfargues C, Chalabi M, Billah SM, Susini C, Vezzosi D, Caron P & Pyronnet S 2012 Clinical review: current scientific rationale for the use of somatostatin analogs and mTOR inhibitors in neuroendocrine tumor therapy. Journal of Clinical Endocrinology and Metabolism 97 727737. (https://doi.org/10.1210/jc.2011-2088)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al.2014 Lanreotide in metastatic enteropancreatic neuroendocrine tumors. New England Journal of Medicine 371 224233. (https://doi.org/10.1056/NEJMoa1316158)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Carter Y, Jaskula-Sztul R, Chen H & Mazeh H 2013 Signaling pathways as specific pharmacologic targets for neuroendocrine tumor therapy: RET, PI3K, MEK, growth factors, and Notch. Neuroendocrinology 97 5766. (https://doi.org/10.1159/000335136)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chan JA, Ryan DP, Zhu AX, Abrams TA, Wolpin BM, Malinowski P, Regan EM, Fuchs CS & Kulke MH 2012 Phase I study of pasireotide (SOM 230) and everolimus (RAD001) in advanced neuroendocrine tumors. Endocrine-Related Cancer 19 615623. (https://doi.org/10.1530/ERC-11-0382)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cives M, Kunz PL, Morse B, Coppola D, Schell MJ, Campos T, Nguyen PT, Nandoskar P, Khandelwal V & Strosberg JR 2015 Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocrine-Related Cancer 22 19. (https://doi.org/10.1530/ERC-14-0360)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Colucci R, Blandizzi C, Ghisu N, Florio T & Del Tacca M 2008 Somatostatin inhibits colon cancer cell growth through cyclooxygenase-2 downregulation. British Journal of Pharmacology 155 198209. (https://doi.org/10.1038/bjp.2008.268)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Connolly R, Gates D, Loh N, Baban D, Thakker R, Johnston B, McCance D, Ardill J, O’Connor DT, Taupenot L, et al.2007 Cox-2 promotes chromogranin A expression and bioactivity: evidence for a prostaglandin E2-dependent mechanism and the involvement of a proximal cyclic adenosine 5′-monophosphate-responsive element. Endocrinology 148 43104317. (https://doi.org/10.1210/en.2007-0167)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cook MR, Pinchot SN, Jaskula-Sztul R, Luo J, Kunnimalaiyaan M & Chen H 2010 Identification of a novel Raf-1 pathway activator that inhibits gastrointestinal carcinoid cell growth. Molecular Cancer Therapeutics 9 429437. (https://doi.org/10.1158/1535-7163.MCT-09-0718)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Culler MD 2011 Somatostatin-dopamine chimeras: a novel approach to treatment of neuroendocrine tumors. Hormone and Metabolic Research 43 854857. (https://doi.org/10.1055/s-0031-1287769)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP, et al.2015 Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Molecular Medicine 7 735753. (https://doi.org/10.15252/emmm.201404346)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fazio N, Abdel-Rahman O, Spada F, Galdy S, De Dosso S, Capdevila J & Scarpa A 2014 RAF signaling in neuroendocrine neoplasms: from bench to bedside. Cancer Treatment Reviews 40 974979. (https://doi.org/10.1016/j.ctrv.2014.06.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferolla P, Brizzi MP, Meyer T, Mansoor W, Mazieres J, Do Cao C, Léna H, Berruti A, Damiano V, Buikhuisen W, et al.2017 Efficacy and safety of long-acting pasireotide or everolimus alone or in combination in patients with advanced carcinoids of the lung and thymus (LUNA): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncology 18 16521664. (https://doi.org/10.1016/S1470-2045(17)30681-2)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Gatto F & Hofland LJ 2011 The role of somatostatin and dopamine D2 receptors in endocrine tumors. Endocrine-Related Cancer 18 R233R251. (https://doi.org/10.1530/ERC-10-0334)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Georgieva I, Koychev D, Wang Y, Holstein J, Hopfenmüller W, Zeitz M & Grabowski P 2010 ZM447439, a novel promising aurora kinase inhibitor, provokes antiproliferative and proapoptotic effects alone and in combination with bio- and chemotherapeutic agents in gastroenteropancreatic neuroendocrine tumor cell lines. Neuroendocrinology 91 121130. (https://doi.org/10.1159/000258705)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hendren NS, Panach K, Brown TJ, Peng L, Beg MS, Weissler J & Mirfakhraee S 2018 Pasireotide for the treatment of refractory hypoglycaemia from malignant insulinoma. Clinical Endocrinology 88 341343. (https://doi.org/10.1111/cen.13503)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henry RR, Ciaraldi TP, Armstrong D, Burke P, Ligueros-Saylan M & Mudaliar S 2013 Hyperglycemia associated with pasireotide: Results from a mechanistic study in healthy volunteers. Journal of Clinical Endocrinology and Metabolism 98 34463453. (https://doi.org/10.1210/jc.2013-1771)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Hofland LJ & Lamberts SW 2003 The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocrine Reviews 24 2847. (https://doi.org/10.1210/er.2000-0001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iida S, Miki Y, Ono K, Akahira J, Nakamura Y, Suzuki T & Sasano H 2012 Synergistic antitumour effects of RAD001 with MEK inhibitors in neuroendocrine tumours: a potential mechanism of therapeutic limitation of mTOR inhibitor. Molecular and Cellular Endocrinology 350 99106. (https://doi.org/10.1016/j.mce.2011.11.024)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Janson ET, Gobl A, Kalkner KM & Oberg K 1996 A comparison between the efficacy of somatostatin receptor scintigraphy and that of in situ hybridization for somatostatin receptor subtype 2 messenger RNA to predict therapeutic outcome in carcinoid patients. Cancer Research 56 25612565.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kanakis G, Grimelius L, Spathis A, Tringidou R, Rassidakis GZ, Oberg K, Kaltsas G & Tsolakis AV 2015 Expression of somatostatin receptors 1–5 and dopamine receptor 2 in lung carcinoids: implications for a therapeutic role. Neuroendocrinology 101 211222. (https://doi.org/10.1159/000381061)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kasajima A, Papotti M, Ito W, Brizzi MP, La Salvia A, Rapa I, Tachibana T, Yazdani S, Sasano H & Volante M 2017 High interlaboratory and interobserver agreement of somatostatin receptor immunohistochemical determination and correlation with response to somatostatin analogs. Human Pathology 72 144152. (https://doi.org/10.1016/j.humpath.2017.11.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kidd M, Drozdov I, Joseph R, Pfragner R, Culler M & Modlin I 2008 Differential cytotoxicity of novel somatostatin and dopamine chimeric compounds on bronchopulmonary and small intestinal neuroendocrine tumor cell lines. Cancer 113 690700. (https://doi.org/10.1002/cncr.23700)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim HS, Lee HS & Kim WH 2011 Clinical significance of protein expression of cyclooxygenase-2 and somatostatin receptors in gastroenteropancreatic neuroendocrine tumors. Cancer Research and Treatment 43 181188. (https://doi.org/10.4143/crt.2011.43.3.181)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Kimura N, Pilichowska M, Date F, Kimura I & Schindler M 1999 Immunohistochemical expression of somatostatin type 2A receptor in neuroendocrine tumors. Clinical Cancer Research 5 34833487.

    • Search Google Scholar
    • Export Citation
  • Kulaksiz H, Eissele R, Rossler D, Schulz S, Hollt V, Cetin Y & Arnold R 2002 Identification of somatostatin receptor subtypes 1, 2A, 3, and 5 in neuroendocrine tumours with subtype specific antibodies. Gut 50 5260. (https://doi.org/10.1136/gut.50.1.52)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kulke MH, Ruszniewski P, Van Cutsem E, Lombard-Bohas C, Valle JW, De Herder WW, Pavel M, Degtyarev E, Brase JC, Bubuteishvili-Pacaud L, et al.2017 A randomized, open-label, phase 2 study of everolimus in combination with pasireotide LAR or everolimus alone in advanced, well-differentiated, progressive pancreatic neuroendocrine tumors: COOPERATE-2 trial Annals of Oncology 28 13091315. (https://doi.org/10.1093/annonc/mdx078)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Kumar U, Sasi R, Suresh S, Patel A, Thangaraju M, Metrakos P, Patel SC & Patel YC 1999 Subtype-selective expression of the five somatostatin receptors (hSSTR1-5) in human pancreatic islet cells: a quantitative double-label immunohistochemical analysis. Diabetes 48 7785. (https://doi.org/10.2337/diabetes.48.1.77)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kvols LK, Oberg KE, O’Dorisio TM, Mohideen P, de Herder WW, Arnold R, Hu K, Zhang Y, Hughes G, Anthony L, et al. 2012 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. Endocrine-Related Cancer 14 657666. (https://doi.org/10.1530/ERC-11-0367)

    • Search Google Scholar
    • Export Citation
  • Laskaratos FM, Rombouts K, Caplin M, Toumpanakis C, Thirlwell C & Mandair D 2017 Neuroendocrine tumors and fibrosis: an unsolved mystery? Cancer 123 47704790. (https://doi.org/10.1002/cncr.31079)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mahipal A, Shibata D, Siegel E, Springett G, Almhanna K, Fulp W, Williams-Elson I & Kim R 2015 Phase I trial of combination of FOLFIRI and pasireotide, a somatostatin analogue, in advanced gastrointestinal malignancies. Investigational New Drugs 33 10931099. (https://doi.org/10.1007/s10637-015-0277-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mazziotti G, Mosca A, Frara S, Vitale G & Giustina A 2017 Somatostatin analogs in the treatment of neuroendocrine tumors: current and emerging aspects. Expert Opinion on Pharmacotherapy 18 16791689. (https://doi.org/10.1080/14656566.2017.1391217)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mizuno S, Kato K, Hashimoto A, Sugitani M, Sheikh A, Komuro S, Jike T, Iwasaki A, Arakawa Y & Nemoto N 2006 Expression of cyclo-oxygenase-2 in gastrointestinal carcinoid tumors. Journal of Gastroenterology and Hepatology 21 13131319. (https://doi.org/10.1111/j.1440-1746.2005.04153.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moatassim-Billah S, Duluc C, Samain R, Jean C, Perraud A, Decaup E, Cassant-Sourdy S, Bakri Y, Selves J, Schmid H, et al.2016 Anti-metastatic potential of somatostatin analog SOM230: indirect pharmacological targeting of pancreatic cancer-associated fibroblasts. Oncotarget 7 4158441598. (https://doi.org/10.18632/oncotarget.9296)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Modlin IM, Pavel M, Kidd M & Gustafsson BI 2010 Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Alimentary Pharmacology and Therapeutics 31 169188. (https://doi.org/10.1111/j.1365-2036.2009.04174.x)

    • Search Google Scholar
    • Export Citation
  • Mohamed A, Blanchard MP, Albertelli M, Barbieri F, Brue T, Niccoli P, Delpero JR, Monges G, Garcia S, Ferone D, et al.2014 Pasireotide and octreotide antiproliferative effects and sst2 trafficking in human pancreatic neuroendocrine tumor cultures. Endocrine-Related Cancer 21 691704. (https://doi.org/10.1530/ERC-14-0086)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mohamed A, Romano D, Saveanu A, Roche C, Albertelli M, Barbieri F, Brue T, Niccoli P, Delpero JR, Garcia S, et al. 2017 Anti-proliferative and anti-secretory effects of everolimus on human pancreatic neuroendocrine tumors primary cultures: is there any benefit from combination with somatostatin analogs? Oncotarget 20 4104441063.

    • Search Google Scholar
    • Export Citation
  • Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC & Meric-Bernstam F 2008 Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors Endocrine-Related Cancer 15 257266. (https://doi.org/10.1677/ERC-07-0202)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mundschenk J, Unger N, Schulz S, Hollt V, Steinke R & Lehnert H 2003 Somatostatin receptor subtypes in human pheochromocytoma: subcellular expression pattern and functional relevance for octreotide scintigraphy. Journal of Clinical Endocrinology and Metabolism 88 51505157. (https://doi.org/10.1210/jc.2003-030262)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Oberstein PE & Saif MW 2012 Safety and efficacy of everolimus in adult patients with neuroendocrine tumors. Clinical Medicine Insights: Oncology 6 4151. (https://doi.org/10.4137/CMO.S7319)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ono K, Suzuki T, Miki Y, Taniyama Y, Nakamura Y, Noda Y, Watanabe M & Sasano H 2007 Somatostatin receptor subtypes in human non-functioning neuroendocrine tumors and effects of somatostatin analogue SOM230 on cell proliferation in cell line NCI-H727. Anticancer Research 27 22312239.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Papotti M, Bongiovanni M, Volante M, Allia E, Landolfi S, Helboe L, Schindler M, Cole SL & Bussolati G 2002 Expression of somatostatin receptor types 1–5 in 81 cases of gastrointestinal and pancreatic endocrine tumors. A correlative immunohistochemical and reverse-transcriptase polymerase chain reaction analysis. Virchows Archiv 440 461475. (https://doi.org/10.1007/s00428-002-0609-x)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Pasquali D, Rossi V, Conzo G, Pannone G, Bufo P, De Bellis A, Renzullo A, Bellastella G, Colao A, Vallone G, et al. 2008 Effects of somatostatin analog SOM230 on cell proliferation, apoptosis, and catecholamine levels in cultured pheochromocytoma cells. Journal of Molecular Endocrinology 40 263271.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pavel M, Valle JW, Eriksson B, Rinke A, Caplin M, Chen J, Costa F, Falkerby J, Fazio N, Gorbounova V, et al.2017 ENETS Consensus Guidelines for the standards of care in neuroendocrine neoplasms: systemic therapy – biotherapy and novel targeted agents. Neuroendocrinology 105 266280. (https://doi.org/10.1159/000471880)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Quinn TJ, Yuan Z, Adem A, Geha R, Vrikshajanani C, Koba W, Fine E, Hughes DT, Schmid HA & Libutti SK 2012 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 152 10681077. (https://doi.org/10.1016/j.surg.2012.08.021)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Reubi JC & Waser B 2003 Concomitant expression of several peptide receptors in neuroendocrine tumours: molecular basis for in vivo multireceptor tumour targeting. European Journal of Nuclear Medicine and Molecular Imaging 30 781793. (https://doi.org/10.1007/s00259-003-1184-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Reubi JC, Schaer JC, Waser B & Mengod G 1994 Expression and localization of somatostatin receptor SSTR1, SSTR2, and SSTR3 messenger RNAs in primary human tumors using in situ hybridization. Cancer Research 54 34553459.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Righi L, Volante M, Tavaglione V, Bille A, Daniele L, Angusti T, Inzani F, Pelosi G, Rindi G & Papotti M 2010 Somatostatin receptor tissue distribution in lung neuroendocrine tumours: a clinicopathologic and immunohistochemical study of 218 ‘clinically aggressive’ cases. Annals of Oncology 21 548555. (https://doi.org/10.1093/annonc/mdp334)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Rinke A, Müller HH, Schade-Brittinger C, Klose KJ, Barth P, Wied M, Mayer C, Aminossadati B, Pape UF, Bläker M, et al. 2009 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. Journal of Clinical Oncology 27 46564663. (https://doi.org/10.1200/JCO.2009.22.8510)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ruszniewski P, Ish-Shalom S, Wymenga M, O’Toole D, Arnold R, Tomassetti P, Bax N, Caplin M, Eriksson B, Glaser B, et al.2004 Rapid and sustained relief from the symptoms of carcinoid syndrome: results from an open 6-month study of the 28-day prolonged-release formulation of lanreotide. Neuroendocrinology 80 244251. (https://doi.org/10.1159/000082875)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salmenkivi K, Haglund C, Ristimaki A, Arola J & Heikkila P 2001 Increased expression of cyclooxygenase-2 in malignant pheochromocytomas. Journal of Clinical Endocrinology and Metabolism 86 56155619. (https://doi.org/10.1210/jcem.86.11.8052)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Saveanu A, Muresan M, De Micco C, Taieb D, Germanetti AL, Sebag F, Henry JF, Brunaud L, Enjalbert A, Weryha G, et al.2011 Expression of somatostatin receptors, dopamine D(2) receptors, noradrenaline transporters, and vesicular monoamine transporters in 52 pheochromocytomas and paragangliomas. Endocrine-Related Cancer 18 287300. (https://doi.org/10.1530/ERC-10-0175)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmid HA & Schoeffter P 2004 Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology 80 4750. (https://doi.org/10.1159/000080741)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Somnay Y, Chen H & Kunnimalaiyaan M 2013 Synergistic effect of pasireotide and teriflunomide in carcinoids in vitro. Neuroendocrinology 97 183192. (https://doi.org/10.1159/000341810)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Suleiman Y, Mahipal A, Shibata D, Siegel EM, Jump H, Fulp WJ & Springett GM 2015 Phase I study of combination of pasireotide LAR + gemcitabine in locally advanced or metastatic pancreatic cancer. Cancer Chemotherapy and Pharmacology 76 481487. (https://doi.org/10.1007/s00280-015-2814-8)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Svejda B, Kidd M, Kazberouk A, Lawrence B, Pfragner R & Modlin IM 2011 Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer 117 41414154. (https://doi.org/10.1002/cncr.26011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tirosh A, Stemmer SM, Solomonov E, Elnekave E, Saeger W, Ravkin Y, Nir K, Talmor Y & Shimon I 2016 Pasireotide for malignant insulinoma. Hormones 15 271276.

    • Search Google Scholar
    • Export Citation
  • Tsuta K, Wistuba II & Moran CA 2012 Differential expression of somatostatin receptors 1–5 in neuroendocrine carcinoma of the lung. Pathology, Research and Practice 208 470474. (https://doi.org/10.1016/j.prp.2012.05.014)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Unger N, Serdiuk I, Sheu SY, Walz MK, Schulz S, Saeger W, Schmid KW, Mann K & Petersenn S 2008 Immunohistochemical localization of somatostatin receptor subtypes in benign and malignant adrenal tumours. Clinical Endocrinology 68 850857. (https://doi.org/10.1111/j.1365-2265.2007.03124.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • van Adrichem RC, de Herder WW, Kamp K, Brugts MP, de Krijger RR, Sprij-Mooij DM, Lamberts SW, van Koetsveld PM, Janssen JA & Hofland LJ 2016 Effects of somatostatin analogs and dopamine agonists on insulin-like growth factor 2-induced insulin receptor isoform A activation by gastroenteropancreatic neuroendocrine tumor cells. Neuroendocrinology 103 815825. (https://doi.org/10.1159/000444280)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Walenkamp A, Crespo G, Fierro Maya F, Fossmark R, Igaz P, Rinke A, Tamagno G, Vitale G, Öberg K & Meyer T 2014 Hallmarks of gastrointestinal neuroendocrine tumours: implications for treatment. Endocrine-Related Cancer 21 R445R460. (https://doi.org/10.1530/ERC-14-0106)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA & Thakker RV 2016 Pasireotide therapy of multiple endocrine neoplasia type 1-associated neuroendocrine tumors in female mice deleted for an Men1 Allele improves survival and reduces tumor progression. Endocrinology 157 17891798. (https://doi.org/10.1210/en.2015-1965)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang M, Zhao J, Zhang L, Wei F, Lian Y, Wu Y, Gong Z, Zhang S, Zhou J, Cao K, et al.2017 Role of tumor microenvironment in tumorigenesis. Journal of Cancer 8 761773. (https://doi.org/10.7150/jca.17648)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wolin EM 2012 The expanding role of somatostatin analogs in the management of neuroendocrine tumors. Gastrointestinal Cancer Research 5 161168.