throbber
Annals of Oncology 15 (Supplement 4): iv293 – iv298, 2004
`
`doi:10.1093/annonc/mdh942
`
`Management of neuroendocrine tumours
`
`K. O¨ berg
`
`Department of Endocrine Oncology, University Hospital, Uppsala, Sweden
`
`Introduction
`
`Neuroendocrine (NE) tumours of the gastrointestinal tract and
`pancreas constitute about 2% of all malignant tumours. They
`include a number of different tumours, derived from cells of
`the diffuse NE cell system [1]. The largest group of NE
`tumours are the so-called carcinoids, with an incidence of
`about 2.5/100 000 [2], which by tradition have been divided
`into foregut, midgut and hindgut tumours. This old classi-
`fication is based on the embryonic origin of the different
`tumours, where the foregut primaries have been located in
`the lung, thymus, gastric mucosa; the midgut with primary
`tumours in the ileum, caecum and proximal colon; and the
`hindgut with the primaries in the distal colon and rectum. This
`old classification is now about to be abandoned, and a more
`tumour-based classification has emerged. The new World
`Health Organization (WHO) classification now indicates five
`subtypes [3]:
`
`1 well-differentiated endocrine tumour
`2 well-differentiated endocrine carcinoma
`3 poorly differentiated endocrine carcinoma
`4 mixed exocrine and endocrine carcinomas
`5 tumour-like lesions.
`
`This classification can be used for all types of NE tumour; not
`only for carcinoids.
`A classical midgut carcinoid will, in the new terminology,
`be classified as a well-differentiated endocrine carcinoma of
`the ileum, whereas a benign insulin-producing tumour of the
`pancreas will be a well-differentiated endocrine tumour of the
`pancreas. The differentiation between different tumour types
`is based on histomorphology, tumour size and the presence or
`absence of local invasion and/or metastases. This new classifi-
`cation of NE tumours is a step forward, although the former
`classification of carcinoid tumours into foregut, midgut and
`hindgut remains clinically available and is still used in many
`clinical studies. It will take some time for the new classifi-
`cation to be generally accepted.
`NE tumours exhibit substantial differences in terms of geno-
`type and phenotype. Foregut carcinoids mainly located in the
`lung but also endocrine pancreatic tumours, frequently show
`loss of 11q, which represent a characteristic genetic alteration
`in these tumours. Both typical and atypical carcinoids of the
`lung show loss of heterozygosity at 11q13, harbouring the
`multiple endocrine neoplasia type 1 (MEN-1) gene. Atypical
`carcinoids also show loss of heterozygosity at 3p14 – p21.3.
`
`q 2004 European Society for Medical Oncology
`
`Recent studies have shown that carcinoid tumours of the lung
`and the gastrointestinal
`tract may develop via different
`molecular pathways. Inactivation of one of several tumour
`suppressor genes on chromosome 18 may be important for the
`biological behaviour of gastrointestinal
`tumours. Familial
`midgut carcinoids are rare but bronchial carcinoids as well as
`endocrine pancreatic tumours and gastric carcinoids may be
`part of a MEN-1 syndrome [4, 5].
`
`Clinical presentation
`
`The different NE tumours may be divided into functioning
`and non-functioning tumours. Functioning tumours present
`clinically with symptoms related to overproduction of hor-
`mones and amines such as midgut carcinoids with carcinoid
`syndrome, gastrinoma with Zollinger – Ellison’s syndrome,
`insulinoma with hypoglycaemic symptoms, glucagonoma with
`glucagonoma syndrome and VIPoma with watery diarrhoea –
`hypokalaemia – achlorhydria (WDHA) syndrome.
`Non-functioning tumours produce and secrete peptides that
`do not cause any distinct clinical symptom. The majority of
`endocrine pancreatic tumours is non-functioning tumours
`(40 – 45%).
`The classical carcinoid syndrome includes flushing (80%),
`diarrhoea (70%), abdominal pain (40%), valvular heart disease
`(30 – 40%), telangiectasia (25%), wheezing (15%) and pellagra-
`like skin lesions (5%).
`The flushing observed in patients with classical carcinoid
`syndrome has usually a pink to red colour and involves the
`face or upper trunk. It lasts for a few minutes and may be
`triggered by alcohol, physical exercise, mental stress and
`tyramine-containing foods such as chocolates, walnuts and
`bananas.
`The atypical carcinoid syndrome that is seen in bronchial
`carcinoids has a purple rather than pink colour with telang-
`iectasias, hypertrophy of the skin of the face, headache,
`lacrimation, hypotension, cutaneous edema and bronchocon-
`striction.
`Carcinoid heart disease is characterized by plaque-like
`fibrous endocardial
`thickening that classically involves the
`right side of the heart, occurring in 50 – 70% of patients with
`a carcinoid syndrome. Haemodynamically significant heart
`disease is seen in about 5 – 10% of patients [6, 9, 10].
`Zollinger – Ellison’s
`syndrome is
`related to a gastrin-
`producing tumour located either in the pancreas (45 – 50%) or
`in the duodenum (35 – 55%). These tumours produce gastrin,
`ARGENTUM
`Exhibit 1026
`
`000001
`
`

`

`iv294
`
`which stimulates high acid output with recurrent gastritis
`and gastric ulcers. Nowadays with the use of proton-pump
`inhibitors severe ulcer disease is very rare.
`Insulinoma or hypoglycaemic syndrome is characterized by
`neuroglycopenia, particularly in the morning or after exercise,
`blurred vision and sometimes even psychosis. Secondary to
`low blood glucose the patient might develop palpitations and
`sweating.
`WDHA syndrome or VIPoma syndrome is characterized by
`severe secretory diarrhoea up to 10 – 15 l/day, hypercalcaemia,
`hypokalaemia, achlorhydria, flushing and diabetic glucose
`tolerance.
`Glucagonoma syndrome is very often present with typical
`necrolytic migratory erythema but also anaemia and thrombo-
`sis. About 50% of the patients also show mild diabetes melli-
`tus [6 – 8].
`Non-functioning tumours of the intestine present with large
`abdominal masses, bleeding from the gastrointestinal
`tract
`and/or intestinal obstruction. Non-functioning endocrine pan-
`creatic tumours become very large until they cause abdominal
`discomfort, pain and liver enlargement. Sometimes they also
`cause jaundice [8].
`
`Biochemical diagnosis of NE tumours
`
`Since the majority of NE tumours secretes peptides and
`amines,
`these can be used as markers both for diagnosis
`and monitoring of therapy.
`The most
`important
`tumour marker is chromogranin A,
`which is a glycoprotein stored in the secretory granules of
`tumour cells and is released together with other peptides and
`amines. The level of chromogranin A is increased in 70 – 90%
`of all NE tumours. The sensitivity and specificity are approxi-
`mately 92% and 96%, respectively.
`Another marker
`is pancreatic polypeptide, which is
`increased in 50 – 60% of patients with endocrine pancreatic
`tumours and a somewhat lower number in carcinoid of the
`gastrointestinal tract. The specificity and sensitivity is lower
`for this marker.
`Another tumour marker is human chorionic gonadotropin
`(HCG) a-subunit, which is particularly useful to determine the
`malignant potential of a NE tumour.
`A specific marker for patients with a carcinoid syndrome
`the 24 h urinary level of 5-hydroxyindoleacetic acid
`is
`(5-HIAA), a metabolite of serotonin with a sensitivity of 73%
`and specificity of 100%. The urine should be collected under
`dietary restrictions, excluding bananas, chocolate, tea, coffee,
`walnuts and pecan.
`Patients with foregut carcinoid tumours rarely secrete sero-
`tonin but may release adrenocorticotropic hormone (ACTH),
`growth hormone releasing hormone (GHRH) or histamine
`[11 – 14].
`Serum gastrin together with measurement of the basal and
`stimulated acid output are the best diagnostic tools for gastri-
`noma. Sometimes a secretin infusion test measuring gastrin is
`necessary to demonstrate a gastrinoma.
`
`For the diagnosis of an insulin-producing tumour, measure-
`ment of fasting serum insulin and blood glucose might be
`sufficient, but sensitivity can be improved by analysing pro-
`insulin and C-peptide as well. Sometimes 24 – 72 h fasting is
`necessary to diagnose an insulinoma.
`VIPoma is demonstrated by measuring plasma vasoactive
`intestinal peptide (VIP), which is significantly elevated in
`most patients.
`Plasma glucagon is elevated in most patients with a
`glucagonoma.
`Non-functioning tumours may present with a high chromo-
`granin A level as well as pancreatic polypeptide and HCG-a
`subunit [7, 8].
`
`Imaging of NE tumours
`
`A unique feature of NE tumour cells is the expression of
`peptide hormone receptors on their surface. These receptors
`may be targets for both diagnosis and therapeutic procedures.
`The majority of NE tumour cells expresses somatostatin
`receptors particularly receptor subtypes 2 and 5. They are the
`basis for somatostatin receptor scintigraphy (Octreoscan). This
`procedure is actually the most important staging procedure for
`NE tumours since 55 – 95% of these tumours express somato-
`statin receptors. Somatostatin receptor scintigraphy has a diag-
`nostic accuracy of 83% and a positive predictive value of
`100%. It has a higher sensitivity than I-131-metaiodobenzyl-
`guanidine (MIBG) scanning
`Undifferentiated anaplastic NE tumours with a high pro-
`liferation capacity lack somatostatin receptors and may give
`a negative somatostatin receptor scintigraphy. These tumours
`can be diagnosed by positron emission tomography (PET)
`scanning with [18F]-2-deoxy-2-fluoro-D-glucose (18-FDG).
`Another PET tracer with high sensitivity for well-differen-
`tiated hormone-producing NE tumours is C11-5HTP, with
`very high sensitivity and specificity. Tumours with a size of
`2 mm can be visualized by this method.
`Besides imaging by radionucleotides, computed tomography
`(CT) scan or magnetic resonance imaging (MRI) as well as
`ultrasonography should always be performed to visualize the
`precise location and size of the lesion for evaluation during
`treatment. Most recently a combined PET – CT camera has
`been developed, which makes a computerized fusion image of
`both methods.
`For endocrine tumours of the pancreatic head, endoscopic
`ultrasonography is particularly useful. Other
`localization
`procedures are bronchoscopy, gastroscopy and colonoscopy
`[7, 8, 15 – 18].
`
`Treatment of NE tumours
`
`Surgery
`
`The clinical management of metastatic NE tumours requires a
`multi-modal approach including surgery and other means of
`cytoreductive treatment, radiotherapy and medical treatment.
`
`000002
`
`

`

`Surgery remains the treatment of choice and is the only
`approach that can achieve a complete cure in patients with NE
`tumours. In cases of metastases, surgery has been used to
`improve hormone-mediated symptoms, quality of life and sur-
`vival in certain groups of patients, as well as to reduce tumour
`bulk and prevent further local and systemic effects. Surgical
`resection of primary tumours as well as lymph nodes and liver
`metastases can improve survival. In addition, surgery can also
`be employed after medical treatment to achieve substantial
`tumour reduction in an attempt to maximize the disease-free
`interval. Surgery and thermal ablation (radiofrequency treat-
`ment) are new promising methods for treatment of liver
`metastases. Significant clinical improvement and reduction in
`tumour size have been reported.
`Liver transplantation has been suggested in selected patients
`without residual extrahepatic manifestations. However, long-
`term results are not that encouraging at the moment and liver
`transplantation should be reserved for a very few patients,
`where other means of therapy cannot control
`the disease
`[7, 19, 20].
`
`Embolization and chemoembolization
`
`A significant number of patients have liver metastases at diag-
`nosis. Therefore treatment aimed at reducing the tumour bulk
`in the liver may significantly improve quality of life and survi-
`val. Such procedures include embolization of liver metastasis
`with or without concomitant cytotoxic agents (chemoemboli-
`zation). Chemoembolization is embolization combined with
`intra-arterial administration of chemotherapy.
`By chemoembolization, the concentration of chemothera-
`peutic drugs may reach a higher local concentration and their
`action may be more effective due to the cellular tumour
`
`Table 1. Cytotoxic therapy for carcinoid tumours
`
`Drug
`
`Regimen
`
`iv295
`
`ischaemia. Contraindications for liver embolization include
`complete portal vein obstruction and hepatic insufficiency.
`If liver metastases are the only site of metastasis, emboli-
`zation may be the first-line treatment
`in patients with NE
`tumours poorly responsive to biological treatment or systemic
`cytotoxic treatment.
`in
`Chemoembolization gives symptomatic improvement
`75 – 100%, biochemical responses in 57 – 90% and significant
`tumour reduction in 35 – 80% of patients with NE tumours.
`The response duration is between 8 and 40 months.
`Side effects are usually mild and transient nausea, vomiting,
`abdominal pain, mild fever and raised liver enzymes. Patients
`with extensive disease may suffer necrosis of the tumour with
`carcinoid crises. Other rare but important complications are
`acute renal
`failure (hepato-renal syndrome), peptic ulcer
`bleeding and necrosis of the gall bladder [21, 22].
`
`Radiotherapy
`
`External radiotherapy has limited value in the treatment of NE
`tumours. It is reserved mainly for treatment of brain meta-
`stases and pain related to bone metastases.
`Tumour-targeted radiolabelled somatostatin analogues have
`been used during the past few years with some encouraging
`results. The various compounds used are 111indium-DTPA-
`octreotide, 90yttrium-DOTA-octreotide, 90yttrium-DOTATOC
`and MAURITIUS, all giving a symptomatic improvement in
`40% of the patients, biochemical responses in 24 – 30% and
`significant tumour reduction in 5 – 10%.
`To overcome the limitation of administering radiotherapy to
`non-octreotide avid lesions and lack of uptake due to tumour
`heterogeneity, several other isotopes such as 177lutetium and
`186rhenium are being examined.
`177Lu-DOTA-octreotate
`
`Number of
`patients
`
`Overall
`response (%)
`
`Median duration
`(months)
`
`Single agents
`
`Doxorubicin
`
`5-Fluorouracil
`
`Streptozotocin
`
`Dacarbazine
`
`Cisplatin
`
`Combinations
`
`Streptozotocin
`
`+5-Fluorouracil
`
`Streptozotocin
`
`+Doxorubicin
`
`Streptozotocin
`
`+Cyclophosphamide
`
`Etoposide
`
`+Cisplatin
`
`60 mg/m2 every 3 – 4 weeks
`500 mg/m2/day 5 every 5 weeks
`500 – 1500 mg/m2/day 5 every 3 – 5 weeks
`250 mg/m2/day 5 every 4 – 5 weeks
`45 – 90 mg/m2 every 3 – 4 weeks
`
`81
`
`30
`
`14
`
`15
`
`16
`
`21
`
`17 – 26
`
`0 – 17
`
`13
`
`6
`
`6
`
`3
`
`2
`
`4.5
`
`4.5
`
`500 mg/m2/day 5 every 3 – 6 weeks
`400 mg/m2/day 5 every 3 – 6 weeks
`1000 mg/m2/week 4
`25 mg/m2/week then every 2 weeks
`
`500 mg/m2/day every 6 weeks
`
`100 mg/m2 once every 3 weeks
`130 mg/m2/day 3
`45 mg/m2/day on day 2 and 3,
`repeat cycle every 4 weeks
`
`175
`
`7 – 33
`
`3 – 7
`
`10
`
`24
`
`13
`
`40
`
`39
`
`0
`
`5
`
`6.5
`
`–
`
`000003
`
`

`

`iv296
`
`shows a high tumour uptake with a very good ratio of tumour
`to kidney uptake. This isotope has recently been administered
`to 80 patients with a variety of progressive NE tumours and
`49% showed a partial response [23 – 25].
`
`Medical treatment
`
`treatment of NE tumours includes treatment with
`Medical
`both chemotherapy and biological agents, such as somatostatin
`analogues and interferon (IFN)-a.
`
`Chemotherapy
`
`Chemotherapy has been considered the gold standard for treat-
`ment of most NE tumours. However,
`its use is usually
`reported in a limited number of patients and with variable
`criteria for assessment of antitumour response.
`Cytotoxic treatment is predominantly used in patients with
`tumours with a high proliferative capacity shown by a pro-
`liferation index of >10 – 15% measured by the antibody Ki67
`and with a large tumour burden. Patients with a classical mid-
`gut carcinoid with a low proliferating capacity (Ki67 usually
`<2%) do not benefit from cytotoxic treatment.
`The most common chemotherapy for the treatment of endo-
`crine pancreatic tumours is a combination of streptozotocin
`plus 5-fluorouracil or doxorubicin. Response rates are between
`40% and 70%. In classical midgut carcinoids the same combi-
`nation induces responses of short duration in <10%.
`
`Table 2. Chemotherapy of endocrine pancreatic tumours
`
`No. of
`patients
`
`Objective
`response (%)
`
`Duration
`(months)
`
`Streptozotocin + 5-fluorouracil
`
`170
`
`Streptozotocin + doxorubicin
`
`Cisplatinum + etoposide
`
`Dacarbazine
`
`Paclitaxel
`
`50
`
`14
`
`11
`
`15
`
`45 – 63
`
`40 – 69
`
`50
`
`9
`
`7
`
`18 – 36
`
`12 – 24
`
`9
`
`6
`
`5
`
`For anaplastic tumours with a high proliferative capacity
`(Ki67 > 15%) combinations of cisplatin and etoposide have
`been useful with a response rate of 67% and a tendency to
`prolonged survival [26, 27]. Newer cytotoxic agents such as
`paclitaxel and gemcitabine have not been of substantial value.
`Results of phase II studies with different chemotherapeutic
`regimens are shown in Tables 1 and 2.
`
`Somatostatin analogues
`
`The rationale for the clinical use of somatostatin analogues
`is based on the identification of high-affinity somatostatin
`receptors in 80 – 90% of NE tumours. Regular octreotide at a
`subcutaneous daily dose of 200 – 450 mg is associated with a
`median 60% symptomatic, 70% biochemical and 8% tumour
`response. A limited number of patients has been reported with
`partial tumour regression during treatment with somatostatin
`analogues and very few cases have shown complete tumour
`regression. However, a high number of patients reached
`disease stabilization.
`(Sandostatin
`Slow release
`formulations of octreotide
`LARw) and somatuline (Somatuline Autogelw) have been
`effective with a monthly dose of 20 – 30 mg octreotide or 60 –
`120 mg somatuline. In clinical practice the patient is treated
`subcutaneously with immediate release octreotide 100 mg 2 – 3
`times/day for at least 3 – 4 days to see whether the patient can
`tolerate somatostatin analogue treatment. Thereafter, a long-
`acting formulation of octreotide 20 mg or somatuline 90 mg is
`given intramuscularly every 4 weeks. The immediate release
`formulation of octreotide is continued during the first 2 weeks
`to prevent symptoms until the long-acting formulation of the
`drug reaches a steady state concentration. If symptoms return
`before the next administration, the interval can be shortened
`to 2 or 3 weeks. The patient should also have a supply of
`immediate release octreotide for particular situations.
`When a patient develops resistance to a somatostatin ana-
`logue, which may occur after 9 – 12 months of treatment, dose
`escalation may be tried with doses up to 60 mg for octreotide
`or 150 mg of somatuline.
`
`Table 3. NE tumours: somatostatin analogue therapy (summary of several trials)
`
`Response
`
`Symptomatic
`
`Biochemical
`
`Standard dose
`(100 – 1500 mg/day) (%)
`
`High dose
`(>3000 mg/day) (%)
`
`Slow release
`(20 – 30 mg/day every 2 – 4 weeks) (%)
`
`64 (146/228)
`
`42 (11/26)
`
`63 (76/119)
`
`Complete response
`
`11 (6/54)
`
`Partial response
`
`55 (116/211)
`
`Stable disease
`
`34 (72/211)
`
`Progressive disease
`
`11 (23/211)
`
`Tumour
`
`Complete response
`
`–
`
`Partial response
`
`5 (7/131)
`
`Stable disease
`
`38 (50/131)
`
`Progressive disease
`
`56 (74/131)
`
`3 (1/33)
`
`72 (24/83)
`
`21 (7/33)
`
`3 (1/33)
`
`2 (1/53)
`
`11 (6/53)
`
`47 (25/53)
`
`39 (21/51)
`
`3 (3/119)
`
`64 (76/119)
`
`18 (21/119)
`
`15 (19/119)
`
`–
`
`3 (4/119)
`
`79 (94/119)
`
`18 (21/119)
`
`000004
`
`

`

`to somatostatin analogue
`If a patient becomes resistant
`treatment, IFN-a might be an alternative to up-regulate the
`number of somatostatin receptors type 2 or to give a period of
`rest to somatostatin receptors. Somatostatin analogue therapy
`can be re-instituted after 2 – 3 months, by using either the
`immediate release or the long-acting formulation.
`SOM230 is a new somatostatin analogue that has a
`prolonged half-life, ( 24 h) and exerts a more potent inhibi-
`tory effect than the compounds currently available, as it binds
`with much higher affinity to somatostatin receptors 1, 2, 3 and
`5. The introduction of SOM230 into clinical practice will
`address a long-standing question as to whether somatostatin
`receptor subtypes 1 and 3, which mediate antitumour effects
`(cell cycle inhibition and induction of apoptosis) will be clini-
`cally beneficial in NE tumours [28 – 31]. Results of trials with
`somatostatin analogues are given in Table 3.
`
`Interferons
`
`Interferons are compounds known to exert a combination of
`effects directed to several groups of tumours and are con-
`sidered as biological response modifiers as they interact with
`other soluble or cell-associated regulatory factors. The rec-
`ommended dose of IFN-a is 3 – 9 MU subcutaneously every
`other day, or slow release formulation pegylated IFN-a 80 –
`100 mg subcutaneously once a week. The dose should be
`titrated individually and the leucocyte count may be used
`as guidance: a leucocyte count of <3.0 109/l indicates an
`optimal IFN-a dose.
`Several studies in patients with carcinoid tumours have
`reported a median symptomatic and biochemical response rate
`of 40 – 70%, biochemical responses in 40 – 60% and significant
`tumour reduction in 10 – 12% (Table 4). Disease stabilization
`is noted in a further 35% of the patients. Flu-like symptoms
`are almost universal with interferon treatment but are usually
`short
`lasting. Chronic fatigue and mild depression may
`develop in  50% of patients. Autoimmune reactions appear
`in  15% of patients [27, 32, 33].
`
`Table 4. Therapy with IFN-a in patients with midgut carcinoids
`
`Number of
`patients
`
`Biochemical
`response (%)
`
`Subjective
`response (%)
`
`Tumour value
`response (%)
`
`PR 53 (13/25)
`
`72 (32/29)
`
`PR 10 (3/29)
`
`SD 36 (9/25)
`
`PR 39 (9/23)
`
`65
`
`PR 16 (1/6)
`
`SD 86 (25/29)
`
`PR 20 (4/20)
`
`PR 0 (0/16)
`
`SD 50 (3/6)
`
`80 (4/5)
`
`SD 66 (10/15)
`
`29a
`
`27b
`
`16
`
`14
`
`13
`
`PR 44 (4/9)
`
`55
`
`PR 8 (1/13)
`
`PR 0 (0/16)
`
`PR 8 (1/13)
`
`SD 77 (10/13)
`
`SD 31 (4/13)
`50
`aNatural leucocyte IFN-a, 6 MU subcutaneously 8 weeks.
`bHigh-dose IFN-a2a 24 MU/m2 subcutaneously 8 weeks.
`PR, partial response; SD, stable disease.
`
`iv297
`
`Figure 1. Algorithm of NE tumours.
`
`Combination of IFN and somatostatin analogue
`
`The value of combining IFN-a with somatostatin analogues
`has been discussed during the past few years. Early non-
`randomized studies indicated a beneficial effect of the combi-
`nation, with significant antitumour and biochemical responses
`in patients
`resistant
`to either
`IFN or
`a
`somatostatin
`analogue alone [34, 35].
`Recent randomized trials have not supported this early
`observation, but these studies have several flaws. One of these
`studies included a low number of patients and the statistical
`analyses were not performed correctly [36]. The combination
`of IFN-a and a somatostatin analogue showed a non-signi-
`ficant trend to improved survival. The other study included
`different types of NE tumours with different tumour biology
`and it was not easy to evaluate the combination therapy [37].
`The tolerance of IFN-a is improved by use of a concomi-
`tant somatostatin analogue and experimental data indicate an
`up-regulation of the somatostatin receptor type 2 by IFN-a.
`IFN-a with a somatostatin
`Therefore the combination of
`analogue should be evaluated in randomized studies
`in
`pre-defined patient populations. Current concepts in therapy
`are summarized in Figure 1.
`
`New compounds
`
`transduction by tyrosine
`intracellular signal
`Inhibition of
`kinase receptors may be a new target in the treatment of NE
`tumours. Many NE tumours express platelet-derived growth
`factor-a and -b receptor subtypes and ligands, and also
`vascular endothelial growth factor and epidermal growth
`factor receptors.
`
`000005
`
`

`

`iv298
`
`Another interesting new compound is rapamycin, which
`may block signal transduction through the mTOR pathway.
`Clinical trials with this compound as a single agent or in
`combination with cytotoxic agents are planned.
`Over the next 5 years the precise role of tumour-targeted
`radioactive treatment with somatostatin analogue-based com-
`pounds will be defined. New somatostatin analogues, such as
`SOM230 and somatostatin receptor subtype-specific analogues
`will also be developed. The tumour biology for different sub-
`types of NE tumour will be defined and thus new treatments
`including tyrosine kinase inhibitors, anti-angiogenic com-
`pounds as well as combinations of these, will be applied in
`clinical trials.
`
`References
`
`1. Kimura W, Kuroda A, Morioka Y. Clinical pathology of endocrine
`tumors of the pancreas. Dig Dis Sci 1991; 36: 933– 942.
`2. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carci-
`noid tumors. Cancer 2003; 97: 934 – 959.
`3. Solcia E, Kloppel G, Sobin L (eds). Histological Typing of Endocrine
`Tumours. New York, USA: Springer 2000; 38 – 74.
`4. Rindi G, Villanacci V, Ubiali A. Biological and molecular aspects
`of gastroenteropancreatic neuroendocrine tumors. Digestion 2000; 62
`(Suppl 1): 19 – 26.
`5. Oberg K. Carcinoid tumors: molecular genetics, tumor biology, and
`update of diagnosis and treatment. Curr Opin Oncol 2002; 14:
`38 – 45.
`6. Oberg K. Neuroendocrine gastrointestinal tumors—a condensed over-
`view of diagnosis and treatment. Ann Oncol 1999; 10 (Suppl 2):
`S3– S8.
`7. Thompson GB, van Heerden JA, Grant CS et al. Islet cell carcinomas
`of
`the pancreas: a twenty-year experience. Surgery 1988; 104:
`1011– 1017.
`8. Bieligk S, Jaffe BM. Islet cell tumors of the pancreas. Surg Clin
`North Am 1995; 75: 1025 – 1040.
`9. Caplin ME, Buscombe JR, Hilson AJ et al. Carcinoid tumour. Lancet
`1998; 352: 799– 805.
`10. Kulke MH, Mayer RJ. Carcinoid tumors. N Engl J Med 1999; 340:
`858 – 868.
`11. Granberg D, Wilander E, Stridsberg M et al. Clinical symptoms,
`hormone profiles, treatment, and prognosis in patients with gastric
`carcinoids. Gut 1998; 43: 223– 228.
`12. Feldman JM, O’Dorisio TM. Role of neuropeptides and serotonin in
`the diagnosis of carcinoid tumors. Am J Med 1986; 81: 41 – 48.
`13. Lamberts SW, Hofland LJ, Nobels FR. Neuroendocrine tumor mar-
`kers. Front Neuroendocrinol 2001; 22: 309– 339.
`14. Stridsberg M, Eriksson B, Oberg K, Janson ET. A comparison
`between three commercial kits for chromogranin A measurements.
`J Endocrinol 2003; 177: 337 – 341.
`15. Ricke J, Klose KJ, Mignon M et al. Standardisation of imaging in
`neuroendocrine tumours: results of a European delphi process. Eur
`J Radiol 2001; 37: 8 – 17.
`16. Kwekkeboom D, Krenning EP, de Jong M. Peptide receptor imaging
`and therapy. J Nucl Med 2000; 41: 1704– 1713.
`17. Krenning EP, de Jong M, Kooij PP et al. Radiolabelled somatostatin
`analogue(s)
`for peptide receptor
`scintigraphy and radionuclide
`therapy. Ann Oncol 1999; 10 (Suppl 2): S23– S29.
`
`18. Orlefors H, Sundin A, Ahlstrom H et al. Positron emission tomo-
`graphy with 5-hydroxytryprophan in neuroendocrine tumors. J Clin
`Oncol 1998; 16: 2534– 2541.
`19. Hellman P, Lundstrom T, Ohrvall U et al. Effect of surgery on the
`outcome of midgut carcinoid disease with lymph node and liver
`metastases. World J Surg 2002; 26: 991– 997.
`20. Le Treut YP, Delpero JR, Dousset B et al. Results of liver trans-
`plantation in the treatment of metastatic neuroendocrine tumors.
`A 31-case French multicentric report. Ann Surg 1997; 225: 355– 364.
`21. Ruszniewski P, Malka D. Hepatic arterial chemoembolization in the
`management of advanced digestive endocrine tumors. Digestion 2000;
`62 (Suppl 1): 79 – 83.
`22. Ajani JA, Carrasco CH, Wallace S. Neuroendocrine tumors metastatic
`to the liver. Vascular occlusion therapy. Ann NY Acad Sci 1994; 733:
`479– 487.
`23. Anthony LB, Woltering EA, Espenan GD et al. Indium-111-pentetreo-
`tide prolongs survival in gastroenteropancreatic malignancies. Semin
`Nucl Med 2002; 32: 123– 132.
`24. De Jong MBW, Bernard HF et al. Response to radionuclide therapy
`using 90Y and 111In-labeled somatostatin analogs is dependent on
`tumor size. J Nucl Med 2000; 41: 147.
`25. Kwekkeboom DJ, Bakker WH, Kooij PP et al. [177Lu-DOTAOTyr3]-
`octreotate: comparison with [111In-DTPAo]octreotide in patients. Eur
`J Nucl Med 2001; 28: 1319 – 1325.
`26. Oberg K. Advances in chemotherapy and biotherapy of endocrine
`tumors. Curr Opin Oncol 1998; 10: 58 – 65.
`27. Oberg K. Chemotherapy and biotherapy in the treatment of neuro-
`endocrine tumours. Ann Oncol 2001; 12 (Suppl 2): S111 – S114.
`28. Eriksson B, Oberg K. Summing up 15 years of somatostatin analog
`therapy in neuroendocrine tumors: future outlook. Ann Oncol 1999;
`10 (Suppl 2): S31– S38.
`29. Oberg K. Established clinical use of octreotide and lanreotide in
`oncology. Chemotherapy 2001; 47 (Suppl 2): 40 – 53.
`30. Oberg K, Kvols L, Caplin M et al. Consensus report on the use
`of somatostatin analogs for
`the management of neuroendocrine
`tumors of the gastroenteropancreatic system. Ann Oncol 2004; 15:
`966– 973.
`31. Bruns C, Lewis I, Briner U et al. SOM230: a novel somatostatin pep-
`tidomimetic with broad somatotropin release inhibiting factor (SRIF)
`receptor binding and a unique antisecretory profile. Eur J Endocrinol
`2002; 146: 707– 716.
`32. Oberg K, Eriksson B. The role of interferons in the management of
`carcinoid tumors. Acta Oncol 1991; 30: 519 – 522.
`33. Jakobsen M, Hansen LE, Kolmannskog F, Schrumpf E, Vatn MH,
`Bergan A. Interferon-a 2b, with or without prior hepatic artery embo-
`lization: clinical response and survival in midgut carcinoid patients.
`Scand J Gastroenterol 1995; 30: 789 – 796.
`34. Fjallskog ML, Sundin A, Westlin JE et al. Treatment of malignant
`endocrine pancreatic tumors with a combination of alpha-interferon
`and somatostatin analogs. Med Oncol 2002; 19: 35 – 42.
`35. Frank M, Klose KJ, Wied M et al. Combination therapy with octreo-
`tide and alpha-interferon: effect on tumor growth in metastatic endo-
`crine gastroenteropancreatic tumors. Am J Gastroenterol 1999; 94:
`1381– 1387.
`36. Kolby L, Persson G, Franzen S, Ahren B. Randomized clinical trial of
`the effect of interferon alpha on survival in patients with disseminated
`midgut carcinoid tumours. Br J Surg 2003; 90: 687– 693.
`randomized,
`37. Faiss S, Pape UF, Bohmig M et al. Prospective,
`multicenter trial on the antiproliferative effect of lanreotide, interferon
`alfa, and their combination for therapy of metastatic neuroendocrine
`gastroenteropancreatic
`tumors—the
`International Lanreotide
`and
`Interferon Alfa Study Group. J Clin Oncol 2003; 21: 2689 – 2696.
`
`000006
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket