throbber
REVIEW
`
`Neuroendocrine tumours
`
`Endocrine-Related Cancer (2004) 11 1–18
`
`M T Barakat, K Meeran and S R Bloom
`
`Department of Metabolic Medicine, Division of Investigative Science, Imperial College London at Hammersmith
`Campus, Du Cane Road, London W12 ONN, UK
`
`(Requests for offprints should be addressed to S R Bloom; Email: s.bloom@imperial.ac.uk)
`
`Abstract
`
`carcinoid,
`for example,
`Neuroendocrine tumours are a heterogeneous group including,
`gastroenteropancreatic neuroendocrine tumours, pituitary tumours, medullary carcinoma of
`the
`thyroid and phaeochromocytomas. They have attracted much attention in recent years, both because
`they are relatively easy to palliate and because they have indicated the chronic effect of the particular
`hormone elevated. As neuroendocrine phenotypes became better understood,
`the definition of
`neuroendocrine cells changed and is now accepted as referring to cells with neurotransmitter,
`neuromodulator or neuropeptide hormone production, dense-core secretory granules, and the
`absence of axons and synapses. Neuroendocrine markers, particularly chromogranin A, are invaluable
`diagnostically. Study of several neuroendocrine tumours has revealed a genetic etiology, and
`techniques such as genetic screening have allowed risk stratification and prevention of morbidity in
`patients carrying the particular mutation. Pharmacological
`therapy for these often slow-growing
`tumours, e.g. with somatostatin analogues, has dramatically improved symptom control, and
`radiolabelled somatostatin analogues offer targeted therapy for metastatic or inoperable disease. In
`this review, the diagnosis and management of patients with carcinoid, gut neuroendocrine tumours,
`multiple endocrine neoplasia types 1 and 2, and isolated phaeochromocytoma are evaluated.
`
`Endocrine-Related Cancer (2004) 11 1–18
`
`Introduction and definition
`
`The definition of a neuroendocrine cell has changed over
`the last few years as our understanding and experimental
`techniques have advanced. In 1969, Pearse proposed the
`APUD (amine precursor uptake and decarboxylation)
`classification to describe cells producing polypeptide
`hormones and biogenic amines identical to those found
`in neurons (Pearse 1968, 1969). These cells would make up
`the tightly coordinated diffuse neuroendocrine system,
`present either in endocrine organs or dispersed through-
`out the body (Polak & Bloom 1979). The gastroentero-
`pancreatic system alone provides the richest source of
`regulatory peptides outside the brain (Polak & Bloom
`1986a). Indicating a unified origin to these cells, it was
`originally thought that they all derived from neuroecto-
`derm, but increasingly this was found not to be the case
`for all neuroendocrine cells (Le Douarin 1988). The
`following criteria are now generally accepted as defining
`neuroendocrine cells (Langley 1994): the production of a
`neurotransmitter, neuromodulator or neuropeptide hor-
`mone; the presence of dense-core secretory granules from
`which the hormones are released by exocytosis in response
`
`to an external stimulus; and the absence of axons and
`synapses. In practical
`terms, molecular markers are
`invaluable in defining neuroendocrine cells, and in
`particular chromogranin A (Winkler & Fischer-Colbrie
`1992, Taupenot et al. 2003). More recently, the subtilase
`proprotein convertases (SPC), particularly SPC2 and
`SPC3, have been added to the list of useful markers
`(Bergeron et al. 2000). With the discovery of neuroendo-
`crine phenotypes developing in cells such as immunocytes
`and certain neoplastic cells such as small cell carcinomas,
`it has recently been proposed that activation of specific
`genetic switches leading to neuroendocrine phenotypes
`should also be included in the definition (Day & Salzet
`2002).
`Neuroendocrine tumours are therefore a very hetero-
`geneous group arising from these neuroendocrine cells,
`and include carcinoid, non-carcinoid gastroenteropan-
`creatic tumours (such as insulinoma, gastrinoma and
`VIPoma
`(VIP,
`vasoactive
`intestinal
`polypeptide)),
`catecholamine-secreting tumours (phaeochromocytomas,
`paragangliomas, ganglioneuromas, ganglioneuroblasto-
`mas, sympathoblastoma, neuroblastoma), medullary car-
`cinoma of the thyroid, chromophobe pituitary tumours,
`
`Endocrine-Related Cancer (2004) 11 1–18
`1351-0088/04/011–1 # 2004 Society for Endocrinology Printed in Great Britain
`
`Online version via http://www.endocrinology.org
`
`Ex. 1072-0001
`
`

`

`Barakat et al.: Neuroendocrine tumours
`
`lung cancer and Merkel cell tumours. The
`small cell
`World Health Organization’s definition of neuroendo-
`crine tumours is ‘morphofunctional’ and is primarily
`based on microscopic characteristics, but incorporates
`immunohistological data (with such markers as the
`chromogranins, synaptophysin and non-specific enolase),
`special stains (e.g. silver), in addition to immunohisto-
`chemical stains for specific hormones which result in
`endocrine hyperfunction syndromes (Solcia et al. 2000).
`For the purposes of this review, the following topics are
`covered in the vast field of neuroendocrine tumours:
`etiology, general markers and the specific syndromes
`related to a limited subset of neuroendocrine tumours,
`namely
`carcinoid and other
`gastroenteropancreatic
`neuroendocrine tumours, multiple endocrine neoplasia
`types 1 and 2, and phaeochromocytoma.
`
`Etiology
`
`The genetic predisposition to certain neuroendocrine
`tumours has generated much interest, particularly with
`advanced techniques facilitating the identification of
`mutated genes. Similarly, with analysis of gene expression
`microarray profiles and multivariate analysis of complex
`traits (Phillips & Belknap 2002), important information
`can be used to prognosticate and risk stratify patients. A
`plethora of genes are known to be involved in neuroendo-
`crine tumorigenesis, including MEN1, RET, VHL, TSC1
`and TSC2 (Calender 2000), with mutations in MEN1
`remaining the most common form of genetic predisposi-
`tion to neuroendocrine tumours. Despite the lack of direct
`evidence, it is likely that neuroendocrine tumorigenesis is
`similar to that of the well-studied colorectal carcinoma
`model of a series of multiple genetic alterations leading to
`activation of oncogenes and/or inactivation of tumour
`suppressor genes and failure of apoptosis (Fearon &
`Vogelstein 1990, Shannon & Iacopetta 2001). Intrigu-
`ingly,
`the exact
`sequence of events
`is crucial
`for
`determining the phenotype, as demonstrated by the effect
`of early loss of Lkb1 in protection against transformation
`in Peutz-Jeghers syndrome (Bardeesy et al. 2002).
`In multiple endocrine neoplasia type 1 (MEN1),
`germline inactivating mutations in the tumour suppressor
`MEN1 gene (chromosome 11q13; Larsson et al. 1988) are
`found in 95% of patients. Somatic mutations of MEN1
`are also found in sporadic cases: 21% of parathyroid
`adenomas, 33% of gastrinomas, 17% of insulinoma, 36%
`of bronchial carcinoid, and 50–70% of sporadic thymic
`and duodenal carcinoid (Jakobovitz et al. 1996, Lubensky
`et al. 1996, Emmert-Buck et al. 1997, Marx et al. 1998,
`1999). The absence of detectable mutations in MEN1 may
`reflect deficiencies of current technology, or that the
`inactivation process is via non-mutation mechanisms such
`
`as methylation in the CpG-rich 50-region of MEN1.
`Indeed, such hypermethylation is found in the VHL
`promoter in von Hippel Lindau (Herman et al. 1994,
`Prowse et al. 1997).
`In keeping with Knudson’s model (Knudson 1971,
`Pannett & Thakker 2001), a two-hit process is required for
`MEN1 tumorigenesis (Larsson et al. 1988, Thakker et al.
`1989, Bystrom et al. 1990). Familial and somatic MEN1
`mutations differ in terms of the former usually presenting
`with tumour expression at an earlier age, multiple organs
`affected, and multiple tumours in one organ (Marx et al.
`1999). Although many mutations have been found in
`MEN1,
`there seems to be poor genotype–phenotype
`correlation (Kouvaraki et al. 2002). The majority of
`patients with familial MEN1 will develop non-functioning
`pancreatic tumours, while 40% will develop gastrinoma,
`10% insulinoma and 2% other functioning pancreatic
`tumours, such as glucagonoma, VIPoma and somatosta-
`tinoma (Marx et al. 1999).
`Based on Fearon and Vogelstein’s (1990) genetic
`model for colorectal tumorigenesis, Fig. 1 illustrates the
`possible pathways from induction to metastases for
`neuroendocrine
`tumours
`(Calender 2000),
`including
`MEN1, MEN2 and phaeochromocytoma.
`
`General markers for neuroendocrine
`tumours
`
`The group of neuroendocrine tumours can be character-
`ized both by general and specific markers, the most
`strikingly consistent general markers being the chromo-
`granins. Other general staining markers include pancrea-
`tic polypeptide, neuron-specific enolase (NSE), peptide
`histidine-methionine and human chorionic gonadotro-
`phin subunits (Tapia et al. 1981, Hamid et al. 1986,
`Yiangou et al. 1987). Chromogranin A (CgA), a secretory
`granule, is located alongside specific hormones in large
`dense-core vesicles of neuronal and neuroendocrine cells.
`Chromogranin B (CgB) is also widely distributed in
`neuroendocrine cells, although CgA seems to be more
`widespread (Winkler & Fischer-Colbrie 1992). Staining
`for
`the
`chromogranins
`in different neuroendocrine
`tumours
`is
`summarized in Table 1 (adapted from
`Taupenot et al. 2003), where CgA seems ubiquitous with
`the exception of the CgB-staining pituitary lactotrophs
`and some pancreatic beta-cell tumours.
`In one study, plasma CgA was elevated in 94% of
`endocrine pancreatic tumours, and pancreatic polypeptide
`in 74% (Eriksson et al. 1990). Elevated CgA levels were
`even more frequent (99%) in malignant carcinoid and
`gastroenteropancreatic tumours (99%) (Stridsberg et al.
`1995), with the highest levels seen in metastatic carcinoid
`(particularly midgut). Plasma CgB (and its 74-amino-acid
`
`2
`
`www.endocrinology.org
`
`Ex. 1072-0002
`
`

`

`INITIATION
`
`TRANSFORMATION
`PROLIFERATION
`
`Acetylation
`Methylation
`TGF-α
`
`MALIGNANT EVOLUTION
`
`deficiency in
`MMR
`(mismatch
`repair)
`
`METASTASIS
`
`Normal
`neuroendocrine
`cells
`
`Hyperplastic
`cells
`
`Dysplastic cells
`
`Well differentiated
`tumour
`
`Moderately
`differentiated tumour
`
`Poorly differentiated
`tumour
`
`Metastasis
`
`Endocrine-Related Cancer (2004) 11 1–18
`
`Inactivation of MEN1 (1st hit) VHL, NF1, TSC1, TSC2,
`?3q, ?1q
`
`
`Activation of RET (MEN-2)
`
`Growth factors:
`? NGF, TGF , bFGF, VEGF
`MEN1 (2nd hit)
`
`activation of oncogenes
`e.g. c-myc, K-ras
`
`Large LOHs
`3p-, 1p-, 18q-, 17p-, 8p-
`Loss of tumour suppressors e.g. PTEN
`Loss of apoptosis gene(s)
`Chromosomal instability
`
`Loss of adhesion (CD44, NCAMs)
`Oncogene activation
`?VEGF induction
`? ineffective nm23 (MEN1)
`
`Figure 1 Process of tumorigenesis from initiation to metastatic cancer in neuroendocrine tumours. Based on Calender (2000) and
`Fearon and Vogelstein (1990). Genes: MEN1, multiple endocrine neoplasia type 1; VHL, von Hippel Lindau; NF1, neurofibromatosis
`type 1; TSC1 and TSC2, tuberous sclerosis genes 1 and 2; PTEN, tumour suppressor gene PTEN (‘phosphatase and tensin
`homolog, deleted on chromosome 10’). NGF, nerve growth factor; TGF, transforming growth factor; bFGF, basic fibroblast growth
`factor; VEGF, vasculoendothelial growth factor; NCAM, neural cell adhesion molecule; nm23, tumour metastasis suppressor nm23
`(in MEN1); LOH, loss of heterozygosity.
`
`fragment termed GAWK) is usually a better marker for
`benign insulin-producing tumours (Sekiya et al. 1989,
`Yasuda et al. 1993). Finally, multiple hormones may be
`secreted by some tumours (Wood et al. 1983), and up to
`62% have elevated gastrin despite only 30% presenting
`with peptic ulcer disease.
`
`Although these two categories differ in specific histology
`and location, there are many diagnostic and therapeutic
`similarities between them, and these will be covered in the
`sections below. In general, neuroendocrine tumours are
`slow-growing. The classification into benign and malignant
`depends on various features summarized in Table 2 (Rindi
`et al. 1998).
`
`Specific syndromes
`
`Carcinoid and other gut neuroendocrine
`tumours
`
`Over the last few years, a new classification of neuroendo-
`crine gastroenteropancreatic tumours (GEPs) has been
`developed based on clinicopathologic features (Capella et
`al. 1994, Kloppel & Heitz 1994, Rindi et al. 1998). Tumours
`are termed functioning neuroendocrine tumours according
`to their leading clinical and endocrine profile, while those
`without plasma hormone elevation and lacking endocrine
`symptoms are labelled non-functioning neuroendocrine
`tumours (Taheri & Meeran 2002). In excess of 50% of
`neuroendocrine tumours are of the ’carcinoid’ type, with
`the remainder being mostly pancreatic and including
`insulinomas, gastrinomas, VIPomas and glucagonomas.
`
`Carcinoid
`Mostly derived from serotonin-producing enterochrom-
`affin (EC, or Kulchitsky’s) cells, these tumours termed
`’carcinoid’ have a wide clinical spectrum of presentation
`and symptomatology (McStay & Caplin 2002). Indeed, it
`is felt by some that the term should be made archaic
`because of this wide spectrum (Gilligan et al. 1995). Fewer
`than 10% of patients with carcinoid suffer from the
`classical carcinoid syndrome of flushing, hypotension,
`diarrhoea, wheezing and heart disease. These symptoms
`seem to be related directly to serotonin levels (elevated in
`93–94% of 600 patients with carcinoid syndrome),
`although serotonin can still be elevated in asymptomatic
`individuals (elevated in 25–30% of 7000 asymptomatic
`patients) (Vinik 2001). High serotonin levels seem to
`predict the development of carcinoid heart disease (Moller
`
`www.endocrinology.org
`
`3
`
`Ex. 1072-0003
`
`

`

`Barakat et al.: Neuroendocrine tumours
`
`Table 1 Detection of granins in neuroendocrine tumours. Adapted from Taupenot et al. (2003).
`
`Chromogranin A
`
`Chromogranin B
`
`Secretogranin III
`
`Carcinoid
`Gastrinoma
`Glucagonoma
`Insulinoma
`PPoma
`Somatostatinoma
`VIPoma
`Non-functioning islet cell carcinoma
`
`Corticotrophinoma
`Gonadotrophinoma
`Somatotrophinoma
`Thyrotrophinoma
`Prolactinoma
`Non-functioning pituitary adenoma
`
`Phaeochromocytoma
`
`Parathyroid adenoma
`
`Ganglioneuroblastoma
`Ganglioneuroma
`Neuroblastoma
`Medulloblastoma
`Paraganglioma
`
`Medullary thyroid carcinoma
`
`Prostate tumour with neuroendocrine differentiation
`Breast tumour with neuroendocrine differentiation
`
`ND, not detected.
`
`+
`+
`+
`+
`+
`+
`+
`+
`
`+
`+
`+
`+
`
`+
`
`+
`
`+
`
`+
`+
`+
`+
`+
`
`+
`
`+
`+
`
`+
`+
`+
`+
`ND
`ND
`ND
`ND
`
`+
`+
`+
`+
`+
`+
`
`+
`
`+
`
`ND
`ND
`+
`ND
`+
`
`+
`
`+
`+
`
`+
`ND
`+
`+
`ND
`ND
`ND
`ND
`
`+
`+
`+
`+
`+
`+
`
`+
`
`+
`
`ND
`+
`+
`ND
`+
`
`+
`
`+
`+
`
`et al. 2003). Other carcinoid tumours deriving from the
`gastric histamine-secreting enterochromaffin-like (ECL)
`cells produce an ‘atypical’ carcinoid syndrome. These
`tumours, which have low serotonin levels, frequently
`secrete the serotonin precursor 5-hydroxytryptophan (5-
`HTP). This produces the ‘atypical’ symptoms seen in
`foregut carcinoids with more intense and protracted
`purplish flushing; the limbs as well as the upper trunk
`are often affected and frequently result in telangiectasia
`(Snow et al. 1955, Sandler & Snow 1958, Vinik 2001).
`From the most recent and largest US epidemiology
`series (using the 1992–1999 cohort), the age-adjusted
`incidence of carcinoid tumours varies between 2.47 and
`4.48 per 100 000 population, with the rates being highest
`
`Table 2 The proposed classification into benign, uncertain and
`malignant gastroenteropancreatic tumours (Rindi et al. 1998).
`
`Tumour characteristic
`
`Size of tumour (cm)
`Local spread
`Vascular invasion
`Nuclear atypia
`Gross invasion
`Metastases
`
`Benign
`2
`No
`No
`No
`No
`No
`
`Uncertain Malignant
`
`> 2
`Yes
`Yes
`Yes
`No
`No
`
`> 2
`Yes
`Yes
`Yes
`Yes
`Yes
`
`in black males, then black females, then white Americans
`(Modlin et al. 2003). In this series,
`the population-
`corrected male-to-female ratio for all carcinoid sites was
`0.86, and the average age at diagnosis for all carcinoids
`was 61.4 years (compared with 63.9 years for non-
`carcinoid tumours). Table 3 summarizes the frequency
`of carcinoid at different sites (most occurring in the gut at
`67.5%, then the lung at 25.3%), with the associated 5-year
`survivals. Predisposition to metastatic spread depends on
`location and size (Lauffer et al. 1999, Modlin et al. 2003).
`In addition to the general markers mentioned above,
`the specific markers for carcinoid include urinary 5HIAA,
`neuropeptide K, substance P and other tachykinins.
`Urinary 5-hydroxyindole acetic acid (5HIAA) and neu-
`ropeptide K show high sensitivity in midgut carcinoid,
`with less diagnostic usefulness in foregut and hindgut
`carcinoid (Janson et al. 1997). If a diagnosis of carcinoid is
`suspected with normal baseline urinary testing,
`the
`pentagastrin test with measurements of plasma tachykinins
`is helpful (Norheim et al. 1986).
`
`Non-carcinoid gastroenteropancreatic
`neuroendocrine tumours
`Most of the non-carcinoid gastroenteropancreatic neuro-
`endocrine tumours arise in the pancreas. Making up less
`
`4
`
`www.endocrinology.org
`
`Ex. 1072-0004
`
`

`

`Table 3 Carcinoid tumours – relation between site, metastatic potential and 5-year survival. Data from the 1992–1999 cohort
`(Modlin et al. 2003).
`
`Endocrine-Related Cancer (2004) 11 1–18
`
`% of all
`carcinoids
`
`Regional
`metastases
`(%)
`
`Distant
`metastases
`(%)
`
`5-year
`survival with
`no metastases
`(%)
`
`5-year
`survival with
`regional metastases
`(%)
`
`5-year
`survival with
`distant metastases
`(%)
`
`25.3
`
`28.2
`2.4
`7.6
`18.5
`5.9
`1.4
`
`5.2
`
`35.9
`28.9
`25.8
`2.2
`3.1
`5.2
`
`0.5
`
`22.4
`9.9
`29.5
`1.7
`6.5
`0.5
`
`81.1
`
`59.9
`80.8
`76
`90.8
`69.1
`90.9
`
`76.7
`
`72.8
`88.1
`71.6
`48.9
`N/A
`N/A
`
`25.6
`
`50
`9.6
`30
`32.3
`21.2
`28.3
`
`Site
`
`Lung
`Small
`intestine
`Appendix
`Colon
`Rectum
`Stomach
`Ovary
`
`N/A, not available.
`
`than half of all neuroendocrine tumours and only 1–2%
`of all pancreatic tumours, pancreatic neuroendocrine
`tumours (PET) form an important group with a better
`prognosis than non-neuroendocrine tumours. Deriving
`from the diffuse neuroendocrine system of the gut (Polak
`& Bloom 1986a), PETs were formerly classified as
`APUDomas (tumours of the amine precursor uptake
`and decarboxylation system), and can secrete a vast
`number of hormones depending on the cell of origin.
`Physiologically,
`these hormones are involved in an
`intricate network of autocrine, paracrine, endocrine and
`neurotransmitter communication.
`Although the annual incidence of PETs is approxi-
`mately 3.5 to 4 per million population, post-mortem data
`suggests a much higher incidence. Indeed they have been
`detected in 0.3% to 1.6% of unselected autopsies in which
`only a few sections of pancreas were examined, and in up
`to 10% of autopsies in which the whole pancreas was
`systematically inspected (Kimura et al. 1991).
`Depending on whether secreted hormone is detectable
`and associated symptoms are present, gastroenteropan-
`creatic neuroendocrine tumours (GEPs) can be divided
`into ‘functioning’ and ‘non-functioning’ tumours. The
`‘functioning’ tumours may secrete several peptides, only
`some of which may produce symptoms. Similarly, a
`tumour which originally secreted one peptide may de-
`differentiate to co-secrete other peptides. Typically, these
`tumours are slow-growing, and often morbidity is from
`the secreted hormone (or hormones) rather than tumour
`bulk. The presence of a specific endocrine hyperfunction
`syndrome seems to be as important as purely pathological
`features for predicting tumour behaviour (Kloppel &
`Heitz 1988, Solcia et al. 1990) (see Table 4 taken from
`data from Aldridge & Williamson 1993, Arnold et al.
`2000, Schindl et al. 2000, Jensen 2001, Taheri et al. 2001).
`In the case of ‘non-functioning tumours’, it is accepted
`that there may be secreted, but as yet undetectable,
`peptides. These non-functioning tumours tend to be more
`
`aggressive and often present after metastasizing with
`symptoms of tumour bulk (Legaspi & Brennan 1988).
`The following are specific markers for functioning
`non-carcinoid GEPs: fasting hormones such as gastrin,
`glucagon, pancreatic polypeptide, somatostatin, neuro-
`tensin, and vasoactive intestinal polypeptide (VIP), and
`random parathyroid hormone-related peptide
`levels
`(PTHrP) (with simultaneous calcium and parathyroid
`hormone (PTH) measurements). In the case of gastrin, the
`patient has to be off proton pump inhibitors for at least
`two weeks and off H2-blockers for at least three days.
`Caution, however, is required if the clinical likelihood of
`gastrinoma is high, since there is a high risk of peptic ulcer
`perforation when medical therapy is stopped for the
`gastrin test. Even on proton pump inhibitors, very high
`gastrin levels (> 500 pg/ml or > 250 pmol/l) are indicative
`of gastrinoma, and repeat testing off therapy should not
`be recommended (Ashrafian et al. 2002). Differential
`diagnoses, which include atrophic gastritis, hypercalcae-
`mia and renal impairment, may be excluded by measuring
`basal acid output: spontaneous basal acid outputs of 20 to
`25 mmol/h are almost diagnostic and > 10 mmol/h
`suggestive. If the test results are equivocal, the secretin
`test is helpful: a rise of gastrin (instead of the normal fall)
`in response to intravenous secretin of greater than 200 pg/
`ml
`(100 pmol/l) has a sensitivity of 80–85% for
`gastrinoma (McGuigan & Wolfe 1980, Frucht et al. 1989).
`The dynamic test for the diagnosis of an insulinoma is
`a three-day fast, allowing unlimited non-caloric fluids
`(Service 1995, Service & Natt 2000). Elevated plasma
`insulin and C-peptide levels are diagnostic in the presence
`of hypoglycaemia (glucose below 2.2 mmol/l (40 mg/dl)),
`and this is achieved by 48 h of the fast in > 95% of
`insulinomas (Friesen 1987). If no hypoglycaemia is
`achieved by the end of the fast, the sensitivity can be
`further increased by exercising the patient for 15 min. The
`fast is terminated after the exercise period, or prior to this
`if hypoglycaemia is achieved, with simultaneous plasma
`
`www.endocrinology.org
`
`5
`
`Ex. 1072-0005
`
`

`

`Barakat et al.: Neuroendocrine tumours
`
`Hypoglycaemia
`Weight gain
`Abdominal pain
`Diarrhoea
`Peptic ulceration
`Secretory diarrhoea
`Hypokalaemia
`Achlorhydria
`Metabolic acidosis
`Flushing
`Necrolytic migratory erythema
`Diabetes
`Cachexia
`Thromboembolic disease
`Pain
`Weight loss
`Diarrhoea
`Steatorrhoea
`Diabetes
`Gallstones
`Weight loss
`Classical carcinoid
`Flushing
`Diarrhoea
`Wheeze
`Cardiac fibrosis
`Pellagra dermatosis
`Cushing’s syndrome
`Pigmentation
`Hypercalcaemia
`Nephrolithiasis
`Nephrocalcinosis
`Osteoporosis
`Diarrhoea, flushing
`Acromegaly
`
`Symtoms of tumour bulk
`Weight loss
`
`Glucagonoma
`
`1–2
`
`Pancreas 100%
`
`1–20
`
`> 70
`
`PPoma
`
`<1
`
`Pancreas 100%
`
`Somatostatinoma
`
`<1
`
`Pancreas 55%
`Duodenum + jejunum 44%
`
`Carcinoid
`
`<1 mostly extrapancreatic
`
`Midgut 75–87%
`Foregut 2–33%
`Hindgut 1–8%
`Unknown 2–15%
`
`18–44
`
`45
`
`> 60
`
`> 50
`
`Rare
`
`90
`
`Corticotrophinoma
`CRFoma
`PTHrPoma
`
`<1
`
`<1
`
`Calcitoninoma
`Somatotrophinoma
`
`<1
`
`‘‘Non-functioning’’
`
`<1
`
`Pancreas 4–14% (of all ectopic ACTH)
`
`Rare
`
`Pancreas
`
`Pancreas
`Pancreas 30%
`Lung 54%
`Jejunum 7%
`Pancreas + gastrointestinal tract
`
`Rare
`
`16
`16
`
`18–44
`
`> 99
`
`> 99
`
`> 80
`50
`
`> 80
`
`PPoma, pancreatic polypeptide secreting tumour; CRF, corticotrophin releasing factor.
`
`www.endocrinology.org
`
`6
`
`Table 4 Tumour syndromes of gastrointestinal neuroendocrine tumours (NET). Adapted from Aldridge & Williamson (1993), Arnold et al. (2000), Jensen (2001), Schindl et al.
`(2000) and Taheri et al. (2001). The percentage with MEN1 of each tumour is independent of family history.
`
`Tumour
`
`Insulinoma
`
`Gastrinoma
`
`Frequency of pancreatic
`neuroendocrine tumours (%)
`
`70–75
`
`20–25
`
`VIPoma
`
`3–5
`
`Tumour location
`
`Pancreas > 99%
`
`Duodenum 70%
`Pancreas 25%
`
`Pancreas 90%
`
`% with MEN1
`
`Malignancy (%)
`
`Clinical syndrome
`
`4–5
`
`20–25
`
`6
`
`<10
`
`> 50
`
`> 50
`
`Ex. 1072-0006
`
`

`

`Endocrine-Related Cancer (2004) 11 1–18
`
`Table 5 Imaging modalities for gastroenteropancreatic neuroendocrine tumours.
`
`Somatostatin- receptor- scintigraphy
`
`CT
`
`Endoscopic ultrasound of pancreatic neuroendocrine
`tumours
`
`Visceral angiography
`
`Sensitivity 90% (excluding insulinoma)
`Specificity 80% (excluding insulinoma)
`Sensitivity 10–50% for insulinoma
`
`Useful preoperatively for localizing tumours, but difficulty detecting
`tumours <1 cm
`Sensitivity of 29% in one study for detecting pancreatic insulinoma
`compared with endoscopic ultrasound
`
`Sensitivity 80–90%
`Most sensitive for detecting insulinomas (94%)
`Smallest lesion detectable 5 mm in pancreatic head
`Lower sensitivities for extrapancreatic tumours (50%)
`
`Helpful for subcentimetre tumours, where a tumour blush is seen
`Futhermore, can be combined with calcium stimulation in
`individual pancreatic arteries with venous sampling from the hepatic
`vein (see text for details)
`
`MRI
`
`In general no more helpful than CT
`
`Positron emission tomography with 11C-serotonin
`precursor 5-HTP instead of 18F-deoxyglucose
`
`Selective uptake in carcinoid with high resolution allowing liver
`and lymph node lesion detection
`
`Intraoperative ultrasound for pancreatic tumours
`
`Additional invaluable information for the surgeon operating on the
`delicate pancreas
`
`Intraoperative gamma probes with somatostatin
`analogue (111InDTPA-D-Phe1)-pentetreotide tracer
`
`Recently introduced, allows detection of lesions <5 mm and
`identified 57% more lesions than the ‘‘palpating finger’’
`
`MIBG for gastroenteropancreatic neuroendocrine tumours
`
`Very few gastroenteropancreatic tumours take up MIBG, since
`they derive from endoderm and not neuroectoderm
`
`and urine samples for sulphonylurea analysis, which must
`be shown to be negative for the diagnosis of insulinoma
`(Todd & Bloom 2002). Finally, the reader needs to be
`aware of the various insulin assays: the competitive and
`increasingly used two-site noncompetitive immunoassays.
`The main problems relate to the variable specificity, with
`some detecting proinsulin, the interference from insulin-
`degrading enzymes
`in haemolysed samples and the
`presence of anti-insulin antibodies (Sapin 2003)
`
`Radiological localization of carcinoid and other gut
`neuroendocrine tumours
`The Delphi consensus has recently been published for
`standardizing the diagnostic imaging of neuroendocrine
`(Ricke et al. 2001). Somatostatin receptor
`tumours
`scintigraphy (SRS)
`forms
`the mainstay of
`imaging
`techniques, with a sensitivity of up to 90% and specificity
`of 80% (Krenning et al. 1993, Lebtahi et al. 1997,
`Termanini et al. 1997, Kwekkeboom & Krenning 2002),
`although only 10 to 50% of insulinomas are SRS-positive
`(Modlin & Tang 1997, Warner & O’dorisio 2002). Table 5
`summarizes the different modalities and their usefulness,
`including computed tomography (CT), endoscopic ultra-
`sonography (EUS) (Lightdale et al. 1991, Glover et al.
`1992, Palazzo et al. 1992, Rosch et al. 1992, de Kerviler et
`al. 1994, Thompson et al. 1994, Ruszniewski et al. 1995,
`Zimmer et al. 2000), positron emission tomography
`
`(Eriksson et al. 2000, 2002, Warner & O’dorisio 2002),
`intraoperative ultrasound and intraoperative gamma
`probes with the tracer (111)In DTPA-D-Phe1-pentetreo-
`tide (Adams & Baum 2000). Given that GEPs derive from
`endoderm and not catecholamine-producing neuroecto-
`derm, there is little place for 131I-metaiodobenzyl-guani-
`dine (MIBG) in GEP imaging (European Neuroendocrine
`Tumour Network (ENET) 2000).
`With pancreatic tumours, the surgeon requires as much
`information regarding location as possible. Selective
`angiography with secretagogue injection into the main
`pancreatic arteries allows biochemical
`in addition to
`angiographic localization. In this procedure, the main
`pancreatic arteries (gastroduodenal, superior mesenteric,
`inferior pancreaticoduodenal and splenic) are cannulated
`separately and examined for a tumour blush. A secretago-
`gue (calcium has taken the place of secretin; Turner et al.
`2002) is injected into each of these arteries individually, and
`venous samples are collected from the hepatic vein for
`biochemical analysis of
`the suspected hypersecreted
`hormone (e.g. gastrin or insulin). In the presence of a
`tumour, the hormone levels double after 30 s, whereas the
`normal effect is a reduction in levels (Doppman et al. 1990,
`1991, Fedorak et al. 1993, Goldstone et al. 1996, O’Shea et
`al. 1996). The tremendous advantage of this technique is
`that it allows biochemical localization of the tumour (e.g. a
`very small
`insulinoma) even if the radiology appears
`
`www.endocrinology.org
`
`7
`
`Ex. 1072-0007
`
`

`

`Barakat et al.: Neuroendocrine tumours
`
`Table 6 The specific medical treatments for some neuroendocrine tumour syndromes.
`
`Hypersecretion syndrome
`
`Medical therapy
`
`Carcinoid
`
`Insulinoma
`
`Gastrinoma
`
`Glucagonoma
`
`VIPoma
`
`Somatostatinoma
`
`PTHrPoma
`
`Somatostatin analogue for SRS positive carcinoid
`Interferon- for SRS negative carcinoid
`Histamine antagonists (H1 and H2)
`Cyproheptadine, nicotinamide
`
`Frequent slow-release complex carbohydrate intake
`Guar gum
`Diazoxide
`Intravenous dextrose if period of fasting
`Somatostatin analogue if SRS positive (usually malignant)
`
`High dose proton pump inhibitor (life-long in patients with MEN1, since high recurrence
`rate with surgery)
`
`High dose somatostatin analogue
`Anticoagulation since associated with thrombophilia
`Insulin for diabetes mellitus
`
`High dose somatostatin analogue
`Aggressive intravenous rehydration in acute attack of diarrhoea
`Potassium + bicarbonate in acute attacks
`
`Pancreatic enzyme supplementation
`Insulin for diabetes mellitus
`
`High dose somatostatin analogue
`Intravenous rehydration
`Bisphosphonates for often life-threatening hypercalcaemia
`Somatostatin analogue
`
`Non-functioning
`
`Somatostatin analogue if SRS scan positive and progressive disease
`
`normal. Furthermore, with MEN1, different tumours may
`co-exist, and this technique will help to distinguish them
`biochemically. Finally, and perhaps the procedure’s great-
`est advantage is that it allows the detection of liver
`metastases, since the hepatic artery is always cannulated
`at the end of the procedure and a rise in hormone levels
`detectable in the hepatic vein after calcium injection into
`the hepatic artery is diagnostic of liver metastases.
`
`Medical treatment
`Until curative surgical treatment is performed, or if
`surgery is not indicated, medical treatment has to be
`initiated for symptom control. Table 6 summarizes the
`medical
`treatments
`for
`the various hypersecretion
`syndromes. The greatest impact for control of symptoms
`has been afforded by the somatostatin analogues in
`SRS-scan positive tumours, and it
`is known that
`somatostatin immunoreactivity is often found in neuro-
`endocrine tumours (Polak & Bloom 1986b). There are
`five somatostatin receptors (sstr1–5), but
`the longer-
`acting
`analogues,
`lanreotide
`and octreotide, bind
`preferentially to sstr2 and to a lesser extent to sstr5
`(Patel & Srikant 1994, Reisine & Bell 1995, de Herder &
`Lamberts 2002). The effectiveness of these analogues at
`reducing hormone production and tumour stabilization
`(Arnold et al. 1996, di Bartolomeo et al. 1996, Eriksson
`
`et al. 1997, Faiss et al. 1999) has been attributed to the
`sstr2 receptor (Patel & Srikant 1994, Buscail et al. 1995,
`Reisine & Bell 1995, Wulbrand et al. 1998). Given the
`low sensitivity of SRS imaging for insulinomas, it is not
`surprising that only 50% of insulinomas express sstr2
`(Wulbrand et al. 1998), and it therefore follows that
`long-acting somatostatin analogues benefit only those
`with
`sstr2-positive
`tumours.
`In
`‘non-functioning’
`tumours, somatostatin analogues are beneficial where
`the SRS scan is positive and there is evidence of tumour
`progression or high mitotic rate (European Neuroendo-
`crine Tumour Network (ENET) 2000). An exciting
`prospect
`in this field is
`the development of new
`somatostatin analogues that have differing specificities
`to the various somatostatin receptors,
`improving their
`therapeutic effectiveness
`in tumours
`that
`lack,
`for
`instance, sstr2. Until these are available, for progressive
`SRS-negative (i.e. sstr2/5 negative) carcinoid tumours,
`interferon-alpha should be the treatment of choice.
`
`Interferon-alpha and chemotherapy for gut
`neuroendocrine tumours
`Interferon-alpha has been used in the treatment of
`carcinoid for some time (Oberg et al. 1983) to control
`hormone secretion, symptoms and tumour growth. Many
`trials looking at its effectiveness in malignant and mostly
`
`8
`
`www.endocrinology.org
`
`Ex. 1072-0008
`
`

`

`Endocrine-Related Cancer (2004) 11 1–18
`
`Table 7 Non-medical treatment options for gastroenteropancreatic neuroendocrine tumours.
`
`Non-medical treatment
`
`Effectiveness
`
`Curative surgery
`Resection of primary
`Resection of resectable liver metastases
`Liver transplantation
`Hepatic arterial embolization
`
`Chemotherapy with hepatic arterial occlusion
`
`Hepatic chemoembolization
`
`Hepatic radioembolization
`
`Hepatic cryosurgery
`
`Palliative surgery
`
`Radio-labelled somatostatin analogues
`
`Best option if feasible
`Often pancreatic insulinomas can be enucleated easily
`70–80% 4–5 year survival with curative surgery
`In highly selected cases can improve survival
`Most effective for liver metastases from functioning neuroendocrine tumours
`producing symptoms. Significant reduction in symptoms (40–90%)
`Difficult to com

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