Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
Contents lists available at ScienceDirect
Best Practice & Research Clinical Endocrinology & Metabolism journal homepage: www.elsevier.com/locate/beem
6
Neuroendocrine tumours e Medical therapy: Biological Anja Rinke, Dr., Consultant Internal Medicine a, *, Sebastian Krug, Dr. med. b a b
Department of Gastroenterology, Philipps University Marburg, Germany Department of Internal Medicine I, Martin Luther University Halle, Germany
a r t i c l e i n f o Article history: Available online xxx Keywords: somatostatin analogue octreotide lanreotide pasireotide interferon-a symptomatic treatment antiproliferative treatment
Somatostatin analogues (SSA) are well established antisecretory drugs that have been used as first line treatment for symptomatic control in hormonally active neuroendocrine tumours (NET) for three decades. Both available depot formulations of SSA, longacting repeatable (LAR) octreotide and lanreotide autogel, seem similarly effective and well tolerated, although comparative trials in NET have not been performed. The importance of SSA as antiproliferative treatment has been increasingly recognized during recent years. Two placebo-controlled trials demonstrated significant prolongation of progression free survival under SSA treatment. However, objective response as assessed by imaging is rare. Interferon-a (IFNa) also has antisecretory and antiproliferative efficacy in NET. Due to the less favourable toxicity profile it mainly has a role as add-on option in the refractory setting, especially in carcinoid syndrome patients. Further studies are needed to evaluate the antiproliferative efficacy of the multiligand SSA pasireotide and the role of pegylated IFNa. © 2015 Elsevier Ltd. All rights reserved.
Introduction and definition of biotherapy Medical therapy of neuroendocrine tumours (NETs) aims to ameliorate hormonal symptoms caused by the tumour and/or to control tumour growth. Traditionally, systemic treatments are divided into biotherapy, chemotherapy and so-called molecular targeted agents. Biotherapy implies that substances occurring naturally within the body or pharmacological derivatives thereof are used for medical * Corresponding author. Department of Gastroenterology University Hospital Marburg Baldingerstraße 1 35043 Marburg, Germany. Tel.: þ49 6421 58 66460; Fax: þ49 6421 58 68922. E-mail address:
[email protected] (A. Rinke). http://dx.doi.org/10.1016/j.beem.2015.09.004 1521-690X/© 2015 Elsevier Ltd. All rights reserved.
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
2
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
treatment. Biotherapy of NETs mainly comprises somatostatin analogues (SSAs) and interferon-a (IFNa). SSA: history, available drugs Somatostatin and somatostatin receptors Somatostatin (SST) is a natural polypeptide with two bioactive forms of 14 and 28 amino acids, respectively, discovered incidentally as growth hormone (GH) inhibitor in 1972 [1,2]. SST was subsequently found to inhibit not only GH but also the release of many gastrointestinal hormones including insulin, gastrin and glucagon. It additionally reduces exocrine gastric and pancreatic secretion and motility, portal blood flow and intestinal absorption [2]. The physiological effects of SST are the result of specific interaction with somatostatin receptors, a family of G-coupled proteins with five receptor subtypes (SSTR1-5). Natural SST binds to all five subtypes with high affinity [2]. NETs often express SSTRs at high levels with SSTR2 being the most prevalent subtype [3,4]. Levels of SSTR expression and predominant subtype vary between tumour types. While half of the insulinoma patients express SSTR2, high levels of SSTR2 are found in more than 90% of gastrinoma patients [2]. In patients with bronchopulmonary NETs SSTR1 was recently reported to be the most prevalent subtype [5]. Somatostatin analogues (SSAs) e developmental history The natural SSTs have a very short circulation half-life of about two minutes. For clinical use a continuous infusion is necessary. The development of synthetic peptides with longer half-lives e including the octapeptides octreotide and lanreotide - in the early eighties was a major step towards therapeutic applicability [6]. These synthetic SSAs retain the binding affinity for SSTRs e at least with high affinity for SSTR2 and moderate affinity for SSTR5 e and are more resistant to peptidases. In 1988 octreotide was approved for treatment of hormone syndromes like carcinoid syndrome. With a halflife of two hours this first clinically available SSA provided control of symptoms by subcutaneous (sc) injections three times a day. In the 1990ies the development of depot formulations of SSA provided more sustained drug levels and improved quality of life. In 1995 octreotide long-acting repeatable (LAR) was introduced which is an incorporation of octreotide into microspheres of the biodegradable polymer Poly-DL-lactide-co-glycolide [2]. Octreotide LAR is typically administered intramuscularly (im) every 4 weeks. For initiation of octreotide LAR treatment application of subcutaneous octreotide for about two weeks is recommended until the plateau level of serum concentration is reached. Simultaneously with octreotide LAR a slow release formulation of lanreotide e lanreotide microparticles (MP) e was licensed in France. This formulation was injected im every 14 days. Lanreotide autogel is a viscous aqueous solution composed of only lanreotide and water that is usually administered deep sc every four weeks. After a single injection the peak serum concentration is reached on the first day, a steady state level after four injections [7]. This formulation was licensed in France in 2001, in Germany in 2007 and was very recently also approved by the FDA. The depot formulations octreotide LAR and lanreotide autogel are the most widely used SSAs in clinical practice today. They are similar with regard to SSTR binding affinity, clinical efficacy and side effects. There are no comparative randomized trials of both formulations available in NET patients. In 2004 pasireotide was developed. Its binding affinity to SSTR2 is lower compared to octreotide and lanreotide. However, its affinity to SSTR1, 3 and 5 is much higher [8]. The drug is available as shortacting subcutaneous formulation and long-acting release intramuscular formulation, usually administered at 60 mg monthly. While pasireotide has been approved for Cushing's disease and acromegaly, the role in NET still has to be defined. Mechanism of biological activity Binding of SSA results in inhibition of calcium channels and stimulation of various potassium channels, thus leading to depletion of intracellular calcium concentration and inhibition of adenylate Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
3
cyclase. Decrease in cyclic adenosine monophosphate (cAMP) levels and inhibition of protein phosphatases leads to inhibition of exocytosis. This mechanism explains the antisecretory effects of SSA [2]. In addition, direct antiproliferative mechanisms as a result of SSA binding to SSTR on the tumour cell and indirect mechanisms have been described. Indirect mechanisms include inhibition of circulating growth factors as well as antiangiogenic effects through interaction with SSTRs on cells of the surrounding microenvironment [2]. Upon activation, all SSTRs induce cell cycle inhibitors such as p27 and p21, thus leading to cell cycle arrest [9]. Binding of SSA to SSTR activates phosphotyrosine phosphatases resulting in suppression of mitogenic pathways such as the ERK1/2/PI3K/AKT pathway. Further potential antiproliferative mechanisms of SSA that have been described in various reports include: direct induction of apoptosis, restoration of functional gap junctions and changes in natural killer cell activity [10,11]. SSAs in hypersecretion syndromes SSAs have been the mainstay for the control of hormonal syndromes for three decades. Symptomatic relief is reported in about 70% (40e100%) of treated patients, biochemical response (decrease of the marker hormones of at least 50%) in about 50% of patients (18%e100%) [2]. Symptom control is dose dependent [12]. The duration of symptom control is variable. Escape mechanisms include desensitization to the inhibitory SST effects (tachyphylaxis) [13]. SSAs in carcinoid syndrome SSAs are the treatment of choice for palliating flushing and diarrhoea in patients with carcinoid syndrome. An early trial reported symptom control in 88% of the patients with octreotide 150 mg three times daily [14]. A randomized trial of subcutaneous octreotide versus octreotide LAR demonstrated equivalence of both formulations [15]. Numerous studies confirmed similar results for octreotide and lanreotide in terms of symptomatic response [16e21]. A cross over study on 33 patients with carcinoid syndrome compared the efficacy of 30 mg lanreotide MP every 10 days and 200 mg octreotide three times a day. Both drugs were equally efficacious in terms of symptom control and reduction in biomarkers [16]. 71 patients with carcinoid syndrome were included in an open multicentric trial of lanreotide autogel for 6 months [19]. At the end of the study 80% of patients with flushing and 75% of patients with diarrhoea had an improvement of symptoms compared to baseline. A release of at least 50% of flushing episodes was reported in 65%, a 50% reduction of diarrhoea in 18% of patients. The less pronounced effect on diarrhoea may be explained by other causes for diarrhoea than hormone hypersecretion like bile acid malabsorption after ileal resection, short bowel syndrome and side effects of SSAs. In a study of 108 patients with metastatic midgut NETs and carcinoid syndrome, octreotide LAR treatment resulted in sufficient long term symptomatic control in 45% of cases [20]. The same centre provided data on long term treatment with lanreotide autogel in 69 patients with carcinoid syndrome. In 74% of patients the carcinoid syndrome was well controlled with lanreotide autogel alone throughout the study period [21]. The international non-interventional SymNET trial (NCT01234168) assessed patient-reported satisfaction with diarrhoea control in 273 patients with carcinoid syndrome. 75.7% of patients were satisfied with the control of diarrhoea after lanreotide autogel treatment [22]. Pasireotide was evaluated in NETs with carcinoid syndrome refractory to available SSAs. In a phase II study subcutaneous pasireotide was effective in controlling the symptoms of carcinoid syndrome in 27% of the cohort [23]. However, a randomized phase III study of pasireotide LAR versus high-dose octreotide LAR (40 mg every 28 days) among patients with refractory carcinoid syndrome failed to prove an advantage of pasireotide LAR in symptom control [24]. Treatment with SSAs is associated with >50% decrease of urinary 5- hydroxyindoleacetic acid (5HIAA) in about half of the patients treated with octreotide and 40% of patients treated with lanreotide [2,21]. Carcinoid heart disease (CHD) is a severe complication of the carcinoid syndrome, usually involving the right-sided heart valves and eventually leading to right heart failure. As high levels of 5-HIAA are associated with increased risk for CHD [25], decrease in hormone levels under SSA treatment could Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
4
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
contribute to reduced risk or delay of development of CHD. A clear proof of an SSA effect on CHD, however, is lacking so far. Carcinoid crisis manifests by extreme fluctuations of blood pressure, flushing, bronchoconstriction and arrhythmias that can occur during anaesthetic induction and manipulation of tumour masses during surgical or radiological procedures. Therefore, in patients with known carcinoid syndrome who undergo surgery or radiological interventions prophylactic supplementary sc or iv octreotide is recommended [26,27]. SSAs in insulinomas Most insulinomas are benign and can be cured by surgery. In the rare metastasizing insulinomas SSA treatment often is of limited value for glycaemic control. One probable reason is the low expression of SSTR2 [28]. Even deterioration of hypoglycaemia may occur as a result of inhibition of the counter regulatory hormone glucagon [29]. Therefore, in this indication SSA treatment should not be commenced in an outpatient setting. Further studies are needed to evaluate the potential role of pasireotide in patients with malignant insulinoma. Pasireotide does not inhibit counter regulatory glucagon secretion and often induces hyperglycaemia. Therefore, it could be helpful for treating hypoglycaemia in insulinoma patients. SSAs in Zollinger Ellison Syndrome (gastrinoma) SSA lower gastrin levels and can ameliorate symptoms of Zollinger Ellison Syndrome [30]. However, proton pump inhibitors are the treatment of choice for symptom control as they are highly effective and oral available [31]. SSAs in Verner Morrison Syndrome (VIPoma) Treatment with SSAs results in a rapid reduction of the excessive secretory diarrhoea caused by vasoactive intestinal polypeptide (VIP) secreting pancreatic NETs [32,33] and is therefore indicated in this rare disease. SSAs in glucagonoma syndrome The necrolytic migratory erythema-a characteristic skin rash caused by glucagon secreting pancreatic NETs e can resolve rapidly after initiation of SSA treatment [34,35]. The European Neuroendocrine Tumour Society (ENETS) therefore recommends treatment with SSA in patients with glucagonoma syndrome [36]. SSAs in ectopic ACTH syndrome A Cushing's syndrome caused by release of adrenocorticotropin (ACTH) rarely occurs in patients with NETs and there is insufficient data to conclude on the role of SSA in this condition. Some reports have described relief of symptoms following treatment with SSA [37,38] although other medical (ketoconazole, metyrapone) and non-medical treatments (adrenalectomy) may be necessary. Pasireotide is approved for Cushing's disease but its role for Cushing's syndrome due to ectopic ACTH release by NETs needs further investigation. Adverse effects of SSA SSAs are generally well tolerated. Most common side effects are diarrhoea, abdominal discomfort, flatulence, nausea and local reactions at the site of injection. Gastrointestinal side effects often decrease in intensity with duration of treatment. Cholelithiasis is reported in 3e11% [21,39,40] of patients, in most cases asymptomatic but complications such as acute cholecystitis, bilious attacks or acute pancreatitis may occur. Therefore, a Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
5
prophylactic cholecystectomy should be discussed in patients on SSAs who undergo surgery for other reasons [41]. Steatorrhoea is reported in different frequencies, most cases can be sufficiently treated with pancreatic enzymes. Octreotide and lanreotide can cause hyperglycaemia in about 5%e10% of patients. Rare side effects include hypoglycaemia, hair loss, hypothyreoidism, headache, myalgia, acute hepatitis, hyperbilirubinaemia, bradycardia, obstipation and paralytic ileus. Pasireotide has a similar tolerability profile except for a higher rate of hyperglycaemia [23,24]. Interferon (IFN) short history and mechanism of action Interferons (IFN) form a family of cytokines, initially described in the middle of the 20th century by Isaacs and Lindenmann [42]. To date, three IFN types are characterized, based on their respective cell surface receptors, of which the type I IFN (IFNeI) comprises IFN-a and IFN-b [43]. The IFN-I members bind the heterodimeric IFNa/b receptor, thereby leading to phosphorylation and initiation of signal transduction [44]. Since 1971 IFN-a has been used for the treatment of certain solid tumours. Direct as well as indirect antitumour effects of IFN-a have been described: IFN-a can target tumour cells itself and induce cell cycle arrest and apoptosis, but also has immunomodulatory and anti-angiogenic activities [45,46]. However, the complexity of its impact in neuroendocrine tumours is not fully understood [47e50]. Interferon (IFN), available drugs IFN-a has been approved for treatment of carcinoid syndrome. Most data have been acquired using human leucocyte IFN-a. Nowadays, however, predominantly recombinant IFN-a2a, IFN-a2b and polyethylene glycol-modified (pegylated) forms of IFN-a are in clinical use. The dose has to be individually titrated based on a balance between effect and quality of life. Following schedules can be applied: IFN-a2A 3e9 MU/day sc 3e5/week IFN-a2B 3e9 MU/day sc 3e5/week Pegylated IFN-a2A or IFN-a2B 50e150 mg sc 1/week
IFN-a in hypersecretion syndromes € In 1983 Oberg published first data on the clinical efficacy of IFN-a in six patients with carcinoid syndrome [51]. Subsequently, various non-randomized, mainly retrospective trials of heterogeneous cohorts reported symptom control and biochemical response between 30e70% and 50e60%, respec€lby and colleagues compared monotherapy with tively [52e56]. The randomized prospective trial by Ko octreotide (100e200 mg 3x/day) with combination treatment (octreotide plus IFN-a 3e5 MU 3/week) in a homogeneous population of patients with carcinoid syndrome [57]. Both treatment regimens resulted in significant reduction of 5-HIAA excretion without differences between the treatment arms. Data for efficacy of IFN-a with regard to symptomatic control in other hormone syndromes caused by NETs are limited. According to the ENETS guidelines IFN-a can be considered for symptomatic treatment of functional pNET and carcinoid syndrome [58,59] in case of intolerance of SSAs and insufficient antisecretory effects of SSA. However, due to the unfavourable toxicity profile, IFN-a is not first therapeutic choice in functioning NETs. Adverse effects of IFN-a Compared to SSA, IFN-a is associated with more side effects, mostly grade 1 and 2. Temporary flulike symptoms, fatigue and discrete weight loss are common adverse events occurring in 50e80% of the patients [60]. Additionally, myelodepression comprising anaemia (~25%), leukopenia (up to 40% grade Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
6
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
I/II and 20% grade III/IV) and thrombocytopenia (~10e20%) as well as elevated liver enzymes (~30%) frequently occur during therapy [60e62]. Abdominal complaints with pain, diarrhoea, vomiting and nausea were less frequently reported (10e40%) [62e64]. Rare adverse events include depression, autoimmune phenomena, skin rash, headache and renal impairment.
Practice Points Hypersecretion syndromes SSA are well established for the control of symptoms of patients suffering from carcinoid syndrome, Verner Morrison Syndrome and glucagonoma syndrome Lanreotide and Octreotide seem to be equally effective in controlling hypersecretion syndromes Perioperative and periinterventional application of iv octreotide is indicated to prevent carcinoid crisis In patients with refractory carcinoid syndrome medical treatment options include increasing doses of SSA, addition of IFNa and treatment with pasireotide (not approved for this indication)
Biotherapy in antiproliferative intention SSA for tumour control In addition to their well established role in symptom control, somatostatin analogues also demonstrated inhibition of growth in cancer cell lines. Initial case reports showed tumour shrinkage under octreotide treatment [65], in the following years the antiproliferative efficacy was questioned as remissions were rarely found. Most of these uncontrolled phase II studies included only small numbers and heterogeneous cohorts. They reported high rates of stabilization (30e80%), but tumour progression at study entry was not always required. As expected, the rate of stabilization was usually higher for patients without disease progression at treatment onset [66,67]. It was also higher in intestinal than in pancreatic NETs [68]. Prognosis in non-responders to SSA treatment was worse [63,68]. In the recent Spanish study with lanreotide autogel in GEP-NET patients with documented tumour progression within 6 months prior to study entry, Martin-Richard reported a progression free survival (PFS) of 12.9 months [69]. The results on morphologic response under SSA treatment are summarized in Table 1. A significant antitumour activity of SSAs was documented in two prospective randomized placebo controlled trials in recent years. The PROMID study [39] compared octreotide LAR 30 mg every 4 weeks versus placebo in 85 treatment naïve patients with well-differentiated midgut NETs. Both patients with nonfunctioning tumours or mild carcinoid syndrome were included. The primary endpoint was time to tumour progression (TTP). TTP in the octreotide LAR group was significantly longer with 14.3 months compared to 6 months in the placebo group (HR 0.34; p ¼ 0.000072). The most favourable results were seen in patients with low hepatic tumour load (<10% liver involvement). More recently, the phase III placebo-controlled CLARINET trial expanded the role of SSAs for tumour control in NET [40]. Within this protocol 204 patients with well- or moderately differentiated, nonfunctioning, somatostatin receptor-positive GEP-NETs with a Ki-67 of <10% were randomized to receive either lanreotide autogel 120 mg every 4 weeks or placebo. Patients had to have documented disease status during a three to six months preobservation period prior to randomization. The vast majority of patients (96%) had stable disease at randomization. Lanreotide was associated with a significant prolongation of PFS, with a median not reached versus a median of 18 months in the placebo arm (hazard ratio (HR) 0.47; p < 0.001). The estimated rates of PFS at 24 months were 65.1% in the lanreotide group and 33% in the placebo group. The benefit in the patients with midgut NET (HR 0.35; p ¼ 0.009) was greater than in the pancreatic subset (HR 0.58; p ¼ 0.06). It is important to note that the study was not powered for statistical significance of the subgroups. No benefit could be demonstrated for the small subgroup of patients with hindgut NET (HR 1.47). Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
Author, year
Na
Included patients
Progression at start documented?
Treatment
Tumour response (% of patients) PR
SD
Oberg et al., 1991 [78] Saltz et al., 1993 [79] Arnold et al., 1996 [67]
22 34 103
Metastatic midgut NET Metastatic GEPNET Metastatic GEPNET
Octreotide sc 50 mg bid to 200 mg tid Octreotide sc 50 mg bid to 250 mg tid Octreotide sc 200 mg tid
28 0 0
36 50 PD at entry:36.5 SD at entry: 53.8
Di Bartolomeo et al., 1996 [17] Wymenga et al., 1999 [66]
58 31
Mixed cohort, mainly midgut NET Functioning GI-NET
no yes PD prior to start in 52 pts, SD in 13pts, Unknown in 38pts yes no
3 6
47 81
Faiss et al., 1999 [80] Ducreux et al., 2000 [81] Ricci et al., 2000 [18] Faiss et al., 2003 [64] Arnold et al., 2005 [63]
24 39 25 25 48
yes yes no yes yes
4 5 8 4 2
46 49 40 28 46
Bajetta et al., 2005 [82]
31
no
Octreotide LAR im 30 mg q28d
6
52
Bajetta et al., 2006 [83]
28 each
Progressive, metastatic GEP-NET GI-NET Metastatic NET Progressive GEP-NET Progressive advanced midgut or pancreatic NET Treatment naïve well-differentiated NET Well-differentiated NET
Octreotide sc 500 mge1000 mg tid Lanreotide ATG 30 mg q2wk, escalated to weekly in 27% Lanreotide 5 mg SC tid Lanreotide 30 mg IM q10-14d Lanreotide 30 mg IM q2wk Lanreotide 1 mg SC tid Octreotide sc 200 mg tid
no
Panzuto et al., 2006 [68]
31
advanced pNET and intestinal
yes
Lanreotide ATG: 0 Lan LA: 4 0
Lanreotide ATG: 68 Lan LA: 64 45
Butturini et al., 2006 [84]
21
no
0
38
Rinke et al., 2009 [39] Khan et al., 2011 [21] Bianchi et al., 2011 [85] Martin-Richard et al., 2013 [69]
42 76 23 30
no no yes yes
Octreotide LAR 30 mg q28d Lanreotide ATG 60e120 mg q28d Lanreotide ATG 120 mg q28d Lanreotide ATG 120 mg q28d
2 0 8.7 4
67 75 at 3 years 65 89
Caplin et al., 2014 [40]
101
NA
65 at 24 months
28
96% SD prior treatment no
Lanreotide ATG 120 mg q28d
Cives M et al., 2015 [71]
Well-differentiated nonfunctioning pNET, somatostatin receptor positive Well-differentiated midgut Midgut with carcinoid syndrome Well-differentiated metastatic NET Advanced and/or metastatic, welldifferentiated NET Nonfunctioning somatostatin receptor positive GEPNET Treatment naïve grade 1 and 2 NET
Lanreotide LA 60 mg q3wk or Lanreotide ATG 120 mg q6wk (1:1) Octreotide LAR 30 mg q28d: 21patients Lanreotide SR 60 mg q28d: 10 patients Octreotide LAR 20 mg q28d
Pasireotide LAR 60 mg q28d
4
60
7
ATG ¼ autogel; bid ¼ two times daily; GEP ¼ gastroenteropancreatic; GI ¼ gastrointestinal; IM ¼ intramuscular; LA ¼ long acting; Lan ¼ lanreotide; NET ¼ neuroendocrine tumour(s); pNET ¼ pancreatic neuroendocrine tumour(s); pts ¼ patients; q ¼ every; SC ¼ subcutaneous; SR ¼ Slow release; tid ¼ three times daily. NA: not assessed. a N is for those patients with result for morphological tumour response.
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
Table 1 Studies with reported morphological response under SSA treatment (only trials with at least 20 patients included).
8
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
The main characteristics of both placebo-controlled trials are summarized in Table 2. Due to the small number of deaths, the crossover to open treatment and the impact of other poststudy treatments neither the PROMID trial nor the CLARINET trial was able to demonstrate a significant overall survival benefit. Therefore, it is a matter of debate whether there is still a place for a “watch and wait” approach or whether the treatment with SSA should start immediately to prevent tumour progression. Indirect data to support a possible overall survival benefit of SSA therapy originate from the Surveillance, Epidemiology and End Results (SEER) database: Longer survival durations have been shown in patients diagnosed from 1988 to 2004 if compared to survival durations in NET patients who were diagnosed in the pre-SSA period (1973e1987) [70]. The antiproliferative data for pasireotide are limited. In a recent phase II trial of pasireotide LAR 60 mg every four weeks in treatment naïve patients with NET G1 and G2 of different origins the PFS was 11 months [71]. Interestingly, the randomized trial in patients with refractory carcinoid syndrome which failed to demonstrate an advantage of pasireotide over high dose octreotide with regard to symptom control, showed a significant longer PFS for pasireotide compared to octreotide [24]. Further investigations of the antiproliferative capacity of pasireotide are warranted. IFN-a for tumour control After first reports of IFN-a (3e5 MU/d, 3e5/w sc) demonstrating notable anti-tumour effects in € neuroendocrine tumours [51,53], the Oberg's group published the largest series of IFN-a in 111 patients with metastatic NETs (75% midgut) with a disease control rate (DCR) of 54% (including 15% objective response (OR)) and median duration of response of 32 months [55]. Data for pNET patients are more limited with the largest retrospective cohort published again from the Uppsala group. They reported 12% and 24.5% OR and stabilization, respectively, in 57 pNET patients [72]. There are only few prospective data of IFN-a monotherapy available, placebo-controlled data are lacking. In the randomized GEPNET study of Faiss et al. DCR at 12 months was 29.6% [64]. In Dahan's trial the median PFS was 14.1 months with 1-year and 2-year PFS rates of 53% and 33%, respectively [62]. Combination treatment for tumour control The combination of SSA and IFN-a has been investigated in various non-randomized and five randomized trials over the past decades. Reasons for simultaneous administration were possible synergistic effects as well as better tolerability than IFN monotherapy. Four small non-randomized studies reported promising efficacy data of the combination treatment of SSA plus IFN-a after biochemical or morphological progression under SSA monotherapy: Biochemical response rates and disease control rates ranged from 62.5% to 77% and 21%e100%, respectively. OR was achieved in 4%e 36% [73e76].
Table 2 Comparison of the 2 placebo controlled randomized trials of SSA in well-differentiated NET.
Study drug and dose Primary tumour localization Number of patients included Functional status Preobservation period Histology SSTR positivity Primary end point Assessment of imaging Primary end point met?
PROMID
CLARINET
Ocreotide LAR 30 mg every 28 days versus placebo midgut and unknown 85 FNA and mild carcinoid syndrome no Well differentiated, >90% G1 Majority positive, but no inclusion criterion TTP WHO Yes
Lanreotide autogel 120 mg every 28 days versus placebo pNET, midgut, hindgut, unknown 204 FNA (and gastrinoma controlled by PPI) Yes, 96% SD before randomization G1 and G2 up to Ki67 10% Required for inclusion PFS RECIST 1.0 Yes
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
Author, year
Study design
n
Location Functionality Characteristics primary
€lby, 2003 [57] Ko
Randomized prospective 2 arms multicentric
68 midgut
Faiss, 2003 [64]
Open randomized prospective 3 arms imulticentric
80 GEPNET 29 (36.3%)
68 (100%)
105 GEPNET 47 (44.8%) Arnold, 2005 [63] Randomized prospective 2 arms multicentric
Yao, 2008 [61]
Randomized prospective phase II 2 arms cross-over design
Dahan, 2009 [62] Randomized prospective phase III 2 arms multicentric
44 GEPNET n.a.
64 GEPNET n.a.
Treatment
Octreotide 100 e200ug 2e3/ d sc ± IFN-a 3e5 MU 3e5/w sc Treatment naive; Lanreotide 1 mg PD at entry 3/d sc vs. IFN-a 5 MU 3/w sc vs. combination Prior primary tumour resection þ TAE
PFS þ OS
Symptomatic response
Biochemical response
Disease control rate
n.a.
5-HIAA overall P ¼ 0.005
45.7%vs. 81.8% PD 5-year survival rate 36.6 vs. 19 vs. 6 pts. 56.8% P ¼ 0.132 P ¼ 0.008
Serotonin þ CgA decreased sig. in all arms
32 vs. 29.6 vs. 25% PFS 1-year rate at 12 m 44% vs. 44.4% vs. 50%
Only combination achieved response P ¼ 0.037 for diarrhoea and flush PD at entry Octreotide 200ug Flushing, 3/d sc ± 4.5 MU diarrhoea and IFN-a 3/w sc abd. complaints improved between 33e83% Concomitant SSA Bevacizumab n.a. 15 mg/kg every (100%) therapy 3w iv vs. PEG IFN52.3% PD a-2b 0.5 mcg/kg 2/w sc Concomitant SSA 5-FU 400 mg/ Stool frequency m2 þ STZ (17.2%) 63 pts. and flushing 500 mg/m2 d1-5 decreased with prior PD every 6w iv vs. P ¼ 0.072 and IFN-a 3MU 3/w sc P ¼ 0.26 without differences
CgA 33.3 vs. 50% 45% vs. 50% at 3 m OS 35 vs. 51 m (P ¼ 0.55) (P ¼ 0.46) 5-HIAA 35.3 vs. 66.7% (P ¼ 0.22) Survival rates 5-HIAA 19 vs. 33% 95 vs. 68% P ¼ 0.33 CgA 6 vs. P ¼ 0.02 at week 1-year 93% 2-year 67% 18 14% P ¼ 0.43 3-year 56% mPFS 63w mPFS 5.5 m vs. 59 vs. 72% Biochemical 14.1 m P ¼ 0.12 progression 22 (midguts: 65 vs. P ¼ 0.34 vs. 9% mOS 88% P ¼ 0.31) 30.4 m vs. 44.3 m P ¼ 0.83
GEPNET ¼ gastroenteropancreatic-neuroendocrine tumours; TAE ¼ transarterial embolization; n.a. ¼ not assessed; PD ¼ progressive disease; SSA ¼ somatostatin analogues; vs ¼ versus; w ¼ week; d ¼ day; m ¼ months; sc ¼ subcutaneous; iv ¼ intravenous; 5-HIAA ¼ 5-hydroxyindoleacetic acid; CgA ¼ chromogranin A; (m)PFS ¼ (median) progression-free survival; (m) OS ¼ (median) overall survival.
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
Table 3 Prospective randomized trials with IFN-a in NETs.
9
10
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
Encouraged by these results three randomized trials were initiated to further evaluate the role of SSA and IFN-a combination treatment compared to monotherapies. As described above, the first €lby, exclusively focused on metastatic midgut NETs with carcinoid randomized trial, published by Ko syndrome (n ¼ 68) [57]. Intention-to-treat analysis showed no significant differences in 5-year survival rates (36.6% octreotide versus 56.8% combination, HR 0.62, P ¼ 0.132). However, the risk of tumour progression was lower in the combination treatment (HR 0.28, P ¼ 0.008). The three-arm study of Faiss included 80 progressive GEPNET patients [64]. No difference in PFS rate after 1 year was demonstrated: 56%, 55% and 50% for lanreotide, IFN-a and the combination group, respectively (P ¼ 0.69). The largest randomized trial (n ¼ 109) to explore the role of combined treatment (octreotide 200 mg 3/d ± 4.5 MU IFN-a 3/w sc) was published by our group [63]. Only progressive duodenopancreatic and midgut NETs were included. Median time to treatment failure was 6 months in both treatment arms (P ¼ 0.59). Response to treatment after 12 months exhibited no significant differences (2.0% vs. 9.3%, stable disease 15.8% vs. 14.7% and progressive disease 49% vs. 25.9%). Additionally, median survival with 35 months for mono- and 51 months for combination therapy was not statistically different (P ¼ 0.55). The data of the randomized trials are summarized in Table 3. Although nowadays in clinical practice pegylated IFN-a is used more frequently than IFN-a, there is insufficient data published to draw conclusions on its value in NETs. One small study of 17 GEPNET patients treated with pegylated IFN-a2b in combination with SSA showed promising results with 2 partial responses and 11 stabilizations. Tolerability of pegylated IFN-a2b seemed to be better than IFNa 2b [77]. In a second phase II study 22 patients received PEG IFN-a-2b 0.5 mg/kg 2/w sc with concomitant octreotide LAR (versus bevacizumab þ octreotide). This combination achieved 15 disease stabilizations (68%), while 6 patients progressed (27%). The median PFS was 56 weeks [61]. Insufficient statistical power of the randomized trials, heterogeneity of patient cohorts and different treatment schedules as well as methods of response evaluation hinders a final conclusion. As IFN-a adds toxicity and clear superiority of the combination treatment could not been demonstrated, the combination treatment is not recommended as first line therapy.
Research agenda for biotherapy Conduction of a phase III trial of pasireotide versus octreotide or lanreotide in midgut tumours Assessment of the role of pegylated IFN-a in patients with midgut tumours Comparison of SSA plus IFN-a versus SSA monotherapy in patients with metastatic midgut tumours in a statistically adequate fashion Evaluate high dose SSA in antiproliferative intention
Practice points biotherapy in antiproliferative purpose SSA exert antiproliferative action by delaying tumour progression in patients with metastasized well-differentiated and low proliferative (Ki67 < 10%) GEPNETs SSA treatment rarely leads to objective tumour response SSA have a favourable toxicity profile IFN-a treatment can induce tumour stabilization and e in a minority of patients e objective tumour response The combination therapy of SSA and IFN-a is not recommended as first line treatment since no advantage of the combination treatment as compared to monotherapy has been demonstrated
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
11
Summary Somatostatin analogues (SSA) and IFN-a have favourable antisecretory and antiproliferative effects in well-differentiated neuroendocrine tumours regardless of the origin of primary tumour site. Both biotherapeutic agents obtain similar results for symptomatic response and disease-control rate. However, due to the better tolerability of SSA and the landmark studies PROMID and CLARINET, SSA are considered to be first choice for functioning tumours as well as non-functioning tumours. Although the clinical use of IFN-a is restricted, its clinical impact as salvage therapy in patients not responding to SSA or progressive during SSA treatment should not be underestimated. So far, the combination of SSA and IFN-a did not present any statistically significant differences in prospective randomized trials. But to definitely clarify this issue well-designed randomized trials are needed with pegylated IFN-a and pasireotide being incorporated. To this end, biotherapies provide an ideal platform to investigate novel drugs and treatment approaches to receive synergistic or additive effects. Therefore, future trials should be measured against the clinical benefit of biotherapies. Conflict of interest AR has received travel grants for scientific meetings by Ipsen and Pfizer, honoraria for presentations by Novartis and Ipsen and was member of advisory boards of Ipsen, Novartis and Pfizer. SK has received travel grants for scientific meetings by Ipsen and Novartis. Acknowledgement We thank Patrick Michl for critical revision of the manuscript. References [1] Brazeau P, Vale W, Burgus R, et al. Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science 1973;179:77e9. *[2] Modlin IM, Pavel M, Kidd M, et al. Review article: somatostatin analogues in the treatment of gastroenteropancreatic neuroendocrine (carcinoid) tumours. Aliment Pharmacol Ther 2010;31:169e88. [3] Caplin M, Sundin A, Nillson O, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms: colorectal neuroendocrine neoplasms. Neuroendocrinology 2012;95:88e97. €ssler D, et al. Identification of somatostatin receptor subtypes 1, 2A, 3, and 5 in neuroendocrine [4] Kulaksiz H, Eissele R, Ro tumours with subtype specific antibodies. Gut 2002;50:52e60. €nger J, et al. Somatostatin receptors in bronchopulmonary neuroendocrine neoplasms: new [5] Kaemmerer D, Specht E, Sa diagnostic, prognostic, and therapeutic markers. J Clin Endocrinol Metab 2015;100:831e40. [6] Bauer W, Briner U, Doepfner W, et al. SMS 201-995: a very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sci 1982;31:1133e40. [7] Bronstein M, Musolino N, Jallad R, et al. Pharmacokinetic profile of lanreotide autogel in patients with acromegaly after four deep subcutaneous injections of 60, 90 or 120 mg every 28 days. Clin Endocrinol (Oxf) 2005;63:514e9. [8] Schmid HA, Schoeffter P. Functional activity of the multiligand analog SOM230 at human recombinant somatostatin receptor subtypes supports its usefulness in neuroendocrine tumors. Neuroendocrinology 2004;80(Suppl. 1):47e50. [9] Weckbecker G, Lewis I, Albert R, et al. Opportunities in somatostatin research: biological, chemical and therapeutic aspects. Nat Rev Drug Discov 2003;2:999e1017. [10] Guillermet J, Saint-Laurent N, Rochaix P, et al. Somatostatin receptor subtype 2 sensitizes human pancreatic cancer cells to death ligand-induced apoptosis. Proc Natl Acad Sci U S A 2003;100:155e60. *[11] Susini C, Buscail L. Rationale for the use of somatostatin analogs as antitumor agents. Ann Oncol 2006;17:1733e42. [12] Eriksson B, Oberg K. Summing up 15 years of somatostatin analog therapy in neuroendocrine tumors: future outlook. Ann Oncol 1999;10(Suppl. 2):S31e8. [13] Hofland LJ, Lamberts SW. The pathophysiological consequences of somatostatin receptor internalization and resistance. Endocr Rev 2003;24:28e47. [14] Kvols LK, Moertel CG, O'Connell MJ, et al. Treatment of the malignant carcinoid syndrome. Evaluation of a long-acting somatostatin analogue. N Engl J Med 1986;315:663e6. [15] Rubin J, Ajani J, Schirmer W, et al. Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol 1999;17:600e6. [16] O'Toole D, Ducreux M, Bommelaer G, et al. Treatment of carcinoid syndrome: a prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient acceptability, and tolerance. Cancer 2000;88:770e6. [17] di Bartolomeo M, Bajetta E, Buzzoni R, et al. Clinical efficacy of octreotide in the treatment of metastatic neuroendocrine tumors. A study by the Italian trials in medical oncology group. Cancer 1996;77:402e8. [18] Ricci S, Antonuzzo A, Galli L, et al. Long-acting depot lanreotide in the treatment of patients with advanced neuroendocrine tumors. Am J Clin Oncol 2000;23:412e5.
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
12
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
[19] Ruszniewski P, Ish-Shalom S, Wymenga M, et al. 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 2004;80:244e51. [20] Toumpanakis C, Garland J, Marelli L, et al. Long-term results of patients with malignant carcinoid syndrome receiving octreotide LAR. Aliment Pharmacol Ther 2009;30:733e40. [21] Khan MS, El-Khouly F, Davies P, et al. Long-term results of treatment of malignant carcinoid syndrome with prolonged release lanreotide (Somatuline Autogel). Aliment Pharmacol Ther 2011;34:235e42. [22] Ruszniewski PB, Caplin ME, Valle JW, et al. Patient-reported satisfaction with symptom control during lanreotide autogel/ depot (LAN) treatment for carcinoid syndrome (CS) in gastroenteropancreatic neuroendocrine tumor (GEP-NET) patients: symnet, a large multinational, cross-sectional, observational study. ASCO Meet Abstr 2014;32:273. [23] Kvols LK, Oberg KE, O'Dorisio TM, et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study. Endocr Relat Cancer 2012;19:657e66. *[24] Edward M, Wolin EM, Jarzab B, Eriksson B, et al. A multicenter, randomized, blinded, phase III study of pasireotide LAR versus octreotide LAR in patients with metastatic neuroendocrine tumors (NET) with disease-related symptoms inadequately controlled by somatostatin analogs. In: JCO; 2013. [25] Dobson R, Burgess MI, Banks M, et al. The association of a panel of biomarkers with the presence and severity of carcinoid heart disease: a cross-sectional study. PLoS One 2013;8:e73679. €ppel G, Krenning E, et al. Consensus guidelines for the management of patients with digestive neuro[26] Eriksson B, Klo endocrine tumorsewell-differentiated jejunal-ileal tumor/carcinoma. Neuroendocrinology 2008;87:8e19. [27] Ramage JK, Ahmed A, Ardill J, et al. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut 2012;61:6e32. [28] Papotti M, Bongiovanni M, Volante M, et al. 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 Arch 2002;440:461e75. [29] Healy ML, Dawson SJ, Murray RM, et al. Severe hypoglycaemia after long-acting octreotide in a patient with an unrecognized malignant insulinoma. Intern Med J 2007;37:406e9. [30] Saijo F, Naito H, Funayama Y, et al. Octreotide in control of multiple liver metastases from gastrinoma. J Gastroenterol 2003;38:905e8. [31] Jensen RT, Niederle B, Mitry E, et al. Gastrinoma (duodenal and pancreatic). Neuroendocrinology 2006;84:173e82. [32] Santangelo WC, O'Dorisio TM, Kim JG, et al. Pancreatic cholera syndrome: effect of a synthetic somatostatin analog on intestinal water and ion transport. Ann Intern Med 1985;103:363e7. [33] Nikou GC, Toubanakis C, Nikolaou P, et al. VIPomas: an update in diagnosis and management in a series of 11 patients. Hepatogastroenterology 2005;52:1259e65. [34] Altimari AF, Bhoopalam N, O'Dorsio T, et al. Use of a somatostatin analog (SMS 201-995) in the glucagonoma syndrome. Surgery 1986;100:989e96. [35] Rosenbaum A, Flourie B, Chagnon S, et al. Octreotide (SMS 201-995) in the treatment of metastatic glucagonoma: report of one case and review of the literature. Digestion 1989;42:116e20. [36] O'Toole D, Salazar R, Falconi M, et al. Rare functioning pancreatic endocrine tumors. Neuroendocrinology 2006;84: 189e95. [37] Falhammar H. Cyclic ectopic cushing's syndrome and somatostatin analogue treatment. N Z Med J 2009;122:92e5. *[38] Caplin ME, Baudin E, Ferolla P, et al. Pulmonary neuroendocrine (carcinoid) tumors: European neuroendocrine tumor society expert consensus and recommendations for best practice for typical and atypical pulmonary carcinoids. Ann Oncol 2015 Aug;26(8):1604e20. *[39] Rinke A, Müller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID study Group. J Clin Oncol 2009;27:4656e63. [40] Caplin ME, Pavel M, Ruszniewski P. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014; 371:1556e7. [41] Oberg K, Kvols L, Caplin M, et al. Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Ann Oncol 2004;15:966e73. [42] Isaacs A, Lindenmann J. Virus interference. I. The interferon. By A. Isaacs and J. Lindenmann, 1957. J Interferon Res 1987;7: 429e38. [43] Pestka S, Krause CD, Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev 2004;202:8e32. [44] Schindler C, Levy DE, Decker T. JAK-STAT signaling: from interferons to cytokines. J Biol Chem 2007;282:20059e63. [45] Hanley JP, Haydon GH. The biology of interferon-alpha and the clinical significance of anti-interferon antibodies. Leuk Lymphoma 1998;29:257e68. [46] Pestka S. The interferon receptors. Semin Oncol 1997;24. S9-18-S19-40. [47] Zhou Y, Wang S, Gobl A, et al. Interferon alpha induction of Stat1 and Stat2 and their prognostic significance in carcinoid tumors. Oncology 2001;60:330e8. [48] Wang S, Tyring SK, Townsend CM, et al. Interferon-mediated activation of the STAT signaling pathway in a human carcinoid tumor. Ann Surg Oncol 1998;5:642e9. [49] Detjen KM, Welzel M, Farwig K, et al. Molecular mechanism of interferon alfa-mediated growth inhibition in human neuroendocrine tumor cells. Gastroenterology 2000;118:735e48. [50] Zitzmann K, Brand S, De Toni EN, et al. SOCS1 silencing enhances antitumor activity of type I IFNs by regulating apoptosis in neuroendocrine tumor cells. Cancer Res 2007;67:5025e32. [51] Oberg K, Funa K, Alm G. Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. N Engl J Med 1983;309:129e33. [52] Moertel CG, Rubin J, Kvols LK. Therapy of metastatic carcinoid tumor and the malignant carcinoid syndrome with recombinant leukocyte A interferon. J Clin Oncol 1989;7:865e8.
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004
A. Rinke, S. Krug / Best Practice & Research Clinical Endocrinology & Metabolism xxx (2015) 1e13
13
[53] Oberg K, Norheim I, Lind E, et al. Treatment of malignant carcinoid tumors with human leukocyte interferon: long-term results. Cancer Treat Rep 1986;70:1297e304. [54] Oberg K, Norheim I, Alm G. Treatment of malignant carcinoid tumors: a randomized controlled study of streptozocin plus 5-FU and human leukocyte interferon. Eur J Cancer Clin Oncol 1989;25:1475e9. *[55] Oberg K, Eriksson B. The role of interferons in the management of carcinoid tumors. Acta Oncol 1991;30:519e22. [56] Eriksson B, Skogseid B, Lundqvist G, et al. Medical treatment and long-term survival in a prospective study of 84 patients with endocrine pancreatic tumors. Cancer 1990;65:1883e90. €lby L, Persson G, Franze n S, et al. Randomized clinical trial of the effect of interferon alpha on survival in patients with *[57] Ko disseminated midgut carcinoid tumours. Br J Surg 2003;90:687e93. [58] Jensen RT, Cadiot G, Brandi ML, et al. ENETS consensus guidelines for the management of patients with digestive neuroendocrine neoplasms: functional pancreatic endocrine tumor syndromes. Neuroendocrinology 2012;95:98e119. [59] Pavel M, Baudin E, Couvelard A, et al. ENETS consensus guidelines for the management of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 2012;95:157e76. [60] Oberg K. Interferons in the management of neuroendocrine tumors and their possible mechanism of action. Yale J Biol Med 1992;65:519e29. discussion 531e516. [61] Yao JC, Phan A, Hoff PM, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol 2008; 26:1316e23. [62] Dahan L, Bonnetain F, Rougier P, et al. Phase III trial of chemotherapy using 5-fluorouracil and streptozotocin compared with interferon alpha for advanced carcinoid tumors: FNCLCC-FFCD 9710. Endocr Relat Cancer 2009;16:1351e61. *[63] Arnold R, Rinke A, Klose K, et al. Octreotide versus octreotide plus interferon-alpha in endocrine gastroenteropancreatic tumors: a randomized trial. Clin Gastroenterol Hepatol 2005;3:761e71. €hmig M, et al. Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, *[64] Faiss S, Pape UF, Bo interferon alfa, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumorsethe international lanreotide and interferon alfa study Group. J Clin Oncol 2003;21:2689e96. [65] Clements D, Elias E. Regression of metastatic vipoma with somatostatin analogue SMS 201-995. Lancet 1985;1:874e5. [66] Wymenga AN, Eriksson B, Salmela PI, et al. Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuroendocrine tumors and hormone-related symptoms. J Clin Oncol 1999;17:1111. [67] Arnold R, Trautmann ME, Creutzfeldt W, et al. Somatostatin analogue octreotide and inhibition of tumour growth in metastatic endocrine gastroenteropancreatic tumours. Gut 1996;38:430e8. [68] Panzuto F, Di Fonzo M, Iannicelli E, et al. Long-term clinical outcome of somatostatin analogues for treatment of progressive, metastatic, well-differentiated entero-pancreatic endocrine carcinoma. Ann Oncol 2006;17:461e6. *[69] Martín-Richard M, Massutí B, Pineda E, et al. Antiproliferative effects of lanreotide autogel in patients with progressive, well-differentiated neuroendocrine tumours: a Spanish, multicentre, open-label, single arm phase II study. BMC Cancer 2013;13:427. [70] Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008;26:3063e72. [71] Cives M, Kunz PL, Morse B, et al. Phase II clinical trial of pasireotide long-acting repeatable in patients with metastatic neuroendocrine tumors. Endocr Relat Cancer 2015;22:1e9. [72] Eriksson B, Oberg K. An update of the medical treatment of malignant endocrine pancreatic tumors. Acta Oncol 1993;32: 203e8. € m H, Andersson T, et al. Octreotide and interferon alfa: a new combination for the treatment [73] Tiensuu Janson EM, Ahlstro of malignant carcinoid tumours. Eur J Cancer 1992;28A:1647e50. [74] Frank M, Klose KJ, Wied M, et al. Combination therapy with octreotide and alpha-interferon: effect on tumor growth in metastatic endocrine gastroenteropancreatic tumors. Am J Gastroenterol 1999;94:1381e7. [75] Artale S, Giannetta L, Cerea G, et al. Treatment of metastatic neuroendocrine carcinomas based on WHO classification. Anticancer Res 2005;25:4463e9. €llskog ML, Sundin A, Westlin JE, et al. Treatment of malignant endocrine pancreatic tumors with a combination of [76] Fja alpha-interferon and somatostatin analogs. Med Oncol 2002;19:35e42. [77] Pavel ME, Baum U, Hahn EG, et al. Efficacy and tolerability of pegylated IFN-alpha in patients with neuroendocrine gastroenteropancreatic carcinomas. J Interferon Cytokine Res 2006;26:8e13. [78] Oberg K, Norheim I, Theodorsson E. Treatment of malignant midgut carcinoid tumours with a long-acting somatostatin analogue octreotide. Acta Oncol 1991;30:503e7. [79] Saltz L, Trochanowski B, Buckley M, et al. Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer 1993;72:244e8. €th U, Mansmann U, et al. Ultra-high-dose lanreotide treatment in patients with metastatic neuroendocrine [80] Faiss S, Ra gastroenteropancreatic tumors. Digestion 1999;60:469e76. [81] Ducreux M, Ruszniewski P, Chayvialle JA, et al. The antitumoral effect of the long-acting somatostatin analog lanreotide in neuroendocrine tumors. Am J Gastroenterol 2000;95:3276e81. [82] Bajetta E, Catena L, Procopio G, et al. Is the new WHO classification of neuroendocrine tumours useful for selecting an appropriate treatment? Ann Oncol 2005;16:1374e80. [83] Bajetta E, Procopio G, Catena L, et al. Lanreotide autogel every 6 weeks compared with lanreotide microparticles every 3 weeks in patients with well differentiated neuroendocrine tumors: a phase III study. Cancer 2006;107:2474e81. [84] Butturini G, Bettini R, Missiaglia E, et al. Predictive factors of efficacy of the somatostatin analogue octreotide as first line therapy for advanced pancreatic endocrine carcinoma. Endocr Relat Cancer 2006;13:1213e21. [85] Bianchi A, De Marinis L, Fusco A, et al. The treatment of neuroendocrine tumors with long-acting somatostatin analogs: a single center experience with lanreotide autogel. J Endocrinol Invest 2011;34:692e7.
Please cite this article in press as: Rinke A, Krug S, Neuroendocrine tumours e Medical therapy: Biological, Best Practice & Research Clinical Endocrinology & Metabolism (2015), http://dx.doi.org/10.1016/ j.beem.2015.09.004