621
Views
5
CrossRef citations to date
0
Altmetric
Review article

Treatment of endocrine pancreatic tumors

&
Pages 329-338 | Received 10 Jan 2005, Published online: 08 Jul 2009

Abstract

Endocrine pancreatic tumors are rare with an incidence of 4 per million inhabitants. Most tumors are malignant except for insulinomas that usually are benign. They are slowly growing in the majority of cases but there are exceptions with rapidly progressing malignant carcinomas. Because of the rarity of these tumors large randomized trials are difficult to accomplish. However, most physicians treating these patients agree that surgery should be considered in all cases and that medical treatment with chemotherapy and biotherapy is well established for this group of patients.

Treating patients with endocrine pancreatic tumors (EPTs) is challenging in several different aspects. Both tumor proliferation and hormone production, which can be potentially life threatening, have to be considered. EPTs are classified as functioning if they are associated with clinical syndromes due to hormone release and as non-functioning if they are not. The most common syndromes are insulinomas, which are characterized by hypoglycemias, gastrinomas by repeating ulcers, glucagonomas by necrolytic migrating erythemas and VIPomas by excessive diarrhea Citation[1–4]. Tumors producing somatostatin, ACTH (adrenocorticotropic hormone), GH (growth hormone), calcitonin and recently ghrelin have also been described Citation[5–9]. EPTs have a tendency to switch hormone production thereby causing a change from one syndrome to another. Relevant biomarkers and medical history should be screened on a regular basis to avoid unexpected symptom-generating hormone production.

Another challenge regarding treatment of EPTs is that while most endocrine pancreatic tumors are slowly growing there are quite a few patients with rapidly growing tumors requiring a different type of approach Citation[10]. In the last clinicopathological classification regarding neuroendocrine tumors clinical syndromes, tumor morphology and behavior-predicting parameters are considered. EPTs are divided into well-differentiated endocrine tumors, well-differentiated endocrine carcinomas and poorly differentiated carcinomas where also proliferation, measured as Ki-67, is accounted for Citation[11]. See .

Table I.  Clinicopathological classification of endocrine tumors of the pancreas Citation[11].

Chromogranin A is a protein which is co-stored and co-released with peptides and amines from granules in endocrine cells. Almost all patients with EPTs have elevated plasma levels of Chromogranin A making it an important tool both in the diagnostic work-up and evaluation of treatment efficacy Citation[12,13].

There is some controversy regarding treatment of EPTs, partly due to the limited numbers of patients making prospective randomized trials difficult to accomplish Citation[10]. Hence, treatment decisions based on high evidence levels are infrequent; instead decisions are based on small non-randomized studies and vast clinical experience. Therefore, these uncommon patients should be treated in large and well-experienced centers where a multidisciplinary approach can be applied and studies performed.

The data and treatment suggestions presented in this paper are in accordance with the recently published guidelines for management of gastroenteropancreatic neuroendocrine tumors issued by the Nordic Neuroendocrine Tumour Group Citation[14,15]. In this paper we have focused on the clinical treatment of patients with endocrine pancreatic tumors.

Invasive treatment

Surgery

Complete surgical removal of an endocrine pancreatic tumor can be potentially curative but is rarely possible because of metastatic spread Citation[16]. The proportion of patients with resectable liver metastases is only 10% Citation[17]. However, several studies have shown that surgery should be considered despite advanced disease. Partial hepatectomy combined with removal of the primary tumor can result in prolongation of life and possibly cure for patients with metastatic disease Citation[17,18]. Surgical debulking has also proven effective in providing palliation by decreasing hormonal overproduction and local mechanical pressure Citation[19,20]. Despite these promising data, aggressive surgery in metastatic EPTs is controversial and not always performed because of its possible morbidity and mortality. In 1996, Berlin Consensus Conference on neuroendocrine gastroenteropancreatic tumors stated that in spite of the absence of controlled data available to support an aggressive surgery management policy, “primary tumors, liver metastases and extra hepatic metastases should be extirpated if possible” Citation[21]. Since then morbidity and mortality along with survival benefit have been further elucidated in several recently published papers where aggressive surgery of the primary tumor was combined with partial hepatectomy Citation[22–24]. In these studies mortality and morbidity rates were acceptable (0–5% and 18–45%, respectively). Median survival was 6.3–7.5 years which is superior to previously reported 1.7–3.1 years for unresected patients with hepatic metastases Citation[25,26].

Considering these data all patients with endocrine pancreatic tumors should be discussed with a surgeon who has a special interest in these patients.

Radiofrequency ablation

Tumor debulking in the liver can be achieved by local ablation. Today, radiofrequency ablation (RF) has been increasingly used to induce tumor necrosis. Recently, a paper was published on treatment with RF using a cooled-tip needle in 21 patients with mixed endocrine tumors Citation[27]. Forty-three liver metastases were locally ablated using this technique with a success rate, defined as complete radiological necrosis, of 96%. The procedure was safe and efficient. In another study 42 liver metastases were ablated using laparoscopic RF achieving symptom relief in 95% of the patients with significant or complete symptom control in 80% of the patients for a mean of 10 months. Sixty-five percent of the patients showed a partial or significant decrease in their tumor markers during follow-up Citation[28]. There was no mortality and morbidity was 5%. Although RF treatment may seem promising its role in neuroendocrine tumors needs further clarification.

Liver transplantation

During the last few years, liver transplantation has evolved as a potentially curative therapeutic option for patients suffering from neuroendocrine tumors without tumor spread outside the liver. In 1997, Lang et al. published a paper on liver transplantation for metastatic neuroendocrine tumors in 12 patients of which 5 suffered from EPTs Citation[29]. Nine of 12 patients are still alive after a median survival of 55 months and four have no evidence of tumor with a median follow-up of 2–104 months after transplantation. The same group has later performed liver transplantation on 10 patients with EPTs where 9 of 10 patients are alive after a median follow-up of 33 months and 2 are without evidence of disease Citation[30]. The longest disease-free survival is more than 7 years.

In a more recent publication a Spanish group has reported on four patients with neuroendocrine tumors originating from the pancreas and one from the small bowel undergoing liver transplantation Citation[31]. Two patients are alive and free of tumor 3 and 6 years after transplantation whereas 3 died early.

To sum up, it seems like most patients develop tumor recurrences within the first years after liver transplantation while there are some long-term survivors and occasional patients that seem to be cured. The success rate of liver transplantation is probably correlated to the accuracy of the pretransplantation investigation, and every effort should be made to detect extra hepatic disease before liver transplantation is considered.

Liver embolization

The majority of patients with malignant EPTs have most of their tumor burden in the liver. The hepatic metastases derive almost all of their blood supply from the hepatic artery whereas normal liver parenchyma receives about 20–25% of its blood supply from the hepatic artery and the rest from the portal vein Citation[32]. Because of its dual blood supply, the normal liver is protected against infarction caused by occlusion of hepatic arterial branches, while hepatic metastases undergo varying degrees of necrosis. Several reports have demonstrated that selective embolization of a hepatic artery branch can provide reduction of hormonal symptoms and tumor burden in patients with neuroendocrine gastrointestinal tumors Citation[32–34]. Peripheral arterial embolization allows repeated embolizations since the vessels undergo recanalization.

Overall response rates from liver embolization vary between 50–90% and median duration of response is 10–15 months. Symptomatic and biochemical responses are more frequent than radiological responses (40–90% versus 15–40%). Complications such as postembolic syndrome with pain, fever, nausea, leucocytosis and liver enzyme derangements occur in 50–90% of the patients. Severe complications such as renal failure, liver necrosis, intestinal ischemia or cholecystitis, are noted in 10% of the patients and the mortality rate is 3–7% Citation[32–34].

Liver embolization is considered standard treatment in patients with metastatic EPTs offering good palliation. The proceeding is safe provided that it is performed in well experienced centers with multidisciplinary back-up.

Medical treatment

Chemotherapy

Streptozotocin is a nitrosurea compound which inhibits DNA synthesis by alkylation in all stages of the cell cycle Citation[35]. It was chosen for treatment against islet cell carcinomas because of its previous shown activity in mouse leukemia and its cytotoxic effect on beta cells causing diabetes.

In 1973, Broder and Carter treated 52 patients with metastatic islet cell carcinomas with streptozotocin on a weekly schedule and achieved a response rate of 42% where responders showed a significantly higher 1-year survival rate and a doubling of median survival compared to non-responders Citation[36]. See . One decade later, doxorubicin administered every 3–4 weeks induced objective responses in 4 of 20 (20%) patients with heavily pretreated islet cell carcinomas Citation[37]. Another attempt with a monotherapy regimen using monthly cycles of dacarbazine (DTIC) showed radiological responses in 5 of 10 patients with a median duration of response of 48 months Citation[38].

Table II.  Chemotherapy regimens in EPTs.

Polychemotherapy has produced better results. In a randomized study 84 patients with advanced EPTs received either streptozotocin alone or streptozotocin combined with fluorouracil Citation[39]. The combination treatment had significantly higher response rate (63 versus 36%) and rate of complete responses (33 versus 12%) than streptozotocin alone, but duration of response was equal in both arms (17 months). The combination treatment yielded a non-significant survival advantage of 26 versus 16.5 months over single treatment. Because of promising results with adriamycin as single treatment, Moertel et al. randomized 104 patients with advanced EPTs to treatment with streptozotocin combined with fluorouracil or adriamycin, or single treatment with chlorozotocin Citation[40]. Streptozotocin plus adriamycin had a significantly higher response rate (69 versus 45 and 30%) and survival (2.2 versus 1.4 and 1.4 years) than the two other treatment arms. Durations of responses were 14–18 months. However, a previous study comparing streptozotocin combined with fluorouracil or adriamycin in 44 patients showed higher response rate (58% versus 36%) and also significantly longer duration of response (36 versus. 22 months) for the fluorouracil-containing combination Citation[41]. Median survival was 4.9 years which was more than double as high as reported in previous studies. Because of well-known cardiotoxicity for adriamycin and contradictory study results the combination with streptozotocin plus fluorouracil is most commonly preferred today. During the last years a liposomal adriamycin has been registered showing equal efficacy but much less cardiotoxicity than adriamycin. Moertels promising results in the beginning of the 90's with 69% response rate combining streptozotocin with adriamycin has led to a study combining liposomal adriamycin with streptozotocin. Study results with response rates and incidence of cardiotoxicity are currently being processed and will be published shortly.

Streptozotocin is known to be a nauseating drug and despite adequate premedication with 5-hydroxytryptamine (5-HT3)-blockers approximately 10% vomit Citation[42]. Dose-related renal dysfunction including proteinurea and decrease in creatinine clearance is the dose-limiting factor which occurs in 20–70% of the patients. However, blood counts are rarely affected Citation[35].

There is a subset of patients with EPTs that are poorly differentiated and behave aggressively from initial diagnosis and others that start out as indolent tumors suddenly changing their behavior into rapidly progressing tumors. For these aggressive tumors previous treatments are not enough. The most common malignancy derived from neuroendocrine cells, small cell lung cancer, is a rapidly growing malignancy which responds well to treatment with cisplatin combined with etoposide. The same regimen was tried in 45 patients with neuroendocrine gastroenteropancreatic carcinomas Citation[43]. Among patients with anaplastic tumors 67% achieved partial or complete response whereas only 7% of patients with well-differentiated tumors responded partially. Duration of response was 8 months for anaplastic tumors and median survival 19 months which seemed favorable compared to results from previous studies on this aggressive tumor type. Two years ago data was published from a study using a similar regimen when treating heavily pretreated patients with poorly differentiated and/or rapidly progressing neuroendocrine carcinomas Citation[44]. The results confirm previous good results achieving almost 60% response rate among foregut carcinoids and 50% response rate among EPTs. Duration of response was 9 months which was similar to that achieved by Moertel. However, median survival in the study was 38 months which was higher than expected in this category of aggressive and poorly differentiated carcinomas.

Nephrotoxicity is the dose-limiting factor in treatment with cisplatin and etoposide occurring in two-thirds of the patients. Hematological toxicity is universal, even though septic fevers and/or hospitalizations due to this are uncommon Citation[43,44].

Chemotherapy is efficacious in producing responses, both biochemically and radiologically, and in prolonging lifes in patients with EPTs. Streptozotocin combined with fluorouracil is the first choice of chemotherapy. However, if the tumor is poorly differentiated and/or rapidly growing cisplatin combined with etoposide should be chosen instead.

Biotherapy

α-interferon was initially discovered in 1957 Citation[45] and it has later been shown to induce cell blocking in G0 and G1, reduce mRNA for hormones and growth factors and to generally stimulate the immune system Citation[46,47]. In 1983, Öberg et al. published data showing that α-interferon could control hormone secretion, clinical symptoms and tumor growth in carcinoid tumors Citation[48]. Since EPTs are closely related to carcinoids, a study was conducted on 18 patients with advanced EPTs receiving 3–6 MU human leucocyte α-interferon daily Citation[49]. Objective responses (more than 50% reduction in tumor markers or size) were achieved in 77% of the patients with radiological responses in 35% of the patients with a median duration of 8.5 months. A few years later the same authors reported an objective response rate of 51% with a median duration of 20 months when treating 57 patients with α-interferon at a dose of 15–18 MU weekly. In this study only 12% of the patients achieved a radiological response, which is well in line with later published studies.

Previously reported side-effects from treatment with α-interferon are influenza-like symptoms (77%), weight loss (59%), chronic fatigue syndrome (50%), bone-marrow depression (anemia, leucopenia, thrombocytopenia) (38–66%) and transient liver dysfunction (13%) Citation[49]. Autoimmune phenomena, such as systemic lupus erythematosus, vasculitis, polymyositis or thyroiditis, are experienced by about 10–15% of the patients Citation[50]. About 5% of the patients develop neutralizing antibodies with loss of therapeutic effect, which can be restored after switching to natural interferon Citation[51]. Another side effect is mental depression Citation[52], which can be successfully treated with antidepressant therapy.

Somatostatin is a naturally occurring peptide, which was discovered by Brazeau in 1973 Citation[53]. It has been shown to inhibit hormone release, impair gall bladder and gastrointestinal motility and to inhibit tumor growth in vitro Citation[54]. Somatostatin acts through five different somatostatin receptors and induce different second messenger systems depending on which receptor is stimulated. Inhibition of hormone secretion is mediated through somatostatin receptors 2 and 5 Citation[55], growth inhibition through receptors 1,2 and 5 Citation[56,57] and apoptosis through receptors 2 and 3 Citation[58,59].

It is well known that neuroendocrine tumors express somatostatin receptors to a high extent and this is the rationale for treatment with somatostatin analogs in EPTs Citation[60]. Somatostatin analogs are mainly used for symptomatic treatment because of their ability to decrease the level of circulating hormones Citation[61]. The antiproliferative effect seen in vitro is rarely seen in patients Citation[57]. In 1993, Eriksson et al. reported data from treatment of 19 patients with advanced EPTs with octreotide at a dose of 100 µg twice daily Citation[43]. Significant biochemical responses were achieved in 31% of the patients with a median duration of 16 months. No radiological responses were seen. Arnold et al. claims that octreotide can inhibit tumor growth in metastatic gastroenteropancreatic tumors. They treated 52 patients with progressing disease (about one third EPTs) with 200 µg octreotide thrice daily and managed to achieve stable disease in 36.5% of them lasting for a median of 18 months Citation[62].

Treatment with high-dose (up to 12 000 µg/day) lanreotide has shown biochemical response in 58% and tumor response rate in 11% of patients with neuroendocrine gastrointestinal tumors failing on treatment with standard doses of octreotide Citation[63]. Induction of apoptosis could be demonstrated in the tumors.

Adverse effects from treatment with somatostatin analogs are mild compared to those seen with chemotherapy or treatment with interferon. Most side-effects such as nausea, transient abdominal pains, flatulence, diarrhea and local reaction at the injection site dissolve with time Citation[42,62]. In 20–50% of the patients, gall-stones are formed, but these virtually always remain asymptomatic Citation[64].

Combined treatment with α-interferon and somatostatin analogs has been shown to produce higher biochemical response rates than either drug alone in midgut carcinoids Citation[65]. Recently, data was published from a study examining the effects of combination treatment in 16 patients with metastatic EPTs Citation[66]. Biochemical response was achieved in more than 60% of the patients and objective radiological response in almost 20% of them, median duration of response 23 and 22 months, respectively. The results are encouraging with higher response rates for combination treatment both biochemically and radiologically, but the study did not manage to prove that combination treatment is better than either drug alone in EPTs. The combination treatment was not accompanied with additional adverse effects, apart from those previously described for each drug.

Recently, Faiss et al. published data from a prospective and randomized study comparing the antiproliferative effect of lanreotide, interferon-alfa and their combination in 80 patients with metastatic neuroendocrine gastroenteropancreatic tumors Citation[67]. All treatment arms achieved similar rates of biochemical and objective responses. Symptomatic responses were achieved in all treatment arms but significant differences could only be shown in the combination arm compared to base-line. The antiproliferative effect was lower than that previously reported, only 5%.

Biotherapy is a well-established treatment in metastatic EPTs which mainly induces biochemical responses but also tumor reduction in some patients. Combination treatment with α-interferon and somatostatin analogs can induce responses that are comparable to those achieved by chemotherapy and it can be used as a good alternative treatment in patients that no longer responds to chemotherapy or in those who cannot or do not want to receive chemotherapy.

Treatment with radiolabelled somatostatin analogs

Since neuroendocrine tumors contain high numbers of somatostatin receptors scintigraphy using the radiolabelled somatostatin analog octreotide (Octreoscan) has enabled in vivo visualization of the primary tumor and metastases. The first attempts to treat patients using high dose radiolabelled somatostatin analogs were reported about 10 years ago. In a phase I study, Krenning et al. treated 6 end-stage patients suffering from neuroendocrine tumors with [111In-DTPA-D-Phe1]-octreotide (OCT) up to a cumulative dose of 53 GBq per patient and achieved impressive effects on hormone production and a likely anti-proliferative effect without any major side effects Citation[68]. These promising results have been repeated by a Swedish group treating five patients with neuroendocrine tumors with OCT at high doses (6 MBq every third week) and an American group treating 27 patients with neuroendocrine tumors with at least 2 monthly injections of 180-mCi OCT Citation[69,70]. In the latter study biochemical response was achieved in 81% of the patients and objective tumor response in 8% of the patients. Patients included in the study had a life expectancy of 6 months and survival achieved in the study was threefold longer, 18 months. The treatments were well tolerated with transient side-effects including nephrotoxicity and bone-marrow depression.

In recent years researchers have developed new somatostatin analogs with higher affinity for somatostatin receptors and they have also tried linking different radioisotopes to the analogs. If isotopes emitting β-particles, for example yttrium, are used neighboring cells might also be killed since β-particles can extend over several cell diameters. In a prospective phase II study, thirteen patients with endocrine pancreatic tumors were treated with four equal intravenous injections of a total of 7.4 GBq yttrium labeled octreotide (90Y-DOTATOC) and achieved an objective response rate of 38% with one complete response Citation[71]. Renal insufficiency was the dose limiting toxicity and one patient experienced non-transient renal insufficiency grade 2 despite infusion with amino acid for renal protection. The most common side-effect was nausea which could be successfully treated with domperidone or odansetron.

In a more recent study octreotate, which is a somatostatin analog with nine fold higher affinity for somatostatin receptor subtype 2 than octreotide, was used in conjunction with lutetium (177Lu) which is a beta- and gamma-emitting radioisotope. Previous comparisons between patients receiving 111In-octreotide and patients receiving 177Lu-octreotate had shown comparable uptake in kidneys, liver and spleen but three to fourfold higher uptake in the tumors favoring 177Lu-octreotate Citation[72]. Thirty-five patients with gastroenteropancreatic tumors were treated with a final cumulative dose of 600–800 mCi 177Lu-octreotate and of these one patient (3%) achieved a complete response and 12 (35%) achieved partial response Citation[73]. Nausea was common, but no renal toxicity was reported. In this study even patients with grade 2–3 tumor uptake on Octreoscan were included and tumor response was significantly higher in patients with grade 4 uptake compared to grade 2–3 uptake.

Despite these promising results the place for radioactive targeting therapy is still not clear. The selection of patients and the timing for the procedure have to be further established.

Symptom relieving treatments

All previously described treatments aim to reduce tumor burden and hence decrease hormone production, which will result in symptomatic improvement for the patients. However, sometimes these treatments are not enough to control the patients’ symptoms. In these cases there are symptom relieving treatments that do not effect tumor burden or hormone production, but only alleviate the symptoms. Patients with insulinomas suffering from severe hypoglycemias might benefit from treatment with corticosteroids, which causes glucose intolerance with insulin resistance Citation[74] or diazoxid, which reduces insulin secretion from beta cells resulting in increased blood glucose Citation[75]. If the symptoms still are uncontrolled the patients need continuous intravenous infusion with glucose. Patients with glucagonomas may have the opposite problem suffering from hyperglycemias requiring injections of insulin lowering the blood glucose values. Glucagon production may result in skin lesions called necrolytic migratory erythema which often are successfully treated by injections of somatostatin analogs Citation[76] but in severe cases treatment with amino acid and fatty acid infusions may be successful Citation[77]. The introduction of proton pump inhibitors has been a break through for patients with gastrinomas suffering from repeating ulcers Citation[78]. Severe diarrheas that often torment patients with VIPomas can be reduced by loperamid, opium drops and bulk-forming laxatives Citation[79,80]. Tumors producing ACTH may be very aggressive with rapidly progressing Cushing's syndrome requiring substitution with potassium or medication with ketoconazol or metyrapone which both interfere in the synthesis of corticosteroids Citation[81,82]. In severe cases with life-threatening Cushing's syndrome adrenalectomy has to be considered Citation[83].

Future aspects

New high-affinity and subtype selective somatostatin analogs for each of the five somatostatin receptors have been identified, but are not yet commercially available Citation[84]. A somatostatin analog which binds selectively to somatostatin receptor 3 might induce apoptosis and thereby tumor regression to a greater degree than those available today. It has been suggested that somatostatin analogs act not only directly on tumor cells, but also on peritumoral vessels Citation[85]. In a recent paper 23 tumor specimens from EPTs were examined regarding the expression of somatostatin receptors in intratumoral vessels and the authors found that somatostatin receptors 2 and 4 were highly expressed whereas the other three receptors were more rare Citation[86]. There have been reports claiming that somatostatin potently inhibits angiogenesis Citation[87]. Combination treatment with other known angiogenesis inhibitors might achieve stronger angiogenetic inhibition than single drug treatment.

Molecular targeting with tyrosin kinase inhibitors and monoclonal antibodies is a new and exciting approach to cancer treatment. Today there are tyrosine kinase inhibitors on the market inhibiting signaling through c-kit, EGFR and PDGFRs which have proven effective in the treatment of gastrointestinal stromal tumors, chronic myeloic leukemia and lung cancer Citation[88–93]. In a recently published study, 38 tumor specimens from EPTs were examined immunohistochemically showing that most tumor cells expressed c-kit and PDGFRb whereas about half expressed EGFR Citation[94]. Furthermore, recently ZD 1839, which is an EGFR tyrosine kinase inhibitor, was found to induce growth inhibition, apoptosis and cell cycle arrest in human neuroendocrine gastrointestinal tumor cells Citation[95]. These results are very interesting and it appears as though EGFR tyrosine kinase inhibition may be a promising novel approach for the treatment of neuroendocrine tumors. Clinical studies have been initiated and we await their results.

Conclusion

Despite controversy regarding treatment of EPTs most physicians agree that surgery should be considered for all patients-as first-line treatment offering a chance for cure in a small number of patients Citation[16] or as palliative treatment in patients with advanced stage disease where it may offer symptom relief and a delay for medical treatment Citation[19,96]. Ablation of liver metastases using RF has shown promising results, but needs further clarification before used as standard treatment Citation[27,28]. Liver transplantation is exciting, but additional studies are required to state which patients are suitable for this approach and the shortage of donor organs also needs to be considered Citation[29–31]. Combination treatment with streptozotocin and 5-fluorouracil/doxorubicin have shown high response rates lasting for up to 3 years making it a standard medical treatment in many centers around the world Citation[10]. An alternative to chemotherapy is treatment with somatostatin analogs and/or α-interferon which have shown good results with biochemical responses and symptom relief in most patients, however only few objective tumor responses Citation[42,66,67]. Liver embolization can produce good biochemical symptomatic responses in patients with most tumor burden in the liver Citation[32–34]. Promising results from treatment with radio labeled somatostatin analogs have been published during the last years showing positive effects both on symptoms, hormone levels and tumor size Citation[68–73].

Consequently, there are several treatment options to consider when treating patients suffering from EPTs. The most difficult task is to decide which therapeutic intervention should be initiated and when. There is no obvious order in which different treatments should be begun. For example, in one patient surgery might be the first choice of therapeutic intervention whereas in another patient medical treatment with chemotherapy might be necessary to make the tumor operable. In a third patient, biotherapy and symptom releasing therapy might be first choice of treatments because of uncontrolled hormone production and/or poor general health etc. A multidisciplinary approach is obliged to provide for different needs in different patients. We strongly recommend that these patients be taken care of in centers where this is possible.

The present work with new somatostatin analogs and radioisotopes in the treatment of EPTs with radiolabelled somatostatin analogs is very exciting and we look forward to more studies establishing its role in the standard treatment arsenal. Molecular targeting is becoming increasingly important in oncology and we await clinical studies elucidating the role of tyrosine kinase inhibitors in EPTs.

References

  • Wilder RM, Allan FN, Power MH, Robertson HE. Carcinoma of the islands of the pancreas. Hyperinsulinism and hypoglycemia. J Am Med Assoc 1927; 89: 348–55
  • Zollinger RM, Ellison EH. Primary peptic ulcerations of the jejunum associated with islet cell tumors of the pancreas. Ann Surg 1955; 142: 709–28
  • Mallinson CN, Bloom SR, Warin AP, Salmon PR, Cox B. A glucagonoma syndrome. Lancet 1974; 2: 1–5
  • Verner JV, Morrison AB. Islet cell tumor and a syndrome of refractory watery diarrhea and hypokalemia. Am J Med 1958; 25: 374–80
  • Ganda OP, Weir GC, Soeldner JS, et al. Somatostatinoma”: a somatostatin containing tumor of the endocrine pancreas. N Engl J Med 1977; 296: 963–7
  • Maton PN, Gardner JD, Jensen RT. Cushing's syndrome in patients with Zollinger-Ellison syndrome. N Engl J Med 1986; 315: 1–5
  • Imura H. Ectopic hormone syndromes. Clin Endocrinol Metab 1980; 9: 235–60
  • Jensen RT. Pancreatic endocrine tumors: recent advances. Ann Oncol 1999; 10: 170–6
  • Corbetta S, Peracchi M, Cappiello V, et al. Circulating ghrelin levels in patients with pancreatic and gastrointestinal neuroendocrine tumors: Identification of one pancreatic ghrelinoma. J Clin Endocrinol Metab 2003; 88: 3117–20
  • Eriksson B, Arnberg H, Lindgren PG, et al. Neuroendocrine tumors: clinical presentation, biochemical and histopathological findings in 84 patients. J Intern Med 1990; 228: 103–13
  • Rindi G, Capella C, Solcia. Cell biology, clinicopathological profile, and classification of gastro-enteropancreatic endocrine tumors. J Mol Med 1998; 76: 413–20
  • Eriksson B, Arnberg H, Oberg K, et al. Chromogranins–new sensitive markers for neuroendocrine tumors. Acta Oncol 1989; 28: 325–9
  • Schurmann G, Raeth U, Wiedenmann B, Buhr H, Herfarth C. Serum chromogranin A in the diagnosis and follow-up of neuroendocrine tumors of the gastroenteropancreatic tract. World J Surg 1992; 16: 697–701
  • Oberg K, Astrup L, Eriksson B, et al. Guidelines for the management of gastroenteropancreatic neuroendocrine tumours (including bronchopulmonary and thymic neoplasms). Part I-general overview. Acta Oncol 2004; 43: 617–25
  • Oberg K, Astrup L, Eriksson B, et al. Guidelines for the management of gastroenteropancreatic neuroendocrine tumours (including bronchopulmonary and thymic neoplasms). Part II-specific NE tumour types. Acta Oncol 2004; 43: 626–36
  • Welbourn, RB,Wood, SM,Polak, JM,Bloom, SR, . In:. Pancreatic endocrine tumors.SR Bloom,Polak, JR, , editors., Gut Hormones, 2nd edition., London, Churchill Livingstone; 1981. p., 547–1554
  • Carty SE, Jensen RT, Norton JA. Prospective study of aggressive resection of metastatic pancreatic endocrine tumors. Surgery 1992; 112: 1024–31
  • Que FG, Nagorney DM, Batts KP, Linz LJ, Kvols LK. Hepatic resection for metastatic neuroendocrine carcinomas. Am J Surg 1995; 169: 36–42
  • Montenegro F, Lawrence GD, Macon W, Pass C. Metastatic glucagonoma: improvement after surgical debulking. Am J Surg 1980; 139: 424–7
  • Nagorney DM, Blood SR, Polak JM, Blumgart LH. Resolution of recurrent Verner-Morrison syndrome by resection of metastatic vipoma. Surgery 1983; 93: 348–53
  • Wiedenmann B, Jensen RT, Mignon M, et al. Preoperative diagnosis and surgical management of neuroendocrine gastroenteropancreatic tumors: general recommendations by a consensus workshop. World J Surg 1998; 22: 309–18
  • Sarmiento JM, Que FG, Grant CS, Thompson GB, Farnell MB, Nagorney DM. Concurrent resections of pancreatic islet cell cancers with synchronous hepatic metastases: outcomes of an aggressive approach. Surgery 2002; 132: 976–82
  • Elias D, Lasser P, Ducreux M, et al. Liver resection (and associated extra-hepatic resections) for metastatic well-differentiated endocrine tumors: A 15-year single center prospective study. Surgery 2003; 133: 375–82
  • Norton JA, Kivlen M, Li M, Schneider D, Chuter T, Jensen RT. Morbidity and mortality of aggressive resection in patients with advanced neuroendocrine tumors. Arch Surg 2003; 138: 859–66
  • Thompson GB, van Heerden JA, Grant CS, Carney JA, Ilstrup DM. Islet carcinomas of the pancreas; a twenty-year experience. Surgery 1988; 104: 1011–7
  • Solorzano CC, Lee JE, Pisters PW, et al. Nonfunctioning islet cell carcinoma of the pancreas: survival results in a contemporary series of 163 patients. Surgery 2001; 130: 1078–85
  • Hellman P, Ladjevardi S, Skogseid B, Akerstrom G, Elvin A. Radiofrequency tissue ablation using cooled tip for liver metastases of endocrine tumors. World J Surg 2002; 26: 1052–6
  • Berber E, Flesher N, Siperstein AE. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases. World J Surg 2002; 26: 985–90
  • Lang H, Oldhafer KJ, Weimann A, et al. Liver transplantation for metastatic neuroendocrine tumors. Ann Surg 1997; 225: 347–54
  • Lang H, Schlitt HJ, Schmidt H, et al. Total hepatectomy and liver transplantation for metastatic neuroendocrine tumors of the pancreas-a single center experience with ten patients. Langenbecks Arch Surg 1999; 384: 370–7
  • Fernandez JA, Robles R, Marin C, et al. Role of liver transplantation in the management of metastatic neuroendocrine tumors. Transplant Proc 2003; 5: 1832–3
  • Ackerman NB, Sien WM, Silverman NA. The blood supply of experimental liver metastases. 3. The effects of acute ligation of hepatic artery or portal vein. Surgery 1972; 71: 636–41
  • Ajani JA, Carrasco CH, Charnsangavej C, Samaan NA, Levin B, Wallace S. Islet cell tumor metastatic to the liver: effective palliation by sequential hepatic artery embolization. Ann Intern Med 1988; 108: 340–4
  • Carrasco CH, Chuang V, Wallace S. Apudomas metastatic to the liver: treatment by hepatic artery embolization. Radiology 1983; 149: 79–83
  • Weiss RB. Streptozotocin: A review of its pharmacology, efficacy and toxicity. Cancer Treat Rep 1982; 66: 427–38
  • Broder LE, Carter SK. Pancreatic islet cell carcinoma. II. Results of therapy with streptozotocin in 52 patients. Ann Intern Med 1973; 79: 108–18
  • Moertel CG, Lavin P, Hahn G. Phase II trial of doxorubicin therapy for advanced islet cell carcinoma. Cancer Treat Rep 1982; 66: 1567–9
  • Altimari A, Badrinath K, Reisel HJ, Printz RA. DTIC therapy in patients with malignant intra-abdominal neuroendocrine tumors. Surgery 1987; 102: 1009–17
  • Moertel CG, Hanley JA, Johnson LA. Streptozotocin alone compared to streptozotocin plus fluorouracil in the treatment of advanced islet-cell carcinoma. N Engl J Med 1980; 303: 1189–94
  • Moertel CG, Lefkopoulo M, Lipsitz M, Hahn RG, Klaassen D. Streptozotocin-doxorubicin, streptozotocin-fluorouracil, or chlorozotocin in the treatment of advanced islet-cell carcinoma. N Engl J Med 1992; 326: 519–23
  • Eriksson B, Skogseid B, Lundqvist G, Wide L, Wilander E, Oberg K. Medical treatment and long-term survival in a prospective study of 84 patients with endocrine pancreatic tumors. Cancer 1990; 65: 1883–90
  • Eriksson B, Öberg K. An update of the medical treatment of malignant endocrine pancreatic tumors. Acta Oncol 1993; 32: 203–8
  • Moertel CG, Kvols LK, O′Connell MJ, Rubin J. Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in anaplastic variants of these neoplasma. Cancer 1991; 68: 227–32
  • Fjällskog M-L, Granberg D, Welin S, et al. Treatment with cisplatin and etoposid in patients with neuroendocrine tumors. Cancer 2001; 92: 1101–7
  • Isaac A, Lindenmann J. Virus interference. I. The interferon. By A Isaacs and J Lindenmann, 1957. J Interferon Res 1987; 7: 429–38
  • Andersson T, Wilander E, Eriksson B, Lindgren PG, Oberg K. Effects of interferon on tumor tissue content in liver metastases of carcinoid tumors. Cancer Res 1990; 50: 3413–15
  • Grandér D, Öberg K, Lundqvist M-L, Janson ET, Eriksson B, Einhorn S. Interferon-induced enhancement of 2′-5′-oligoadenylat synthetase in mid-gut carcinoid tumors. Lancet 1990; 336: 337–40
  • 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: 129–33
  • Eriksson B, Oberg K, Alm G, et al. Treatment of malignant endocrine pancreatic tumors with human leucocyte interferon. Lancet 1986; 2: 1307–9
  • Ronnblom LE, Alm GV, Oberg K. Autoimmun phenomena in patients with malignant carcinoid tumors during interferon-alpha treatment. Acta Oncol 1991; 30: 537–40
  • Öberg K, Alm G, Magnusson A, et al. Treatment of malignant carcinoid tumors with recombinant interferon-alfa-2b: development of neutralizing antibodies and possible loss of antitumor activity. J Natl Cancer Inst 1989; 81: 531–5
  • Merimsky O, Chaitchik S. Neurotoxicity of interferon-alpha. Anticancer drugs 1992; 3: 567–70
  • Brazeau P, Vale W, Burgus R, et al. Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science 1973; 179: 77–9
  • Reichlin, S. Somatostatin. Part 1 and 2.N Engl J Med. 1983;309:1495–1501, and 1556–63.
  • Chen C, Vincent JD, Clarke IJ. Ion channels and the signal transduction pathways in the regulation of growth hormone secretion. Trends Endocrinol Metab 1994; 5: 227–33
  • Buscail L, Delesque N, Èsteve JP, et al. Stimulation of tyrosine phosphatase and inhibition of proliferation by somatostatin analogues: mediation by human somatostatin receptors ssTR1 and ssTR2. Proc Natl Acad Sci USA 1994; 91: 2315–9
  • Buscail L, Estéve JP, Saint-Laurent N, et al. Inhibition of cell proliferation by the somatostatin analogue RC-160 is mediated by somatostatin receptors subtypes ssTR2 and ssTR5 through different mechanisms. Proc Natl Acad Sci USA 1995; 92: 1580–4
  • Sharma K, Patel YC, Srikant CB. Subtype-elective induction of wild-type p53 and apoptosis, but not cell-cycle arrest, by human somatostatin receptor 3. Mol Endocrinol 1996; 10: 1688–96
  • Teijeiro R, Rios R, Costoya JA. Activation of somatostatin receptor 2 promotes apoptosis through a mechanism that is independent from induction of p53. Cell Physiol Biochem 2002; 12: 31–8
  • Reubi JC, Hacki WH, Lamberts SW. Hormone producing gastrointestinal tumors contain a high density of somatostatin receptors. J Clin Endocrinol Metab 1987; 65: 1127–34
  • Long RG, Barnes AJ, Adrian TE, et al. Suppression of pancreatic endocrine tumor secretion by long-acting somatostatin analogue. Lancet 1979; 2: 764–7
  • Arnold R, Trautmann ME, creutzfeldt W, et al. Somatostatin analogue octreotide and inhibition of tumor growth in metastatic gastroenteropancreatic tumors. Gut 1996; 38: 430–8
  • Eriksson B, Renstrup J, Imam H, Oberg K. High-dose treatment with lanreotide of patients with advanced neuroendocrine gastrointestinal tumors: clinical and biological effects. Ann Oncol 1997; 8: 1041–4
  • Trendle MC, Moertel CG, Kvols LK. Incidence and morbidity of cholelithiasis in patients receiving chronic octreotide for metastatic carcinoid tumors and malignant islet cell tumors. Cancer 1997; 79: 830–4
  • Tiensuu Janson EM, Ahlström H, Andersson T, Oberg K. Octreotide and interferon-alfa: a new combination for the treatment of malignant carcinoid tumors. Eur J Cancer 1992; 28: 1647–50
  • Fjällskog M-L, Sundin A, Westlin J-E, Oberg K, Janson ET, Eriksson B. Treatment of malignant endocrine pancreatic tumors with a combination of α-interferon and somatostatin analogs. Med Oncol 2002; 19: 35–42
  • Faiss S, Pape UF, Bohmig M, et al. International Lanreotide and Interferon Alfa Study Group. Prospective, randomized, multicenter trial on the antiproliferative effect of lanreotide, interferon alfa, and their combination for therapy for metastatic neuroendocrine gastroenteropancreatic tumors. The international lanreotide and interferon alfa study. J Clin Oncol 2003; 21: 2689–96
  • Krenning E, Kooij P, Pauwels S, et al. Somatostatin receptor: scintigraphy and radionucleide therapy. Digestion 1996; 57(Suppl 1)57–61
  • Tiensuu Janson E, Eriksson B, Oberg K, et al. Treatment with high dose [111In-DTPA-D-Phe1]-octreotide in patients with neuroendocrine tumors- evaluation of therapeutic and toxic effects. Acta Oncol 1999; 38: 373–7
  • Anthony L, Woltering W, Espenan GD, Cronin MD, Maloney TJ, McCarthy KE. Indium-111-penterotide prolongs survival in gastroenteropancreatic malignancies. Semin Nucl Med 2002; 32: 123–32
  • Waldherr C, Pless M, Maecke HR, et al. Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq 90Y-DOTATOC. J Nucl Med 2002; 43: 610–6
  • Kwekkeboom D, Bakker W, Kooij PP, et al. 177Lu-DOTA0Tyr3]octreotate: comparison with [111In-DTPA0]octreotide in patients. Eur J Nucl Med 2001; 28: 1319–25
  • Kwekkeboom D, Bakker B, Kam BL, et al. Treatment of patients with gastroenteropancreatic (GEP) tumors with a novel radiolabelled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate. Eur J Nucl Med 2003; 30: 417–22
  • Cavallo–Perin P, Bruno A, Ozzello A, et al. Comparison of acute and subacute effects of deflazacort and prednisone on glucose metabolism in man. Eur J Pharmacol 1984; 26: 357–62
  • Grant DB, Dunger DB, Burns EC. Long–term treatment with diazoxid in childhood hyperinsulinism. Acta Endocrinol Suppl 1986; 279: 340–5
  • Boden G, Ryan IG, Eisenschmid BL, Shelmet JJ, Owen OE. Treatment of inoperable glucagonomas with the long-acting somatostatin analogue SMS 201-995. N Engl J Med 1986; 314: 1686–9
  • Alexander EK, Robinson M, Staniec M, Dluhy RG. Peripheral amino acid and fatty acid infusion for the treatment of necrolytic migratory erythemas in the glucagonomas syndrome. Clin Endocrinol 2002; 57: 827–31
  • Faiss S, Scherubl H, Riecken EO, Wiedenmann B. Drug therapy in metastatic neuroendocrine tumors of the gastroenteropancreatic system. Recent results cancer res 1996; 142: 193–207
  • Yamashiro Y, Yamamoto K, Sato M. Loperamide therapy in a child with vipoma-associated diarrhea. Lancet 1982; 1: 1413
  • Ewe K. Opium drops in diarrhea caused by Crohn disease. Dtsch Med Wochenschr 1997; 122: 174–5
  • Engelhardt D, Jacob K, Doerr HG. Different therapeutic efficacy of ketoconazole in patients with Cushing's syndrome. Klin Wochenschr 1989; 67: 241–7
  • Verhelst JA, Trainer PJ, Howlett TA, et al. Short and long-term responses to metyrapone in the medical management of 91 patients with Cushing's syndrome. Clin endocrinol 1991; 35: 169–78
  • Iser G, Pfohl M, Dorr U, Weiss EM, Seif FJ. Ectopic ACTH secretion due to a bronchopulmonary carcinoid localized by somatostatin receptor scintigraphy. Clin Invest 1994; 72: 887–91
  • Rohrer SP, Birzin ET, Mosley RT, et al. Rapid identification of subtype-selective agonists of the somatostatin receptor through combinatorial chemistry. Science 1998; 282: 737–40
  • Reubi JC, Horisberger U, Laissue J. High density of somatostatin receptors in veins surrounding human cancer tissue: role in tumor-host interaction?. Int J Cancer 1994; 56: 681–8
  • Fjallskog ML, Ludvigsen E, Stridsberg M, Oberg K, Eriksson B, Janson ET. Expression of somatostatin receptor subtypes 1-5 in tumor tissue and intratumoral vessels in malignant endocrine pancreatic tumors. Med Oncol 2003; 20: 59–67
  • Albini A, Florio T, Giunciuglio D, et al. Somatostatin controls Kaposi's sarcoma tumor growth through inhibition of angiogenesis. FASEB J 1999; 13: 647–55
  • Druker BJ, Tamura S, Buchdunger E, et al. Effects of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med 1996; 2: 561–6
  • Buchdunger E, Cioffi C, Law N, et al. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J Pharmacol Exp Ther 2000; 295: 139–45
  • Ferry D, Hammond L, Ransom M, et al. Intermittent oral Zd1839 (Iressa), a novel epidermal growth factor receptor tyrosine kinase inhibitor (Egfr-Tki), shows good tolerability and activity: final results from a phase I study. Proc Am Soc Clin Oncol 2000; 19: abs 5E
  • Joensuu H, Roberts PJ, Sarlomo-Rikala M, et al. Effect of tyrosine kinase inhibitor STI571 in a patient with a metastatic gastrointestinal stromal tumor. N Engl J Med 2001; 344: 1052–6
  • Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloic leukaemia. N Engl J Med 2001; 344: 1031–7
  • Baselga J, Herbst R, LoRusso P, et al. Continuous administration of ZD1839 (Iressa), a novel oral epidermal growth factor receptor tyrosine kinase receptor inhibitor, in patients with five selected tumor types: evidence of activity and good tolerability. Proc Am Soc Clin Oncol 2000; 19: abs 686
  • Fjällskog ML, Lejonklou MH, Oberg KE, Eriksson BK, Janson ET. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res 2003; 9: 1469–73
  • Hopfner M, Sutter AP, Gerst B, Zeitz M, Scherubl H. A novel approach in the treatment of neuroendocrine gastrointestinal tumors. Targeting the epidermal growth factor receptor gefitinib (ZD1839). Br J Cancer 2003; 89: 1766–75
  • McEntee GP, Nagorney DM, Kvols LK, Moertel CG, Grant CS. Cytoreductive hepatic surgery for neuroendocrine tumors. Surgery 1990; 108: 1091–6

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.