Abstract
Traditional therapies have offered patients with advanced gastrointestinal neuroendocrine tumors limited benefit. Selected patients with hepatic metastases may benefit from surgical debulking, embolization, or other ablative therapies. While somatostatin analogs are highly effective in controlling symptoms of hormonal secretion, they are only rarely associated with tumor regression. The clinical benefit associated with the administration of systemic agents such as interferon-α or cytotoxic chemotherapy is less clear, and the widespread use of such regimens has been limited by their relatively modest anti-tumor activity, as well as concerns regarding their potential toxicity. The mixed clinical results seen with these agents in neuroendocrine tumors have led to great interest in the development of novel treatment approaches for patients with advanced disease. Recent clinical studies of novel agents, particularly those targeting the vascular endothelial growth factor pathway and mammalian target of rapamycin, have demonstrated promising activity in patients with advanced neuroendocrine tumors. Ongoing randomized studies should help better define the role these and other targeted agents will play in the future treatment of patients with this disease.
Introduction
Neuroendocrine tumors comprise a diverse group of malignancies and, when defined broadly, include not only gastrointestinal neuroendocrine tumors but also pheochromocytoma, thyroid cancer, and even small cell lung cancer. Gastrointestinal neuroendocrine tumors are usually subclassified as either pancreatic neuroendocrine tumors or carcinoid tumors. The treatment approaches for both tumor types share many similarities due to their often indolent behavior, characteristic well-differentiated histologic features, and shared ability to secrete neuropeptides resulting in characteristic clinical syndromes.
In contrast to most other malignancies, there is no formal staging system for gastrointestinal neuroendocrine tumors. The absence of such a staging system in part reflects the straightforward treatment approach to localized neuroendocrine tumors, which is almost always surgical resection (Fig. 1 ). Adjuvant therapy has no established role in this disease, even in cases where local lymph nodes are involved and the risk of recurrence is felt to be high. Unfortunately, curative surgery is seldom an option for patients with metastatic disease, and standard cytotoxic therapy offers limited benefit. A number of other treatment approaches, including the use of somatostatin analogs, surgical debulking, and ablation of hepatic metastases offer palliative benefit for such patients. Novel treatment approaches, including the use of agents targeting vascular endothelial growth factor (VEGF) and other pathways involved in neuroendocrine tumorigenesis, may provide additional therapeutic options.
Clinical presentation and diagnosis of metastatic disease
Whereas some patients with advanced neuroendocrine tumors may remain symptom-free for years, others develop symptoms from either tumor bulk or hormonal hypersecretion. In patients with metastatic carcinoid tumors, the secretion of serotonin and other vasoactive substances may cause carcinoid syndrome, which is manifested by episodic flushing, wheezing, diarrhea, and eventual right-sided valvular heart disease ( Thorson et al. 1954). The best-characterized syndromes associated with pancreatic neuroendocrine tumors are those associated with insulinoma, glucagonoma, vasoactive intestinal peptideoma, and gastrinoma. The so-called ‘non-functioning’ pancreatic neuroendocrine tumors often present at a late stage, and may be associated with high serum levels of pancreatic polypeptide.
The predominant site of metastatic spread in patients with neuroendocrine tumors involving the gastrointestinal tract is the liver. Patients with suspected metastatic disease are generally evaluated with an abdominal computerized tomography (CT) scan to rule out liver metastases. Liver function tests are unreliable indicators of tumor involvement, and serum alkaline phosphatase levels are frequently normal despite extensive liver involvement by carcinoid tumor. Carcinoid liver metastases are often hypervascular and may become isodense relative to the liver with the administration of intravenous contrast materials. CT scans should thus be performed both before and after the administration of intravenous contrast agents ( Sugimoto et al. 1995, Woodard et al. 1995).
Somatostatin receptor scintigraphy provides another useful imaging modality for the detection of metastatic disease in patients with neuroendocrine tumors. With the exception of insulinomas (of which only 50% express type-2 somatostatin receptors), over 90% of neuroendocrine tumors, including non-functioning pancreatic tumors and carcinoid tumors, contain high concentrations of somatostatin receptors, and can be imaged with a radiolabeled form of the somatostatin analog octreotide (111-indium pentetreotide; Lamberts et al. 1990, Kvols et al. 1993, Kaltsas et al. 2001). The uptake of radiolabeled octreotide is also predictive of a clinical response to therapy with somatostatin analogs ( Lamberts et al. 1990).
Serial measurement of the serotonin metabolite 5-hydroxyindoleacetic acid (HIAA) in 24-h urine collections has been commonly used in the diagnosis and subsequent monitoring of patients with metastatic carcinoid tumors. Although elevated urinary 5-HIAA levels are highly specific for carcinoid tumors, they are not particularly sensitive; in one study, only 73% of patients with metastatic carcinoid tumors had elevated levels ( Feldman & O’Dirisio 1986). Furthermore, 5-HIAA levels are generally elevated in patients with metastatic midgut carcinoid tumors, but are less useful in patients with either foregut (bronchial, gastric) or hindgut (rectal) carcinoid tumors, which less commonly secrete serotonin.
Chromogranin A (CGA) is a 49 kDa protein that is contained in the neurosecretory vesicles of neuroendocrine tumor cells, and has been identified in the plasma of patients with endocrine neoplasms. Plasma CGA concentrations are a more sensitive marker than urinary 5-HIAA levels in patients with carcinoid tumors, and can also be used as a marker in patients with both functional and non-functional pancreatic endocrine tumors ( Seregni et al. 2001, Stivanello et al. 2001, Tomassetti et al. 2001, Oberg et al. 2004). CGA concentrations should be used with caution as a marker of disease activity in patients treated with somatostatin analogs, since these agents significantly reduce plasma CGA levels ( Oberg et al. 2004). In such cases, changes in CGA concentrations may be more reflective of alterations in hormonal synthesis and release from tumor cells than an actual reduction in tumor mass. In patients on stable doses of somatostatin analogs, consistent increases in plasma CGA levels over time may reflect loss of secretory control and/or tumor growth. Plasma CGA levels have also been shown to have prognostic value; in one series of 71 patients with metastatic carcinoid tumors, CGA levels of more than 5000 μg/ml were independently associated with poor prognosis. ( Janson et al. 1997)
Surgical options
In selected cases, metastatic liver disease can be surgically resected. In one large surgical series involving 170 patients undergoing hepatic resection, more than 90% achieved improvement in symptoms. While the recurrence rate following surgery was high (84%), the 5- and 10-year survival rates were encouraging (61 and 35% respectively; Sarmiento et al. 2003). Several retrospective surgical series have suggested that patients who undergo either complete resection or aggressive ‘debulking’ of hepatic metastases have improved quality of life and improved survival times when compared with patients who do not undergo surgery.( Knox et al. 2004, Touzios et al. 2005, Musunuru et al. 2006, Osborne et al. 2006) In one such series, patients undergoing cytoreductive surgery had a mean survival duration of 43 months, as compared with 24 months for patients undergoing embolization ( Osborne et al. 2006). Similarly, in one study, resection of the primary tumor in patients with documented liver metastases was associated with improved survival ( Givi et al. 2006). While many of these series match patients retrospectively according to tumor bulk and other clinical parameters, the lack of formal randomization and potential for selection bias makes definitive interpretation of these results difficult.
The number of patients with liver-isolated metastatic disease in whom orthotopic liver transplantation (OLT) has been attempted is small, and the role of OLT in such patients remains unclear ( Alsina et al. 1990, Lang et al. 1997, Le Treut et al. 1997, van Vilsteren et al. 2006). Results from one multicenter study demonstrated a 5-year overall survival rate of 69% for patients with carcinoid tumors ( Le Treut et al. 1997). Despite high survival rates in selected patients, however, the majority of patients undergoing transplant ultimately appear to develop recurrent disease ( Le Treut et al. 1997, Florman et al. 2004). The limited availability of orthotopic transplants in many regions has also limited the widespread use of this procedure.
Non-surgical, hepatic-directed therapy
Hepatic artery embolization
Hepatic arterial embolization is commonly used as a palliative technique in patients with hepatic metastases, who are not candidates for surgical resection. Hepatic artery embolization is based on the principle that tumors in the liver derive most of their blood supply from the hepatic artery, whereas healthy hepatocytes derive most of their blood supply from the portal vein. The response rates associated with embolization, as measured either by a decrease in hormonal secretion or by radiographic regression, are generally >50% ( Ajani et al. 1988, Ruszniewski et al. 1993, Moertel et al. 1994, Diamandidou et al. 1998, Drougas et al. 1998, Eriksson et al. 1998, Brown et al. 1999, Dominguez et al. 2000, Gupta et al. 2003, Loewe et al. 2003). However, the duration of response can be brief, ranging from 4 to 51 months in uncontrolled patient series ( Moertel et al. 1994, Gupta et al. 2003). In one of the largest series of 81 patients undergoing embolization or chemoembolization for carcinoid tumors, the median duration of response was 17 months, and the probability of progression-free survival at 1, 2, and 3 years was 75, 35, and 11% respectively ( Gupta et al. 2003). Early studies reported a significant incidence of post-embolization complications that included renal failure, hepatic necrosis, and sepsis. Improved techniques have, in recent years, reduced the incidence of such complications, making embolization an important and generally safe treatment option for patients with neuroendocrine tumors ( Gupta et al. 2003).
Radiofrequency ablation (RFA) and cryoablation
Other approaches to the treatment of hepatic metastases include the use of RFA and cryoablation, either alone or in conjunction with surgical debulking. While these approaches appear to be less morbid than either hepatic resection or hepatic artery embolization, their efficacy, particularly in patients with large volume hepatic disease, has not been well studied. Most published reports are small case studies of fewer than 40 patients ( Chung et al. 2001, Berber et al. 2002, Gulec et al. 2002, Hellman et al. 2002).
Systemic treatment
Somatostatin analogs and interferon (IFN)-α
Carcinoid syndrome, as well as other hormonal syndromes associated with pancreatic neuroendocrine tumors, can often be well controlled with somatostatin analogs. In an initial study, the subcutaneous administration of the somatostatin analog octreotide, administered at a dosage of 150 μcg three times a day, improved the symptoms of carcinoid syndrome in 88% of patients ( Kvols et al. 1986). More recently, the use of a long-acting depot form of octreotide, which can be administered on a monthly basis, has largely obviated the need for patients to inject themselves on a daily basis. Long-acting octreotide is typically initiated at a dose of 20 mg IM after a brief trial of the short-acting formulation, with gradual escalation of the dose as needed for optimal control of symptoms ( Rubin et al. 1999, Oberg et al. 2004). Patients may also use additional short-acting octreotide for breakthrough symptoms. Lanreotide, another somatostatin analog, appears to be similar to octreotide in its clinical efficacy ( Faiss et al. 1999, Wymenga et al. 1999, Ducreux et al. 2000, O’Toole et al. 2000, Faiss et al. 2003). Somatostatin analogs are well tolerated by patients and studies report only mild and occasional toxicities, including hyperglycemia, steatorrhea, an increased risk of cholelithiasis, and irritation at the injection site.
The ability of leukocyte IFN to stimulate T-lymphocyte function and control the secretion of tumor products led to its initial use in patients with carcinoid syndrome ( Oberg et al. 1983). The addition of IFN-α to therapy with somatostatin analogs has subsequently been reported to be effective in controlling symptoms in patients with carcinoid syndrome, who may be resistant to somatostatin analogs alone ( Janson & Oberg 1993, Frank et al. 1999). Therapy with low-dose IFN-α has been reported to result in biochemical responses in ~40% of patients with metastatic neuroendocrine tumors ( Oberg & Eriksson 1991). The more widespread acceptance of IFN-α in the treatment of metastatic neuroendocrine tumors has been limited by studies challenging its efficacy, as well as the potential for side effects, which may include myelosuppression, fatigue, depression, and alteration of thyroid function ( Valimaki et al. 1991).
Whether somatostatin analogs have a direct cytostatic effect, either alone or when combined with IFN-α, is controversial ( Saltz et al. 1993). Radiologic evidence of tumor regression following treatment with these agents is rare. In a small study involving 21 patients with metastatic gastroenteropancreatic neuroendocrine tumors, a combined regimen of a somatostatin analog and IFN-α appeared to significantly slow the rate of tumor progression in 67% of patients during follow-up ( Frank et al. 1999). In a prospective study of 68 patients randomized to receive either octreotide alone or a combination of octreotide and IFN-α, there was no significant difference in the overall survival; however, patients treated with the combination regimen had a reduced risk of tumor progression, suggesting that IFN-α had a cytostatic effect ( Kolby et al. 2003). The efficacy of lanreotide, IFN-α, or combined therapy was evaluated in a prospective randomized trial involving 80 therapy-naive patients with documented progressive metastatic neuroendocrine tumors ( Faiss et al. 2003). The rates of objective partial response (4, 4, and 7% for lanreotide, IFN-α, and combined therapy respectively) were low in all three groups; however, all treatments resulted in apparent disease stabilization in a higher proportion of patients (28, 26, and 18% for lanreotide, IFN-α, and combined therapy respectively).
Cytotoxic chemotherapy
The efficacy of cytotoxic chemotherapy in patients with carcinoid tumors has been relatively limited (Table 1 ). In an initial trial, the Eastern Cooperative Oncology Group (ECOG) randomized 118 patients to receive streptozocin combined with either fluorouracil or cyclophosphamide ( Moertel & Hanley 1979). Response rates, as measured either by tumor regression or a decrease in urinary 5-HIAA levels, were 33% in the streptozocin and fluorouracil arm and 26% in the streptozocin and cyclophosphamide arm. There were no significant differences in survival between the two groups. In a subsequent trial, the dosing interval between cycles of streptozocin and fluorouracil was increased, and this treatment was compared with that of doxorubicin alone ( Engstrom et al. 1984). With this revised schedule, the response rate for streptozocin and fluorouracil was 22%, as compared with 21% for doxorubicin alone; again there were no significant differences in survival. Most recently, streptozocin and fluorouracil was compared with doxorubicin and fluorouracil in a randomized trial of 249 patients ( Sun et al. 2005). The response rates associated with the two regimens were similar (16% vs 15.9%). Although there was a slight survival benefit associated with streptozocin and fluorouracil (24.3 vs 15.7 months) in this trial, over one-third of patients treated with streptozocin developed renal toxicity. This and other toxicities, combined with only modest efficacy, has precluded the common use of streptozocin-based regimens as a first-line treatment for advanced carcinoid disease.
Several studies suggest that pancreatic endocrine tumors are more responsive to chemotherapy than are carcinoid tumors (Table 2 ). In an initial randomized trial, Moertel et al.(1992) reported that the combination of streptozocin and doxorubicin was associated with a combined biochemical and radiologic response rate of 69% and a median overall survival time of 2.2 years. Two subsequent retrospective analyses of patients receiving this regimen questioned the high response rate of this initial trial, and reported objective radiologic response rates, using modern response criteria, of <10% ( Cheng & Saltz 1999, McCollum et al. 2004). A larger retrospective analysis of 84 patients with either locally advanced or metastatic pancreatic endocrine tumors receiving a three-drug regimen of streptozocin, fluorouracil, and doxorubicin showed that this regimen was associated with an overall response rate of 39% and a median survival duration of 37 months, suggesting that this combination is indeed active, though perhaps less active than Moertel’s data suggested initially ( Kouvaraki et al. 2004). This study reported grade 3 or 4 toxicities in 23% of the patient cohort, with myelosuppression, mucositis, diarrhea, and fatigue the most frequently reported adverse reactions.
Dacarbazine (DTIC) has been evaluated as a potential alternative to streptozocin-based therapy in both carcinoid and pancreatic endocrine tumors. The ECOG performed a phase II study of DTIC in 50 patients with advanced pancreatic islet cell carcinoma and reported an objective response rate of 34% (Table 2 ; Ramanathan et al. 2001). A Southwest Oncology Group (SWOG) study reported that treatment with DTIC was associated with an objective radiologic response rate of 16% in 56 patients with metastatic carcinoid tumors (Table 1 ; Bukowski et al. 1994). Toxicity was a concern in both studies: 88% of patients in the SWOG study reported nausea and/or vomiting and there were two lethal toxicities in the ECOG study. In another study involving 61 patients with carcinoid tumors receiving DTIC as a second-line therapy following treatment with combination chemotherapy, the overall response rate was 8% ( Sun et al. 2005).
Temozolomide is a cytotoxic alkylating agent that was specifically developed as an oral and less toxic alternative to DTIC ( Stevens et al. 1987). In a phase II study, 29 patients with metastatic carcinoid, pancreatic or pheochromocytoma neuroendocrine tumors were treated with temozolomide, administered at a dose of 150 mg/m2 for 7 days, followed by a 7-day rest, together with thalidomide administered at doses of 50–400 mg daily without interruption ( Kulke et al. 2006). The overall objective radiologic response rate among patients receiving temozolomide and thalidomide in this study was 25%, a rate that is comparable with prior studies of both DTIC- and streptozocin-based chemotherapy in patients with neuroendocrine tumors ( Engstrom et al. 1984, Bukowski et al. 1987, 1994). Neuropathy is a known toxicity related to thalidomide and occurred in 38% of the patient population. Grade 3 or 4 lymphopenia developed in 69% of the patients. Three of those patients were on the regimen for more than 6 months and developed opportunistic infections, leading to the recommendation that subsequent patients receive prophylaxis with trimethoprim-sulfamethoxazole.
Other chemotherapeutic agents have, to date, proved relatively inactive in neuroendocrine tumors. High-dose paclitaxel, administered with granulocyte colony-stimulating factor, was evaluated in 24 patients with metastatic carcinoid and islet cell tumors ( Ansell et al. 2001). Significant hematologic toxicity was observed, and the objective radiologic response rate was only 8%. Treatment with docetaxel was associated with biochemical responses but no radiologic responses in a recent phase II trial of 21 patients with carcinoid tumors ( Kulke et al. 2004a). No responses were observed in 18 neuroendocrine tumor patients treated with gemcitabine ( Kulke et al. 2004b).
Novel treatment approaches for metastatic neuroendocrine tumors
The modest efficacy of current systemic treatment regimens has led to interest in the development of novel therapeutic approaches for patients with advanced neuroendocrine tumors. Such approaches include the use of targeted radiotherapy, as well as regimens incorporating inhibitors of growth factor signaling pathways.
Somatostatin receptor targeted radiotherapy
Traditional external beam radiation therapy is beneficial in patients with neuroendocrine tumor metastases to bone, but has little utility for more common visceral metastases. A more broadly applicable strategy includes the therapeutic use of radio-labeled somatostatin analogs ( McCarthy et al. 1998, Meyers et al. 2000, Anthony et al. 2002, Paganelli et al. 2002, Waldherr et al. 2002, Buscombe et al. 2003, Kwekkeboom et al. 2003, 2005). Scintigraphy with Indium-111-labelled octreotide has been commonly used to localize previously undetected primary or metastatic neuroendocrine tumor lesions. At higher doses, Indium-111-labelled octreotide has also been evaluated as a potential novel therapeutic. Unfortunately, objective response rates with this agent have been low ( DeJong et al. 2002). More encouraging results have been obtained with octreotide coupled to Yttrium-90, a high-energy β-particle emitter. In early phase II trials, objective radiologic responses were noted in up to 23% of patients with metastatic neuroendocrine tumors ( Virgolini et al. 2002, Waldherr et al. 2002). The longer-term utility of this agent, however, appears to be limited by both renal and hematologic toxicity ( Valkema et al. 2005). Most recently, octreotide labeled with lutetitum (177Lu), a low-energy β-particle emitter, has been evaluated in a phase I study, with encouraging results. In one series, 131 patients with somatostatin receptor-positive, advanced neuroendocrine tumors received 177Lu-octreotate administered every 6–10 weeks, to a final intended dose of 600–800 mCi ( Kwekkeboom et al. 2005). There were 35 objective responses (27%), three of which were complete.
Inhibition of VEGF and other growth factor signaling pathways
Gastrointestinal neuroendocrine tumors overexpress several growth factors, including VEGF, basic fibroblast growth factor (bFGF), transforming growth factor (TGF)-α and -β, platelet-derived growth factor (PDGF), and insulin-like growth factor (IGF-I). In addition, expression of several growth factor receptors, such as PDGF receptor (PDGFR), IGF-I receptor (IGF-IR), epidermal growth factor receptor (EGFR), VEGF receptor (VEGFR), and stem cell factor receptor (KIT) has been observed ( Chaudhry et al. 1992, 1993, 1994, Christofori et al. 1995, Nilsson et al. 1995, Krishnamurthy & Dayal 1997, Ambs et al. 1998, Terris et al. 1998, La Rosa et al. 2003, Lankat-Buttgereit et al. 2005, Hopfner et al. 2006). Disruption of several of these signaling pathways in different experimental models has resulted in inhibition of neuroendocrine cell growth, leading to a number of recent clinical trials with receptor tyrosine kinase inhibitors and mono-clonal antibodies targeting growth factor signaling in patients with advanced neuroendocrine tumors (Table 3 ).
Imatinib mesylate inhibits the Bcr-Abl, PDGFR-α and -β, and KIT tyrosine kinases. In preclinical studies, incubation of neuroendocrine tumor cells in the presence of imatinib resulted in decreased cell growth ( Lankat-Buttgereit et al. 2005). In a phase II study, however, the administration of imatinib to 27 patients with advanced neuroendocrine carcinoid tumors resulted in only one objective response ( Yao et al. 2007). Similarly, while exposure to gefitinib, a tyrosine kinase inhibitor targeting the EGFR, resulted in growth inhibition of neuroendocrine cell lines, few responses were reported when gefitinib was administered to patients with neuroendocrine tumors in a phase II study ( Hopfner et al. 2003, Hobday et al. 2006).
Neuroendocrine tumors have long been known to be highly vascular, and preliminary results from clinical trials of VEGF pathway inhibitors in patients with advanced neuroendocrine tumors have been more encouraging. These studies suggest that inhibition of either VEGF or VEGFR has the potential for tumor growth inhibition, and in some cases, tumor regression. In one phase II trial, 44 patients with advanced or metastatic carcinoid tumors on a stable dose of octreotide were randomly assigned to receive either bevacizumab (15 mg/kg), a humanized monoclonal antibody targeting VEGF, or pegylated IFN-α-2b (0.5 μg/kg; Yao et al. 2005). Four out of twenty-two patients (18%) treated with bevacizumab achieved confirmed radiographic partial responses, when compared with none of the patients who received pegylated IFN-α-2b. After 18 weeks, 96% of patients treated with bevacizumab remained progression-free, when compared with 68% of patients treated with IFN-α-2b. Hypertension was the most common grade 3/4 adverse event observed with bevacizumab, and granulocytopenia and fatigue were commonly associated with pegylated IFN-α-2b.
Sunitinib, a multitargeted tyrosine kinase inhibitor with activity against not only VEGFR-1, -2, and -3, but also PDGFR-α and -β, KIT, RET, FMS-like tyrosine kinase-3 (FLT3), and colony-stimulating factor receptor (CSF-1R), has also recently been shown to have activity in patients with advanced neuroendocrine tumors. A phase I study of sunitinib included four patients with neuroendocrine tumors; out of these patients, one achieved an objective radiologic response, and a second achieved a minor response with prolonged stable disease. The 28 patients in this study received doses between 15 and 59 mg/m2, ranging from 50 mg every other day to 150 mg/day ( Faivre et al. 2006). These observations led to further evaluation of sunitinib in a phase II study, in which 109 patients with advanced neuroendocrine tumors received repeated 6-week treatment cycles of sunitinib, administered at an oral dose of 50 mg once daily for 4 weeks, followed by 2 weeks off treatment ( Kulke et al. 2005). Hematologic toxicities were relatively uncommon, with grade 3 or 4 neutropenia or thrombocytopenia reported in 21 and 10% of patients respectively. The most common grade 3 or 4 non-hematologic adverse events were fatigue (27%), abdominal pain (12%), and hypertension (11%). A total of 11 out of 66 (17%) patients with pancreatic endocrine tumors and one of 41 (2%) patients with carcinoid tumors achieved confirmed partial responses. The median duration of response was 37 weeks in patients with carcinoid tumors and had not been reached in patients with pancreatic tumors. Stable disease was observed in 83% of the patients with carcinoid tumors and 68% of those with pancreatic endocrine tumors. The median time to tumor progression was 44, 33, and 40 weeks for patients with carcinoid tumors, pancreatic endocrine tumors, and for the entire cohort respectively. Based on these results, further evaluation of sunitinib in patients with advanced neuroendocrine tumors is planned.
Inhibition of mammalian target of rapamycin (mTOR)
mTOR is a threonine kinase that mediates downstream signaling from a number of pathways, including the VEGF and IGF signaling pathways implicated in neuroendocrine tumor growth ( Vignot et al. 2005). Activation of the PI3K/AKT/mTOR pathway has been shown to cause increased translation of proteins regulating cell cycle progression, and inhibitors of mTOR have recently shown promising early activity in a number of cancer types ( Smolewski 2006). In one phase II study, 37 patients with progressive neuroendocrine tumors were treated with the mTOR inhibitor temsirolimus. The objective overall response rate was 5.6%, with 63.9% of patients experiencing either partial response or stable disease; in this study, higher baseline tumor levels of mTOR predicted for better outcomes ( Duran et al. 2006). In a second phase II study, 32 patients with neuroendocrine tumors were treated with a combination of the mTOR inhibitor RAD001 (everolimus; 5 mg/day) and depot octreotide (30 mg every 4 weeks; Yao et al. 2006). In a preliminary report, partial responses (by RECIST) were observed in 2 out of 17 (12%) carcinoid tumor patients and 2 out of 13 (15%) patients with pancreatic neuroendocrine tumors. Further trials to confirm the activity of mTOR inhibitors, either administered alone or in combination with other agents, are planned.
Conclusions
The development of novel, targeted agents is of particular interest in neuroendocrine tumors, in which traditional treatment modalities have had only limited success. Promising recent approaches include somatostatin receptor-targeted radiotherapy, inhibition of mTOR, and inhibition of the VEGF signaling pathway. Even with these novel agents, however, response rates, as measured by traditional response criteria, remain low. It is possible that many of these agents also have a cytostatic effect. However, the indolent nature of neuroendocrine tumors, while beneficial from the standpoint of the patient, makes it difficult or impossible to determine from phase II studies whether stable disease reflects drug effect or simply the natural history of the disease. For similar reasons, the prolonged survival times of patients with advanced neuroendocrine tumors, together with varying selection criteria in trials, make the evaluation of survival data in these studies challenging. Randomized trials or, alternatively, the development of surrogate endpoints of response, including the validation of biochemical, may expedite the more definitive evaluation of these potentially promising agents for neuroendocrine tumors.
Selected chemotherapy trials in metastatic carcinoid tumors
Regimen | Patientsa | Radiologic tumor response rate (%) | Reference |
---|---|---|---|
Phase II trials | |||
DTIC | 56 | 16 | Bukowski et al.(1994) |
DTIC | 61 | 8 | Sun et al.(2005) |
Temozolomide+thalidomide | 14 | 7 | Kulke et al.(2006) |
Etoposide | 17 | 12 | Kelsen et al.(1987) |
Paclitaxelb | 24c | 8 | Ansell et al.(2001) |
Docetaxel | 21 | 0 | Kulke et al. (2004aa) |
Gemcitabine | 18d | 0 | Kulke et al. (2004b) |
Streptozocin +fluorouracil+doxorubicin +cyclophosphamide | 56 | 30 | Bukowski et al.(1987) |
Streptozocin+fluorouracil+cyclophosphamide | 9 | 22 | Bukowski et al.(1987) |
Patientsa | Radiological tumor response rate (%) | Median overall, survival (months) | Reference | |
---|---|---|---|---|
DTIC, dacarbazine. | ||||
aNumber of patients evaluable for efficacy endpoints. | ||||
bPatients also received granulocyte colony-stimulating factor. | ||||
cIncludes patients with carcinoid, pancreatic and anaplastic neuroendocrine tumors. | ||||
dIncludes patients with carcinoid, pancreatic and pheochromoctyoma neuroendocrine tumors. | ||||
Randomized trials | ||||
Streptozocin+cyclophosphamide | 47 | 26 | 12.5 | Moertel & Hanley (1979) |
Streptozocin+fluorouracil | 42 | 33 | 11.2 | |
Doxorubicin | 81 | 21 | 11.1 | Engstrom et al.(1984) |
Streptozocin+fluorouracil | 80 | 22 | 14.9 | |
Doxorubicin+fluorouracil | 88 | 16 | 15.7 | Sun et al.(2005) |
Streptozocin+fluorouracil | 88 | 16 | 24.3 |
Selected trials of systemic chemotherapy for metastatic pancreatic endocrine tumors
Regimen | Patientsa | Tumor response rate (%) | Median overall, survival (months) | Reference |
---|---|---|---|---|
DTIC, dacarbazine; NR, not reported. | ||||
aNumber of patients evaluable for efficacy endpoints. | ||||
bIncludes patients with locally advanced disease. | ||||
Prospective studies | ||||
Chlorozotocin | 33 | 30 | 18.0 | Moertel et al.(1992) |
Fluorouracil+streptozocin | 33 | 45 | 16.8 | |
Doxorubicin+streptozocin | 36 | 69 | 26.4 | |
DTIC | 50 | 34 | 19.3 | Ramanathan et al.(2001) |
Temozolomide+thalidomide | 11 | 45 | NR | Kulke et al.(2006) |
Retrospective studies | ||||
Doxorubicin+streptozocin | 16 | 6 | NR | Cheng & Saltz (1999) |
Doxorubicin+streptozocin | 16 | 6 | 20.2 | McCollum et al.(2004) |
Steptozocin+doxorubicin+fluorouracil | 84b | 39 | 37 | Kouvaraki et al.(2004) |
Trials of novel targeted therapies in neuroendocrine tumors
Agent | Molecular target (s) | Patientsa | Tumor type | Tumor response rate (%) | Median TTP or PFS (weeks) | Reference |
---|---|---|---|---|---|---|
CSF-1R, colony-stimulating factor receptor; EGFR, epidermal growth factor receptor; FLT3, FMS-like tyrosine kinase-3; KIT, stem cell factor receptor; mTOR, mammalian target of rapamycin; NR, not reported; PDGFR, platelet-derived growth factor receptor; PFS, progression-free survival; RET, REarranged during transfection or glial cell-line derived neurotrophic factor; TTP, time to progression; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor. | ||||||
aNumber of patients evaluable for efficacy endpoints. | ||||||
bPatients also received treatment with octreotide. | ||||||
Bevacizumab | VEGF | 18 | Carcinoid | 17b | NR | Yao et al.(2005) |
Sunitinib | VEGFR-1, -2, -3; PDGFR-α, -β; KIT; RET; CSF-1R; FLT3 | 41 | Carcinoid | 2 | 44 (TTP) | Kulke et al.(2005) |
66 | Pancreatic endocrine | 17 | 33 (TTP) | |||
Gefitinib | EGFR | 40 | Carcinoid | 3 | NR | Hobday et al.(2006) |
31 | Pancreatic endocrine | 6 | NR | |||
RAD001 | mTOR | 18 | Carcinoid | 11b | NR | Yao et al.(2006) |
13 | Pancreatic endocrine | 15b | NR | |||
Temsirolimus | mTOR | 21 | Carcinoid | 5 | Duran et al.(2006) | |
15 | Pancreatic endocrine | 7 | ||||
Imatinib | PDGFR-α, -β; KIT; Bcr-Abl | 27 | Carcinoid | 4b | 24 (PFS) | Yao et al.(2007) |

Algorithm for treatment of gastrointestinal neuroendocrine tumors.
Citation: Endocrine-Related Cancer Endocr Relat Cancer 14, 2; 10.1677/ERC-06-0061

Algorithm for treatment of gastrointestinal neuroendocrine tumors.
Citation: Endocrine-Related Cancer Endocr Relat Cancer 14, 2; 10.1677/ERC-06-0061
Algorithm for treatment of gastrointestinal neuroendocrine tumors.
Citation: Endocrine-Related Cancer Endocr Relat Cancer 14, 2; 10.1677/ERC-06-0061
The author would like to thank Taylor Spear for assistance in manuscript preparation and Molly Heitz, PhD, senior medical writer at ACUMED, for her editorial contributions. The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work.
Funding
M H Kulke is supported in part by NIH grants K23 CA 093401, K30 HL04095, and gifts from Raymond and Beverly Sackler, the Caring for Carcinoid Foundation, and the Stephen and Caroline Kaufer Fund for Neuroendocrine Tumor Research.
References
Ajani JA, Carrasco CH, Charnsangavej C, Samaan NA, Levin B & Wallace S 1988 Islet cell tumors metastatic to the liver: effective palliation by sequential hepatic artery embolization. Annals of Internal Medicine 108 340–344.
Alsina AE, Bartus S, Hull D, Rosson R & Schweizer RT 1990 Liver transplant for metastatic neuroendocrine tumor. Journal of Clinical Gastroenterology 12 533–537.
Ambs S, Bennett WP, Merriam WG, Ogunfusika MO, Oser SM, Khan MA, Jones RT & Harris CC 1998 Vascular endothelial growth factor and nitric oxide synthase expression in human lung cancer and the relation to p53. British Journal of Cancer 78 233–239.
Ansell S, Pitot H, Burch P, Kvols L, Mahoney M & Rubin J 2001 A phase II study of high-dose paclitaxel in patients with advanced neuroendocrine tumors. Cancer 91 1543–1548.
Anthony L, Woltering E, Espenan G, Cronin M, Maloney T & McCarthy K 2002 Indium-111-pentetreotide prolongs survival in gastroenteropancreatic malignancies. Seminars in Nuclear Medicine 32 123–132.
Berber E, Flesher N & Siperstein AE 2002 Laparoscopic radiofrequency ablation of neuroendocrine liver metastases. World Journal of Surgery 26 985–990.
Brown KT, Koh BY, Brody LA, Getrajdman GI, Susman J, Fong Y & Blumgart LH 1999 Particle embolization of hepatic neuroendocrine metastases for control of pain and hormonal symptoms. Journal of Vascular and Interventional Radiology 10 397–403.
Bukowski R, Johnson K, Peterson R, Stephens R, Rivkin S, Neilan B & Costanzi J 1987 A phase II trial of combination chemotherapy in patients with metastatic carcinoid tumors. Cancer 60 2891–2895.
Bukowski R, Tangen C, Peterson R, Taylor S, Rinehart J, Eyre H, Rivkin S, Fleming T & Macdonald J 1994 Phase II trial of dimethyltriazenoimidazole carboxamide in patients with metastatic carcinoid. A Southwest Oncology Group study. Cancer 73 1505–1508.
Buscombe J, Caplin M & Hilson A 2003 Long-term efficacy of high-activity 111in-pentetreotide therapy in patients with disseminated neuroendocrine tumors. Journal of Nuclear Medicine 44 1–6.
Chaudhry A, Papanicolaou V, Oberg K, Heldin CH & Funa K 1992 Expression of platelet-derived growth factor and its receptors in neuroendocrine tumors of the digestive system. Cancer Research 52 1006–1012.
Chaudhry A, Funa K & Oberg K 1993 Expression of growth factor peptides and their receptors in neuro-endocrine tumors of the digestive system. Acta Oncologica 32 107–114.
Chaudhry A, Oberg K, Gobl A, Heldin CH & Funa K 1994 Expression of transforming growth factors beta 1, beta 2, beta 3 in neuroendocrine tumors of the digestive system. Anticancer Research 14 2085–2091.
Cheng P & Saltz L 1999 Failure to confirm major objective antitumor activity ofr streptozocin and doxorubicin in the treatment of patients with advanced islet cell carcinoma. Cancer 86 944–948.
Christofori G, Naik P & Hanahan D 1995 Vascular endothelial growth factor and its receptors, flt-1 and flk-1, are expressed in normal pancreatic islets and throughout islet cell tumorigenesis. Molecular Endocrinology 9 1760–1770.
Chung MH, Pisegna J, Spirt M, Giuliano AE, Ye W, Ramming KP & Bilchik AJ 2001 Hepatic cytoreduction followed by a novel long-acting somatostatin analog: a paradigm for intractable neuroendocrine tumors meta-static to the liver. Surgery 130 954–962.
Diamandidou E, Ajani JA, Yang DJ, Chuang VP, Brown CA, Carrasco HC, Lawrence DD & Wallace S 1998 Two-phase study of hepatic artery vascular occlusion with microencapsulated cisplatin in patients with liver metastases from neuroendocrine tumors. AJR. American Journal of Roentgenology 170 339–344.
Dominguez S, Denys A, Madeira I, Hammel P, Vilgrain V, Menu Y, Bernades P & Ruszniewski P 2000 Hepatic arterial chemoembolization with streptozotocin in patients with metastatic digestive endocrine tumours. European Journal of Gastroenterology and Hepatology 12 151–157.
Drougas JG, Anthony LB, Blair TK, Lopez RR, Wright JK Jr, Chapman WC, Webb L, Mazer M, Meranze S & Pinson CW 1998 Hepatic artery chemoembolization for management of patients with advanced metastatic carcinoid tumors. American Journal of Surgery 175 408–412.
Ducreux M, Ruszniewski P, Chayvialle JA, Blumberg J, Cloarec D, Michel H, Raymond JM, Dupas JL, Gouerou H, Jian R et al.2000 The antitumoral effect of the long-acting somatostatin analog lanreotide in neuroendocrine tumors. American Journal of Gastroenterology 95 3276–3281.
Duran I, Kortmansky J, Singh D, Hirte H, Kocha W, Goss G, Le L, Oza A, Nicklee T, Ho J et al.2006 A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer 95 1148–1154.
Engstrom P, Lavin P, Moertel C, Folsch E & Douglass H 1984 Streptozocin plus fluorouracil versus doxorubicin therapy for metastatic carcinoid tumor. Journal of Clinical Oncology 2 1255–1259.
Eriksson BK, Larsson EG, Skogseid BM, Lofberg AM, Lorelius LE & Oberg KE 1998 Liver embolizations of patients with malignant neuroendocrine gastrointestinal tumors. Cancer 83 2293–2301.
Faiss S, Rath U, Mansmann U, Caird D, Clemens N, Riecken EO & Wiedenmann B 1999 Ultra-high-dose lanreotide treatment in patients with metastatic neuroendocrine gastroenteropancreatic tumors. Digestion 60 469–476.
Faiss S, Pape U, Bohmig M, Dorffel Y, Mansmann U, Golder W, Rieck E & Wiedenmann B 2003 Prospective, randomized multicenter trial on the antiproliferative effect of lanreotide, interferon alpha, and their combination for therapy of metastatic neuroendocrine gastroenteropancreatic tumors-the International Lanreotide and Interferon Alpha Study Group. Journal of Clinical Oncology 21 2689–2696.
Faivre S, Delbaldo C, Vera K, Robert C, Lozahic S, Lassau N, Bello C, Deprimo S, Brega N, Massimini G et al.2006 Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. Journal of Clinical Oncology 24 25–35.
Feldman J & O’Dirisio T 1986 Role of neuropeptides and serotonin in the diagnosis of carcinoid tumors. American Journal of Medicine 81 41–48.
Florman S, Toure B, Kim L, Gondolesi G, Roayaie S, Krieger N, Fishbein T, Emre S, Miller C & Schwartz M 2004 Liver transplantation for neuroendocrine tumors. Journal of Gastrointestinal Surgery 8 208–212.
Frank M, Klose KJ, Wied M, Ishaque N, Schade-Brittinger C & Arnold R 1999 Combination therapy with octreotide and alpha-interferon: effect on tumor growth in metastatic endocrine gastroenteropancreatic tumors. American Journal of Gastroenterology 94 1381–1387.
Givi B, Pommier SJ, Thompson AK, Diggs BS & Pommier RF 2006 Operative resection of primary carcinoid neoplasms in patients with liver metastases yields significantly better survival. Surgery 140 891–897 (discussion 897–898).
Gulec SA, Mountcastle TS, Frey D, Cundiff JD, Mathews E, Anthony L, O’Leary JP & Boudreaux JP 2002 Cytoreductive surgery in patients with advanced-stage carcinoid tumors. American Surgeon 68 667–671 (discussion 671–662).
Gupta S, Yao J, Ahrar K, Wallace M, Morello F, Madoff D, Murthy R, Hicks M & Ajani J 2003 Hepatic artery embolization and chemoembolization for treatment of patients with metastatic carcinoid tumors: the MD Anderson experience. Cancer Journal 9 261–267.
Hellman P, Ladjevardi S, Skogseid B, Akerstrom G & Elvin A 2002 Radiofrequency tissue ablation using cooled tip for liver metastases of endocrine tumors. World Journal of Surgery 26 1052–1056.
Hobday T, Holen K, Donehower R, Camoriano J, Kim G, Picus J, Philip P, Lloyd R, Mahoney M & Erlichman C 2006 A phase II trial of gefitinib in patients with progressive metastatic neuroendocrine tumors: a phase II consortium study. Journal of Clinical Oncology, 2006 ASCO Annual Meeting Proceedings 24 A4043.
Hopfner M, Sutter AP, Gerst B, Zeitz M & Scherubl H 2003 A novel approach in the treatment of neuroendocrine gastrointestinal tumours. Targeting the epidermal growth factor receptor by gefitinib (ZD1839). British Journal of Cancer 89 1766–1775.
Hopfner M, Baradari V, Huether A, Schofl C & Scherubl H 2006 The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine gastrointestinal tumours. Endocrine-Related Cancer 13 135–149.
Janson E & Oberg K 1993 Long term management of the carcinoid syndrome: treatment with octreotide alone and in combination with alpha-interferon. Acta Oncologica 32 225–229.
Janson E, Holmberg L, Stridsberg M, Eriksson B, Theodorsson E, Wilander E & Oberg K 1997 Carcinoid tumors: analysis of prognostic factors and survival in 301 patients from a referral center. Annals of Oncology 8 685–690.
DeJong M, Valkema R, Jamar F, Kvols L, Kwekkeboom D, Breeman W, Bakker W, Smith C, Pauwels S & Krenning E 2002 Somatostatin-receptor targeted radionucleotide therapy of tumors: preclinical and clinical findings. Seminars in Nuclear Medicine 32 133–140.
Kaltsas G, Korbonits M, Heintz E, Mukherjee J, Jenkins P, Chew S, Reznek R, Monson J, Besser G, Foley R et al.2001 Comparison of somatostatin analog and meta-iodobenzylguanidine radionuclites in the diagnosis and localization of advanced neuroendocrine tumors. Journal of Clinical Endocrinology and Metabolism 86 895–902.
Kelsen D, Fiore J, Heelan R, Cheng E & Magill G 1987 Phase II trial of etoposide in APUD tumors. Cancer Treatment Reports 71 305–307.
Knox CD, Feurer ID, Wise PE, Lamps LW, Kelly Wright J, Chari RS, Lee Gorden D & Wright Pinson C 2004 Survival and functional quality of life after resection for hepatic carcinoid metastasis. Journal of Gastrointestinal Surgery 8 653–659.
Kolby L, Persson G, Franzen S & Ahren B 2003 Randomized clinical trial of the effect of interferon alpha on survival in patients with disseminated midgut carcinoid tumours. British Journal of Surgery 90 687–693.
Kouvaraki M, Ajani J, Hoff P, Wolff R, Evans D, Lozano R & Yao J 2004 Fluorouracil, doxorubicin, and streptozocin in the treatment of patients with locally advanced and metastatic pancreatic endocrine carcinomas. Journal of Clinical Oncology 22 4762–4771.
Krishnamurthy S & Dayal Y 1997 Immunohistochemical expression of transforming growth factor alpha and epidermal growth factor receptor in gastrointestinal carcinoids. American Journal of Surgical Pathology 21 327–333.
Kulke M, Fuchs C, Stuart K, Ryan D, Enzinger P, Vincitore M, Berg D, Clark J & Mayer R 2004a Phase II study of docetaxel in patients with metastatic carcinoid tumors. Cancer Investigation 22 353–359.
Kulke MH, Kim H, Clark JW, Enzinger PC, Lynch TJ, Morgan JA, Vincitore M, Michelini A & Fuchs CS 2004b A phase II trial of gemcitabine for metastatic neuroendocrine tumors. Cancer 101 934–939.
Kulke M, Lenz H, Meropol N, Posey J, Ryan D, Picus J, Bergsland E, Stuart K, Baum C & Fuchs C 2005 Results of a phase II study with sunitinib malate (SU11248) in patients with advanced neuroendocrine tumours ECCO. European Journal of Cancer Supplement 3 204.
Kulke MH, Stuart K, Enzinger PC, Ryan DP, Clark JW, Muzikansky A, Vincitore M, Michelini A & Fuchs CS 2006 Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors. Journal of Clinical Oncology 24 401–406.
Kvols L, Moertel C, O’Connell M, Schutt A, Rubin J & Hahn R 1986 Treatment of the malignant carcinoid syndrome: evaluation of a long-acting somatostatin analog. New England Journal of Medicine 315 663–666.
Kvols LK, Brown ML, O’Connor MK, Hung JC, Hayostek RJ, Reubi JC & Lamberts SW 1993 Evaluation of a radiolabeled somatostatin analog (I-123 octreotide) in the detection and localization of carcinoid and islet cell tumors. Radiology 187 129–133.
Kwekkeboom D, Bakker W, Kam B, Teunissen J, Kooij P, Herder Wd, Feelders R, Eijck Cv, Jong Md, Srinivasan A et al.2003 Treatment of patients with gastro–entero–pancreatic (GEP) tumours with the novel radiolabelled somatostatin analouge [177Lu-DOTA(0), Try3] octreotate. European Journal of Nuclear Medicine and Molecular Imaging 30 417–422.
Kwekkeboom D, Teunissen J, Bakker W, Kooij P, Herder Wd, Feelders R, vanEijck C, Esser J, Kam B & Krenning E 2005 Radiolabled somatostatin analog [177Lu-DOTA0,Try3] octreotate in patients with endocrine gastroenteropancreatic tumors. Journal of Clinical Oncology 23 2754–2762.
Lamberts S, Bakker W, Reubi J & Krenning E 1990 Somatostatin receptor imaging in the localization of endocrine tumors. New England Journal of Medicine 323 1246–1249.
Lang H, Oldhafer K, Weimann A, Schlitt H, Scheumann G, Flemming P, Ringe B & Pichlmayr R 1997 Liver transplantation for metastatic neuroendocrine tumors. Annals of Surgery 225 347–354.
Lankat-Buttgereit B, Horsch D, Barth P, Arnold R, Blocker S & Goke R 2005 Effects of the tyrosine kinase inhibitor imatinib on neuroendocrine tumor cell growth. Digestion 71 131–140.
Loewe C, Schindl M, Cejna M, Niederle B, Lammer J & Thurnher S 2003 Permanent transarterial embolization of neuroendocrine metastases of the liver using cyanoacrylate and lipiodol: assessment of mid- and long-term results. AJR. American Journal of Roentgenology 180 1379–1384.
McCarthy K, Woltering E, Espenen G, Cronin M, Maloney T & Anthony L 1998 In situ radiotherapy with 111In-Pentreotide: initial observations and future directions. Cancer Journal from Scientific American 4 94–102.
McCollum AD, Kulke MH, Ryan DP, Clark JW, Shulman LN, Mayer RJ, Bartel S & Fuchs CS 2004 Lack of efficacy of streptozocin and doxorubicin in patients with advanced pancreatic endocrine tumors. American Journal of Clinical Oncology 27 485–488.
Meyers M, Anthony L, McCarthy K, Drouant G, Maloney T, Espanan G & Woltering E 2000 High-dose indium 111In pentetreotide radiotherapy for metastatic atypical carcinoid tumor. Southern Medical Journal 93 809–811.
Moertel CG & Hanley JA 1979 Combination chemotherapy trials in metastatic carcinoid tumor and the malignant carcinoid syndrome. Cancer Clinical Trials 2 327–334.
Moertel C, Lefkopoulo M, Lipsitz S, Hahn R & Klaassen D 1992 Streptozocin–doxorubicin, stretpozocin–fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. New England Journal of Medicine 326 519–523.
Moertel C, Johnson C, McKusick M, Martin J, Nagorney D, Kvols L, Rubin J & Kunselman S 1994 The management of patients with advanced carcinoid tumors and islet cell carcinoma. Annals of Internal Medicine 120 302–309.
Musunuru S, Chen H, Rajpal S, Stephani N, McDermott JC, Holen K, Rikkers LF & Weber SM 2006 Metastatic neuroendocrine hepatic tumors: resection improves survival. Archives of Surgery 141 1000–1004 (discussion 1005).
Nilsson O, Wangberg B, Kolby L, Schultz GS & Ahlman H 1995 Expression of transforming growth factor alpha and its receptor in human neuroendocrine tumours. International Journal of Cancer 60 645–651.
Oberg K & Eriksson B 1991 The role of interferons in the management of carcinoid tumors. Acta Oncologica 30 519–522.
Oberg K, Funa K & Alm G 1983 Effects of leukocyte interferon on clinical symptoms and hormone levels in patients with mid-gut carcinoid tumors and carcinoid syndrome. New England Journal of Medicine 309 129–133.
Oberg K, Kvols L, Caplin M, Delle Fave G, de Herder W, Rindi G, Ruszniewski P, Woltering EA & Wiedenmann B 2004 Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system. Annals of Oncology 15 966–973.
Osborne DA, Zervos EE, Strosberg J, Boe BA, Malafa M, Rosemurgy AS, Yeatman TJ, Carey L, Duhaine L & Kvols LK 2006 Improved outcome with cytoreduction versus embolization for symptomatic hepatic metastases of carcinoid and neuroendocrine tumors. Annals of Surgical Oncology 13 572–581.
O’Toole D, Ducreux M, Bommelaer G, Wemeau JL, Bouche O, Catus F, Blumberg J & Ruszniewski P 2000 Treatment of carcinoid syndrome: a prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient acceptability, and tolerance. Cancer 88 770–776.
Paganelli G, Bodei L, Handkiewicz DJ, Junak D, Rocca P, Papi S, Lopera-Sierra M, Gatti M, Chinol M, Bartolomei M et al.2002 90Y-DOTA-D-Phe1-Try3-octreotide in therapy of neuroendocrine malignancies. Biopolymers 66 393–398.
Ramanathan RK, Cnaan A, Hahn RG, Carbone PP & Haller DG 2001 Phase II trial of dacarbazine (DTIC) in advanced pancreatic islet cell carcinoma. Study of the Eastern Cooperative Oncology Group-E6282. Annals of Oncology 12 1139–1143.
La Rosa S, Uccella S, Finzi G, Albarello L, Sessa F & Capella C 2003 Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features. Human Pathology 34 18–27.
Rubin J, Ajani J, Schirmer W, Venook A, Bukowski R, Pommier R, Dandona P & Anthony L 1999 Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. Journal of Clinical Oncology 17 600–606.
Ruszniewski P, Rougier P, Roche A, Legmann P, Sibert A, Hochlaf S, Ychou M & Mignon M 1993 Hepatic arterial chemoembolization in patients with liver metastases of endocrine tumors. Cancer 71 2624–2630.
Saltz L, Trochanowski B, Buckley M, Heffernan B, Niedzwiecki D, Tao Y & Kelsen D 1993 Octreotide as an antineoplastic agent in the treatment of functional and nonfunctional neuroendocrine tumors. Cancer 72 244–248.
Sarmiento JM, Heywood G, Rubin J, Ilstrup DM, Nagorney DM & Que FG 2003 Surgical treatment of neuroendocrine metastases to the liver: a plea for resection to increase survival. Journal of the American College of Surgeons 197 29–37.
Seregni E, Ferrari L, Bajetta E, Martinetti A & Bombardieri E 2001 Clinical significance of blood chromogranin A measurement in neuroendocrine tumours. Annals of Oncology 12 S69–S72.
Smolewski P 2006 Recent developments in targeting the mammalian target of rapamycin (mTOR) kinase pathway. Anticancer Drugs 17 487–494.
Stevens MF, Hickman JA, Langdon SP, Chubb D, Vickers L, Stone R, Baig G, Goddard C, Gibson NW, Slack JA et al.1987 Antitumor activity and pharmacokinetics in mice of 8-carbamoyl-3-methyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG 81045; M & B 39831), a novel drug with potential as an alternative to dacarbazine. Cancer Research 47 5846–5852.
Stivanello M, Berruti A, Torta M, Termine A, Tampellini M, Gorzegno G, Angeli A & Dogliotti L 2001 Circulating chromogranin A in the assessment of patients with neuroendocrine tumours. A single institution experience. Annals of Oncology 12 S73–S77.
Sugimoto E, Lorelius L, Eriksson B & Oberg K 1995 Midgut carcinoid tumors: CT appearance. Radiologica 36 367–371.
Sun W, Lipsitz S, Catalano P, Malliard J, Haler D & Group ECO 2005 Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. Journal of Clinical Oncology 23 4897–4904.
Terris B, Scoazec J & Rubbia L 1998 Expression of vascular endothelial growth factor in digestive neuroendocrine tumors. Histopathology 32 133–138.
Thorson A, Biorck G, Bjorkman G & Waldenstrom J 1954 Malignant carcinoid of the small intestine with metastases to the liver, valvular disease on the right side of the heart (pulmonary stenosis and tricuspid regurgitation without septal defects), peripheral vasomotor symptoms, bronch-oconstriction, and an unusual type of cyanosis. American Heart Journal 47 795–817.
Tomassetti P, Migliori M, Simoni P, Casadei R, De Iasio R, Corinaldesi R & Gullo L 2001 Diagnostic value of plasma chromogranin A in neuroendocrine tumours. European Journal of Gastroenterology and Hepatology 13 55–58.
Touzios JG, Kiely JM, Pitt SC, Rilling WS, Quebbeman EJ, Wilson SD & Pitt HA 2005 Neuroendocrine hepatic metastases: does aggressive management improve survival? Annals of Surgery 241 776–783 (discussion 783–775).
Le Treut YP, Delpero JR, Dousset B, Cherqui D, Segol P, Mantion G, Hannoun L, Benhamou G, Launois B, Boillot O et al.1997 Results of liver transplantation in the treatment of metastatic neuroendocrine tumors. A 31-case French multicentric report. Annals of Surgery 225 355–364.
Valimaki M, Jarvinen H, Salmela P, Sane T, Sjoblom S & Pelkonen R 1991 Is the treatment of metastatic carcinoid tumor with interferon not as successful as suggested? Cancer 67 547–549.
Valkema R, Pauwels SA, Kvols LK, Kwekkeboom DJ, Jamar F, de Jong M, Barone R, Walrand S, Kooij PP, Bakker WH et al.2005 Long-term follow-up of renal function after peptide receptor radiation therapy with (90)Y-DOTA(0),-Tyr(3)-octreotide and (177)Lu-DOTA(0), Tyr(3)-octreotate. Journal of Nuclear Medicine 46 83S–91S.
Vignot S, Faivre S, Aguirre D & Raymond E 2005 mTOR-targeted therapy of cancer with rapamycin derivatives. Annals of Oncology 16 525–537.
van Vilsteren FG, Baskin-Bey ES, Nagorney DM, Sanderson SO, Kremers WK, Rosen CB, Gores GJ & Hobday TJ 2006 Liver transplantation for gastroenteropancreatic neuroendocrine cancers: defining selection criteria to improve survival. Liver Transplantation 12 448–456.
Virgolini I, Traub T, Novotny C, Leimer M, Fuger B, Li SR, Patri P, Pangerl T, Angelberger P, Raderer M et al.2002 Experience with indium-111 and yttrium-90-labeled somatostatin analogs. Current Pharmaceutical Design 8 1781–1807.
Waldherr C, Pless M, Maecke H, Schumacher T, Crazzolara A, Nitzsche E, Haldemann A & Mueller-Brand J 2002 Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq 90Y-DOTATOC. Journal of Nuclear Medicine 43 610–616.
Woodard P, Feldman J, Paine S & Baker M 1995 Midgut carcinoid tumors: CT findings and biochemical profiles. Journal of Computer Assisted Tomography 19 400–405.
Wymenga AN, Eriksson B, Salmela PI, Jacobsen MB, Van Cutsem EJ, Fiasse RH, Valimaki MJ, Renstrup J, de Vries EG & Oberg KE 1999 Efficacy and safety of prolonged-release lanreotide in patients with gastrointestinal neuro-endocrine tumors and hormone-related symptoms. Journal of Clinical Oncology 17 1111.
Yao J, NG C, Hoff P, Phan K, Hess H, Chen X, Wang J, Abbruzzese J & Ajani A 2005 Improved progression-free survival and rapid, sustained decrease in tumor perfusion among patients with advanced carcinoid treated with bevacizumab (abstract). Journal of Clinical Oncology Supplement 23.
Yao J, Phan T, Chang D, Jacobs C, Mares J, Rashid A & Meric-Bernstam F 2006 Phase II study of RAD001 (everolimus) and depot octreotide (Sandostatin LAR) in patients with advanced low grade neuroendocrine carcinoma. Journal of Clinical Oncology, 2006 ASCO Annual Meeting Proceedings 24 A4042.
Yao JC, Zhang JX, Rashid A, Yeung SC, Szklaruk J, Hess K, Xie K, Ellis L, Abbruzzese JL & Ajani JA 2007 Clinical and in vitro studies of imatinib in advanced carcinoid tumors. Clinical Cancer Research 13 234–240.