10.1007-s12022-013-9295-2.pdf

download 10.1007-s12022-013-9295-2.pdf

of 15

Transcript of 10.1007-s12022-013-9295-2.pdf

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    1/15

    Neuroendocrine Tumors of the Pancreas: Current Concepts

    and Controversies

    Michelle D. Reid &Serdar Balci &Burcu Saka &

    N. Volkan Adsay

    Published online: 16 January 2014# Springer Science+Business Media New York 2014

    Abstract In the past decade, the clinico-pathologic charac-

    teristics of neuroendocrine tumors (NETs) in the pancreas

    have been further elucidated. Previously termed islet celltumors/carcinomasorendocrine neoplasms, they are now

    called pancreatic NETs (PanNETs). They occur in relatively

    younger patients and may arise anywhere in the pancreas.

    Some are associated with von HippelLindau, MEN1, and

    other syndromes. It is now widely recognized that, with the

    exception of tumorlets (minute incipient neoplasms) that oc-

    cur in some syndromes like MEN1, all PanNETs are malig-

    nant, albeit low-grade, and although they have a protracted

    clinical course and overall 10-year survival of 6070 %, even

    low-stage and low-grade examples may recur and/or metasta-

    size on long-term follow-up. Per recent consensus guidelines

    adopted by both European and North American NET Socie-

    ties (ENETS and NANETs) and WHO-2010, PanNETs are

    now graded and staged separately, unlike previous classifica-

    tion schemes that used a combination of grade, stage, and

    adjunct prognosticators in an attempt to define benign be-

    havior or malignant categories. For staging, the ENETs

    proposal may be more applicable than CAP/AJCC, which is

    based on the staging of exocrine tumors. Current grading of

    PanNETs is based on mitotic activity and ki-67 index. Other

    promising prognosticators such as necrosis, CK19, c-kit, and

    others are still under investigation. It has also been recognized

    that PanNETs have a rather wide morphologic repertoire

    including oncocytic, pleomorphic, ductulo-insular, sclerosing,

    and lipid-rich variants. Most PanNETs are diagnosed by fine

    needle aspiration biopsy, in which single, monotonous

    plasmacytoid cells with fair amounts of cytoplasm and dis-

    tinctive neuroendocrine chromatin are diagnostic. Molecularalterations of PanNETs are also very different than that of

    ductal or acinar tumors. Loss of expression of DAXX and

    ATRX proteins has been recently identified in 45 %. Along

    with these improvements, several controversies remain, in-

    cluding grading, value of current cutoff ranges, and the best

    methods for counting ki-67 index (manual count by computer-

    captured image may be the most practical for the time being).

    More important is the controversial use of the term carcino-

    ma, which was previously employed in WHO-2004 only for

    invasive and metastatic cases but has now been made synon-

    ymous with grade 3 group of tumors. It is becoming clear that

    grade 3 group comprises two distinct categories: (1) differen-

    tiated but proliferatively more active tumors which typically

    have ki-67 indices in the 2050 % range and (2) true poorly

    differentiated NE carcinomas as defined in the lung, with ki-

    67 typically >50 %. Further studies are needed to address

    these controversial aspects of PanNETs.

    Keywords Pancreas.Neuroendocrine tumor. WDNET.

    PDNEC

    Introduction

    Many new developments have taken place in our understand-

    ing of the biology, morphologic, and molecular characteristics

    of neuroendocrine tumors of the pancreas. In 2010, the World

    Health Organization (WHO) proposed a new terminology for

    these tumors [1], which has had far-reaching effects on their

    current diagnosis and management. As a very important and

    positive development, the grading and staging parameters for

    these tumors have now been separated from their name and

    diagnostic sub-categories. In the ensuing article, we will

    M. D. Reid :S. Balci : B. Saka :N. V. Adsay

    Department of Pathology, Emory University School of Medicine,

    Atlanta, GA, USA

    N. V. Adsay (*)

    Department of Pathology, Emory University Hospital, 1364 Clifton

    Rd NE, Room H178, Atlanta, GA 30322, USA

    e-mail: [email protected]

    Endocr Pathol (2014) 25:6579

    DOI 10.1007/s12022-013-9295-2

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    2/15

    review these new developments, their limitations and contro-

    versies, as well as recent advances in the diagnosis, classifi-

    cation, and management of these tumors.

    Definition and Terminology

    Neuroendocrine tumors (NETs) of the pancreas are derivedfrom, and composed of, epithelial cells with phenotypic and

    ultrastructural neuroendocrine differentiation. A spectrum of

    neoplasms with neuroendocrine differentiation occurs in the

    pancreas, ranging from well-differentiated to undifferentiated.

    Differentiated examples are essentially counterparts of carci-

    noid tumors elsewhere, now designated as well-differentiated

    neuroendocrine tumors (WDNETs). The term carcinoid how-

    ever has not been employed in the pancreas, except in the case

    of metastases from the gastrointestinal tract or rare serotonin-

    producing examples. Instead, in the past, pancreatic neuroen-

    docrine tumors were termed islet cell tumors/carcinomasdue

    to their resemblance to islets of Langerhans, the endocrinecomponent of this organ. However, it is now presumed that

    these tumors do not necessarily arise fromthe islets, and in

    fact, many may actually originate from the neuroendocrine-

    type cells residing in the pancreatic ductal epithelium. They are

    also regarded as a part of the age-old APUDoma concept

    since they are fundamentally characterized by amineprecursor

    uptake and decarboxylation activity. Such APUD cells have

    endocrine, autocrine, paracrine, and neuromodulatory functions.

    In the WHO-2004 classification system, these tumors were

    referred to as pancreatic endocrine neoplasms. However, in

    the WHO-2010 system, endocrine tumor was modified to

    neuroendocrine tumorbased on the belief that the latter term

    was a more accurate indicator of the cells neural as well as

    endocrine phenotype and, more importantly, to distinguish

    them from the conventional endocrine cells of the adrenal,

    thyroid, and pituitary gland.

    Although it is widely agreed that the term neuroendocrine

    neoplasm would be a more accurate categorical designation for

    theseneoplasms, the term NETwas adopted by the WHO in

    2010 primarily because the acronym had already been incorpo-

    rated into the very names of the societies dedicated to studying

    them (i.e., the European Neuroendocrine Tumor Society

    (ENETS) and the North American Neuroendocrine Tumor

    Society (NANETs)). Thus, these tumors are currently referred

    to as pancreatic neuroendocrine tumors (PanNETs). The acro-

    nym P-NET is used by some authors; however, because the

    latter can be confused with the primitive neuroectodermal tumor

    acronym, PNET, the former designation is preferred.

    Poorly differentiated neuroendocrine carcinomas are cur-

    rently also included in the generic category of PanNETs and

    are recognized as the high-grade (grade 3 [G3]) version of

    these tumors. However many authors advocate making a

    distinction between ordinary (well-differentiated) PanNETs

    and these poorly differentiated neuroendocrine carcinomas,

    regarding them as a separate category in the pancreas (see

    the following discussion).

    It should be noted that, unlike the WHO-2004 system, in

    the WHO-2010 (Table1) as well as in current practice, the

    terminology of NETs is no longer modified by the tumors

    stage and grade. Previously, the term endocrine carcinoma

    was employed for cases showing extra-pancreatic spread ormetastasis. However, this was problematic because a percent-

    age of cases that were classified as benign demonstrated

    malignant clinical behavior. This necessitated a shift in the

    designation of cases that were originally classified as benign

    but in whom metastases were discovered shortly after. While

    this issue was resolved with the WHO-2010, another poten-

    tially problematic terminology shift was inadvertently created

    because in the latter classification carcinoma was made

    synonymous onlywith G3 (poorly differentiated) NETs. Un-

    fortunately some practitioners still use the term carcinoma

    for metastatic NETs even when they are well differentiated

    (G1 or 2 tumors).

    Clinical Aspects

    PanNETs are relatively rare tumors, accounting for 2 % of all

    pancreatic neoplasms and affecting one to two individuals per

    1,000,000/year. However, with improving technology, more

    and more cases come to attention as incidentalomas [2].

    Tumors are most frequently seen in adults but may rarely

    occur in children. Patients are typically 3060 years (with a

    mean of 50 years). Men and women are equally affected but

    high-grade (G3) carcinomas often occur in older males.

    PanNETs more commonly arise in the pancreatic head but

    can involve any part of the pancreas.

    WDNETs may also arise in a background of familial syn-

    dromes including von HippelLindau (VHL) syndrome, tu-

    berous sclerosis complex (TSC), neurofibromatosis type 1

    (NF1), and multiple endocrine neoplasia type 1 (MEN1). In

    Table 1 Classification system for pancreatic neuroendocrine tumors

    Modified from WHO 2010

    Differentiation Grade Mitosesa/10

    |HPF

    Ki-67b

    proliferation

    |index (%)

    Well-differentiated PanNET Grade 1 20

    WHOWorld Health Organization, HPFhigh power field, PanNETpan-

    creatic neuroendocrine tumor,PanNECpancreatic neuroendocrine carci-

    noma (large and small cell type)aIn at least 50 HPFsb MIB1 antibody

    66 Endocr Pathol (2014) 25:6579

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    3/15

    MEN1 and VHL, tumors are frequently multifocal and occur

    in a younger age group [3,4]. Up to 80 % of MEN1 patients

    develop multiple ( 0.5 cm)

    (Fig. 1) [3]. Up to 17 % of patients with VHL syndrome

    develop PanNETs. Most of these are non-functioning and

    60 % have clear cell morphology due to intracytoplasmic lipid[4,5]. Mutation of the VHLgene has been identified in these

    patients and plays a role in disease development. Although

    lipid-rich cells were once thought to be pathognomonic of

    VHL-related WDNETs, similar cells have now been identified

    in a subset of WDNETs that are either MEN1-related or non-

    syndrome-associated [6]. Up to 10 % of patients with NF1

    develop WDNETs, which have an increased affinity for the

    ampullary region.

    Cell Type and Functionality Status

    Based on clinical presentation, PanNETs were historically

    separated into two clinical categories, functional versus

    non-functional, based on the symptoms elicited by the pre-

    dominant hormone the tumors secrete into the bloodstream.

    Clinical presentation and prognosis were once felt to be

    strongly linked to tumor functionality. However, it has now

    become clear that many tumors secrete multiple hormones

    albeit with variable detectability and functional impact. Addi-

    tionally, tumors may change their hormone productivity over

    time. Moreover, with recent improvements in imaging modal-

    ities, more non-functional PanNETs are being discovered, and

    so the value of a functionality-based classification system has

    lost support.

    Functional tumors are still named based on primary clinical

    presentation and serologic activity (e.g., gastrinoma and

    insulinoma) and NOT on immunohistochemical hormone ex-

    pression. Insulinomas often present with the classicalWhip-

    ple triad due to excessive insulin production [7]. Gastrin

    overproduction is associated with ZollingerEllison syn-

    drome, while overproduction of vasoactive intestinal peptide

    (VIP) can cause severe watery diarrhea, hypokalemia, andachlorhydria, leading to death.

    Hormone status has also been shown to be associated with

    biologic behavior, which is indirectly linked to the stage at

    which a hormone-producing tumor causes clinical symptoms.

    For example, most insulinomas have benign behavior, which

    is largely attributable to their early detection. Over 70 % of

    insulinomas are 2 cm, they are often

    not detected early because glucagon levels are not clinically

    elevated. Having said this, it is possible that cell type may

    have additional yet to be defined biologic characteristics

    that lead to divergent prognoses in these tumors.

    Recently, small PanNETs occurring on the wall of pancre-

    atic ducts, usually of serotonin-producing type, have come to

    clinical attention due to duct obstruction sometimes mimick-

    ing intraductal papillary mucinous neoplasms [8,9].

    Classification and Sub-grouping

    In the previous WHO-2004 classification system, PanNETs

    had been sub-categorized as having benign behavior, as

    having uncertain behavior, or as frank carcinoma (with

    extrapancreatic spread or metastasis) based on an amalgam-

    ation of grade, stage, spread status, and adjunct prognostica-

    tors (vascular and perineural invasion). This was a fairly

    unique approach for any type of tumor in the body. However,

    in the WHO-2010 system, grade and stage were separated out,

    as is the case for tumors of other organs. The latter is discussed

    in detail in the following sections.

    Grading

    In the current 2010-WHO classification system, pancreatic neu-

    roendocrine tumors are divided into well-differentiated [grade 1

    (G1) and 2 (G2)] NETs (WDNETs) and poorly differentiated

    (G3) neuroendocrine carcinoma (PDNECs). This system gives

    the distinct impression that PDNECs are in continuum with

    Fig. 1 In the background of atrophy and hyperplasticislets, there is an

    evolving neuroendocrine tumor that is 3.0 mm in size (microadenoma)

    (hematoxylin & eosin stain, 200 magnification). This patient had a

    history of multiple endocrine neoplasia type 1 syndrome

    Endocr Pathol (2014) 25:6579 67

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    4/15

    ordinary (well-differentiated) PanNETs, which is not neces-

    sarily the case. Evolving evidence strongly suggests that G3

    tumors should be regarded separately and as such will be

    discussed separately later.

    The grading of NETs is based on two calculations: (1)

    mitotic count and (2) ki-67 labeling index (using the MIB1

    antibody). G1 NETs are defined as having a ki-67 index of

    50 %).

    At the other end of the spectrum, for the distinction of G1

    from G2 tumors, some groups advocate using 5 % as the cutoff,

    which may be a more accurate and reproducible cutoff. While

    further studies are needed to clarify this issue, the current 2 %

    cutoff remains widely used. Additionally, the current cutoff

    which is widely used and endorsed by the WHO-2010 is often

    referred to as the 2 % cutoff in publications; however, as

    recently clarified by NANETS, it is actually applied as

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    5/15

    prove to be a fairly reliable method. Of note, currently, there is

    a separate billing code for automated counting. The direct

    real-time eye-count approach, which is widely used in he-

    matology, has also proven difficult in grading PanNETs, unless

    a grid is used. However, even with a grid, cell distribution and

    overlap make the test difficult to perform. We have found that,

    in our current practice, the most practical, cost-effective, and

    reproducible method for counting ki-67-positive cells is amanual count done either on a computer screen or as a cap-

    tured static photomicrograph of tumor hot spots, where nega-

    tive and positive tumor cells are visualized and immediately

    crossed off once counted. The latter can be done with a digital

    camera attachedto themicroscope, a setup that is far less costly

    than automated methods and takes an average of 6 min [18].

    Naturally, tumor heterogeneity and subjectivity in hot spot

    determination are additional factors that may lead to variations

    in results. Whether to count the less intensely labeled cells as

    positive is also a question. For this, normalizationwith the

    background tissue is helpful. There should be no background

    staining in the stroma or cytoplasm in a properly performed ki-67 assay and light brown nuclei should generally be

    disregarded. In tumors with abundant lymphocytes or other

    confounding cell types, ki-67 indices may be falsely elevated.

    In such cases, dual staining with neuroendocrine markers may

    be necessary, especially if the labeling index appears to be

    close to a cutoff, in particular 20 %.

    Despite these challenges, ki-67 has nonetheless been

    shown in numerous studies to correlate highly with clinical

    outcome. Perhaps counter-intuitively, it has proven to be more

    practical and reproducible than mitotic count, and is as, if not

    more applicable, than other quantitative immunohistochemi-

    cal assays that are the norm in other organs such as the breast

    (ER/PR/her2neu) and stomach (EGFR).

    Staging

    The staging of PanNETs is now done separately from the

    grade, unlike the WHO-2004 system where the two were

    combined to determine the category.

    There are currently several different staging protocols for

    PanNETs. The one from The American Joint Commission on

    Cancer (AJCC)/Union of International Cancer Control

    (UICC)/College of American Pathologists (CAP) [1921]

    differs from that of the European Neuroendocrine Tumor

    Society (ENETS) [22] system (Table2). The AJCC has es-

    sentially adopted the staging used for exocrine tumors, which

    is problematic for pT3 tumors, which are defined in part by

    peripancreatic soft tissue involvement. Because the pancreas

    has very irregular lobules, no capsule, and fat placement

    throughout, it is often difficult, if not impossible, to assess

    peripancreatic soft tissueinvasion in these tumors [2325].

    Furthermore, most PanNETs protrude from the pancreas even

    when they are small, especially those located in the tail. That is

    probably why many studies have failed to identify a correla-

    tion between survival and T stage. In contrast, the ENETS

    staging system relies predominantly on more reproducible

    criteria such as tumor size rather than poorly defined and

    irreproducible parameters like peripancreatic extension.

    A recent study of 1,072 post-surgical PanNET patientsshowed that the ENETS system was superior to the

    AJCC/UICC system for stratifying risk of death and creating

    risk-based treatment guidelines [26,27]. Despite the proven

    discrepancies between the two staging systems, there appears

    to be no adverse effect on diagnosis and management [ 28]. In

    our current practice, we typically provide the stage by both

    schemes, separately.

    Early PanNETsIncipient Neoplasia/Dysplasia

    Extensive study of patients with MEN1 has led to the recogni-

    tion of so-called precursor lesions of PanNETs. MEN1 patients

    often develop proliferative or enlarged islets, some of which

    acquire hormonal and morphologic clonality and achieve a size

    beyond that of normal islets. The latter are regarded as dys-

    plastic and are akin to pulmonary tumorlets. Naturally, it is

    difficult to define the dysplasia and determine the point at

    which such proliferations become autonomous and neoplastic.

    These lesions often acquire an enveloping dense fibrous band,

    a feature of neoplastic transformation commonly encountered

    in endocrine organs. They also often show uniform staining

    with pancreatic hormones. Lesions that appear fully established

    but are

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    6/15

    It was in MEN1 syndrome that the idea of preneoplastic

    precursors of pancreatic NETs was first fashioned as it gave

    researchers the unique opportunity to study these tumors in

    their earliest form. The term nesidioblastosiswas coined by

    Laidlaw [30] who believed that pancreatic adenomas (islet cell

    tumors) were a secondary reactive process in ductal epitheli-

    um that occurred after an awakening of their islet and duct

    bui lding capab ility. He nam ed these pluripotent cellsnesidioblasts (Greek for islet builder) and used the term

    nesidioblastosis to describe islet cell proliferation and

    nesidioblastomafor islet cell adenomas.

    Others later proposed that nesidioblastosis might explain

    the patterns of disease seen in familial multiple endocrine

    adenomatosis (MEA) syndrome [31,32]. For almost a centu-

    ry, nesidioblastosis has remained a part of the dysplasia/early

    neoplastic transformation concept of PanNETs. Adult forms

    of diffusenesidioblastosis (abnormality of cells) have also

    been implicated in persistent hyperglycemia [33].

    Characteristics of OrdinaryPanNETs (WDNET)

    Fast-forward to the present day, where our knowledge of the

    clinical aspects, biology, and associated alterations in these

    tumors has expanded significantly: so how do we now view

    and evaluate PanNETs?

    Gross Features

    PanNETs can involve any part of the pancreas. They are

    typically well circumscribed and in sporadic cases are often

    solitary. Their appearance may vary due to the contents of the

    tumor. The vast majority are tan-brown and fleshy due to the

    dense cellularity and relative stromal paucity (Fig. 2). Few

    examples contain a more abundant stromal component and

    may appear more yellow-white and firm/sclerotic. Some cases

    have a complete capsule, but in most cases the capsule is

    incomplete or non-existent. The vast majority of PanNETs

    are solid tumors, but about 5 % present with cystic

    degeneration, typically with clear serosanguinous contents,

    and must be differentiated radiologically from cystic pancre-

    atic tumors. Some examples may have extensive blood (so-

    called peliotic variant) and appear grossly hemorrhagic.

    PanNETs, especially those in the tail, often protrude from

    the pancreatic surface and have a nodular bulge or polypoid

    appearance on gross examination.

    Histopathology

    Unlike the more common pancreatic ductal adenocarcinoma,

    PanNETs are mostly histologically stroma poor with morpho-

    logic features that are dependent on their level of differentia-

    tion. They are typically well circumscribed with variable

    amounts of capsule-like tissue. In WDNETs, cells are ar-

    ranged as sheets of monotonous epithelial cells, with anasto-

    mosing cords, nests, and trabeculae, coarse salt and pepper

    chromatin, and a fair amount of cytoplasm that may be eccen-

    tric and plasmacytoid (Fig. 3). Glandular structures, tubulo-

    acinar units, and pseudorosettes may also be seen, and thelatter may be so striking as to lead to misinterpretation as

    solid-pseudopapillary neoplasms (SPNs). Mitotic activity is

    usually scant. If mitoses are easily found, the possibility of an

    acinar cell neoplasm or other high-grade tumor should be

    excluded.

    The correlation of morphologic findings with hormone

    production (functionality status) is unreliable. Perhaps one

    exception is the close association of psammoma bodies and

    glandular pattern with immunohistochemical positivity for

    somatostatin.

    There are morphologic variants of PanNETs that are pecu-

    liar and diagnostically challenging. The lipid-rich variant of

    NETs is characterized by abundant foamy/microvesicular cy-

    toplasm, which creates a picture virtually indistinguishable

    from adrenocortical cells (Fig. 4). Some but not all of these

    are associated with VHL syndrome. Interestingly, the cyto-

    plasmic lipid frequently pushes and compresses the nucleus

    peripherally, thus obscuring the classical nuclear features of a

    NET, and may lead to their misdiagnosis as metastatic renal

    cell carcinoma or adrenal tumor on morphology. Some

    PanNETs have more uniform cytoplasm and may also show

    prominent rhabdoid [34] or signet ring features. WDNETs are

    also notorious for having oncocytic features characterized by

    abundant granular, eosinophilic cytoplasm, and a single prom-

    inent nucleolus, reminiscent of hepatocellular carcinoma

    (Fig.5). When the oncocytic variant metastasizes to the liver,

    it can be easily misdiagnosed as primary hepatocellular carci-

    noma especially if appropriate immunohistochemical stains

    are not performed. Unlike other morphologic variants, the

    oncocytic PanNET has been found to be more aggressive in

    some studies. Some tumors show a distinctsmall round cell

    appearance and have a very high nucleus-to-cytoplasm ratio.

    However, the nuclear molding, extensive necrosis, and high

    Fig. 2 Well-differentiated pancreatic neuroendocrine tumor is seen aris-

    ing in the pancreatic body as a solitary, well-circumscribed tan yellow

    mass. Note the absence of hemorrhage and necrosis

    70 Endocr Pathol (2014) 25:6579

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    7/15

    mitotic activity seen in the small-cell variant of PDNEC are

    not typically seen. A peliotic variant of PanNETs is also

    described, in which pools of blood and dilated blood vessels

    are conspicuous both grossly and microscopically. Marked

    cystic degeneration is seen in 5 % of tumors, and in such

    cases, there is typically a cuff of ordinary PanNET on the cystwall, making the diagnosis rather straightforward.

    PanNETs may show marked nuclear pleomorphism, so-

    called endocrine atypia, similar to that seen in other endocrine

    organs (Fig. 6). This can be so extensive that it can lead to

    misdiagnosis as adenocarcinoma [35]. Tumors may rarely show

    morphologic features mimicking those of paragangliomas [36].

    Typically, these paraganglioma-like tumors show more

    marked endocrine atypia (Fig. 7). The tumors presence in

    the pancreas, its coexpression of keratin and neuroendocrine

    markers, as well as the absence of basophilia and granularity

    typically seen in paraganglioma help to clinch the diagnosis. In

    some PanNETs with endocrine atypia, the degenerative

    symplastic cells (akin to those seen in symplastic leiomyomas

    or ancient schwannomas) can be so abundant and atypical that

    the case can be misdiagnosed as a high-grade malignancy.

    Some PanNETs contain abundant ductal elements, deemed

    by some as the ductulo-insular variant of PanNET. The

    ducts in such cases show obvious benign cytology. It is

    debatable whether these are entrapped native ductules prolif-erating secondary to the local effects of the tumor or represent

    an unusual transdifferentiation of tumor cells. The former

    appears to be the more plausible explanation. These tumors

    do not seem to behave differently than conventional PanNETs.

    However, if the ductular elements have severe atypia, the

    remote possibility of a mixed ductalneuroendocrine carcino-

    ma ought to be considered, but these are exceedingly uncom-

    mon tumors.

    Although tumors are usually stroma poor, they may exhibit

    marked sclerosis, especially those that secrete serotonin [37].

    Necrosis is not a typical feature of PanNETs and, when

    present, is usually focal, involves single cells, or may be

    comedo-like. If there is extensive tumor necrosis, the

    Fig. 3 a Well-differentiated

    pancreatic neuroendocrine tumor,

    grade 1. The tumor is stroma poor

    with an organoid arrangement of

    bland cells and intervening thin-

    walled blood vessels

    (hematoxylin & eosin stain, 400

    magnification).b Tumor cells are

    strongly positive for

    chromogranin A (400magnification)

    Fig. 4 Well-differentiated pancreatic neuroendocrine tumor, lipid-rich

    variant. Tumor cells are large with abundant, clear, vacuolated cytoplasm

    and oval nuclei lacking the salt and pepperchromatin typical of these

    tumors (hematoxylin & eosin stain, 400 magnification)

    Fig. 5 Well-differentiated pancreatic neuroendocrine tumor, oncocytic

    variant. This example shows tumor cells with abundant granular eosino-

    philic cytoplasm and nuclei with prominent central nucleoli (hematoxylin

    & eosin stain, 200 magnification)

    Endocr Pathol (2014) 25:6579 71

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    8/15

    possibility of an acinar cell neoplasm or high-grade neuroen-

    docrine carcinoma ought to be considered.

    Poorly Differentiated Neuroendocrine Carcinomas

    (PDNEC)

    In the WHO-2010, PDNECs are classified under the rubric of

    grade 3 PanNETs along with proliferatively active ordinary

    PanNETs. However, it is probably more accurate to regard

    poorly differentiated neuroendocrine carcinomas (PDNEC) as

    a separate entity or, at least, as the undifferentiated version of

    differentiated PanNETs. These are essentially defined as theircounterparts in the lung. They are extremely aggressive tumors

    that have often disseminated by the time of diagnosis. Median

    survival is typically less than 2 years. Platinum-based therapeu-

    tic agents have shown some promise in controlling their

    growth; however, their overall prognosis remains grim.

    These poorly differentiated, high-grade (G3) carcinomas are

    subdivided based on cell size into small- and large-cell variants.

    The large-cell variant is more common, often large, and charac-

    terized by large cells with prominent nucleoli and variable

    cytoplasm. Mitotic activity is brisk (often >40/10 HPFs) andnecrosis is often extensive. The small-cell variant shows small to

    intermediate cells with coarsesalt and pepperchromatin, high

    nucleus-to-cytoplasm ratio, inconspicuous nucleoli, prominent

    nuclear molding, and crush artifact (Fig.8). Mitotic figures are

    easily identifiable and there is extensive tumor necrosis.

    Better delineation of PDNECs from ordinary G3 PanNETs

    has been elucidated in some recent studies. Well-differentiated

    but highly proliferative PanNETs typically have a ki-67 index

    below 4050 % [10], whereas PDNECs typically show ki-67

    >55 %, are characterized by highly aggressive behavior, and

    require platinum-based therapy [11].

    It should be noted here that PDNECs are very uncommontumors, and most cases that are diagnosed as such prove to be

    acinar cell carcinomas once studied more carefully [38].

    Ancillary Studies

    Immunohistochemistry

    PanNETs are essentially defined by the production of cyto-

    plasmic neuroendo crin e granules, which are commonly

    highlighted by immunohistochemical markers chromogranin,

    synaptophysin, and CD56 (Fig.3). Ordinary well-differenti-

    ated PanNETs tend to show stronger more diffuse staining

    with neuroendocrine markers than PDNECs. Synaptophysin

    is highly sensitive but less specific. Chromogranin is the most

    specific neuroendocrine marker but is the least sensitive of the

    three. CD56 antibody (123c3) which is directed against neural

    Fig. 6 Well-differentiated pancreatic neuroendocrine tumor, pleomorphic

    variant. Tumor cells show marked nuclear enlargement,pleomorphism, and

    bizarre nuclei (hematoxylin & eosin stain, 400 magnification)

    Fig. 7 Well-differentiated pancreatic neuroendocrine tumor with

    paraganglioma-like features. Tumor cells show focal pleomorphism

    and a prominent nested pattern reminiscent of a paraganglioma (hema-

    toxylin & eosin stain, 100 magnification)

    Fig. 8 Small-cell neuroendocrine carcinoma of the pancreas. Tumor

    cells are arranged as tight groups and sheets of small cells with high

    nuclear to cytoplasmic ratio (hematoxylin & eosin stain, 200

    magnification)

    72 Endocr Pathol (2014) 25:6579

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    9/15

    cell adhesion molecules is the least specific neuroendocrine

    marker but is nonetheless useful in the differential diagnosis of

    these tumors. Neuroendocrine markers are also positive in

    large-cell carcinomas, but staining intensity and distribution

    are not as robust as in WDNETs.

    Additionally, NETs strongly express pancytokeratin as a

    marker of epithelial differentiation. Cytokeratin 19 (CK19),

    which is regarded as a marker of ductal lineage in the pancre-as, is sometimes expressed by PanNETs and has been found in

    some studies to be a marker of more aggressive behavior

    [1315]. Because of the latter, some authors advocate doing

    a CK19 stain on all PanNETs. CD117 (c-kit) has also been

    proposed as an independent adverse prognostic marker in

    PanNETs; however, its use is not routine [16].

    Focal acinar differentiation (with trypsin or chymotrypsin

    positivity) has also been reported in PanNETs, but it is usually

    present as isolated cells. Approximately 45 % of sporadic

    PanNETs show loss of expression of DAXX and ATRX

    immunostains, which correlates with mutations in the DAXX

    andATRXgenes [39]. Loss of Dpc4, a common phenomenon inductal adenocarcinoma of pancreas, is only rarely (if ever) seen

    in neuroendocrine tumors, which suggests that inactivation of

    theSMAD4/DPC4gene is a rare event in these tumors.

    CD99, a transmembrane glycoprotein encoded by theMIC-2

    gene, and progesterone receptor (PR) are also positive in a

    subset of PanNETs, in a manner similar to non-neoplastic islet

    cells. The nuclear transcription factor islet-1 (isl1) is frequently

    expressed in PanNETs [40, 41]. Pancreatic duodenal homeobox

    1 (PDX-1), a homeodomain transcription factor, plays a regu-

    latory role in early pancreatic development and is a marker of

    pancreatic PanNET (in both primary and metastatic tumors).

    Some studies have found isl1 and PDX-1 to be helpful in the

    differential diagnosis of pancreatic NETs from non-pancreatic

    WDNETs at metastatic sites such as liver, but their sensitivity

    and specificity have not been rigorously tested [42].

    Immunohistochemical analysis of PanNETs often lacks cor-

    relation with serologic status and, in some cases, may yield

    opposite results, presumably dueto therapid release and dispersal

    of the hormone product without cellular accumulation, but with

    retention of other hormones produced. Therefore, correlation

    between functional status and immunohistochemical hormone

    expression remains an imperfect science, and the use of immu-

    nohistochemistry to assess hormone production is unreliable.

    In a recent study, small- and large-cell PDNECs were shown

    to be genetically related but distinct from ordinary PanNETs

    [43]. P53 is positive in both small-cell (100 %) and large-cell

    NECs (90 %) but is negative in well-differentiated PanNETs.

    Rb protein is lost in 6090 % of PDNECs, and for tumors that

    retain Rb, there is usually concurrent loss of p16 staining,

    indicating that the two are mutually exclusive in NEC. Small-

    and large-cell NECs show abnormal immunolabelling with p53

    and Rb in 95 and 74 % of cases, respectively, which correspond

    to intragenic mutatedTP53 and retinoblastoma RB-1 genes

    [43]. Conversely, WDNETs retain Rb and p16. PDNECs retain

    DAXX and ATRX immunostains, unlike ordinary PanNETs in

    which these are frequently lost. PDX-1 is positive in up to 40 %

    of NECs, with large-cell tumors showing more positivity.

    PAX8 is positive in 50 % of NETs (including PDNECs and

    WDNETs).

    BCL2 protein, which is overexpressed in small-cell carci-

    noma of the lung, is also overexpressed in PDNECs (100 % ofsmall-cell and 50 % of large-cell tumors) but is negative in G1

    tumors and variably expressed in G2 WDNETs [43]. The

    platinum-based chemotherapeutic agents used to treat small-

    cell lung carcinoma exert their effects by inducing apoptosis

    via a BCL2 regulated pathway. This suggests that BCL2

    expression by PDNECs could potentially be exploited with

    BCL2 antagonists similar to those used in the lung.

    Electron Microscopy

    On ultrastructural analysis, NETs contain membrane-bound,

    dense core neurosecretory granules, which historically havebeen highly useful in their diagnosis. However, ultrastructural

    analysis is no longer accessible in most institutions and has

    been superseded by immunohistochemistry. Electron micros-

    copy may be useful in very poorly differentiated tumors or

    cases with an amphicrine phenotype.

    Molecular Studies

    PanNETs associated with MEN1 and VHL syndromes show

    mutations in theMEN1 andVHL genes. In MEN1, theMEN1

    (menin) gene located on chromosome 11q13 is mutated.

    Among sporadic PanNETs, somatic MEN1 mutation or loss

    of heterozygosity at the MEN1locus is seen in a significant

    number of cases [39, 4447]. MEN1 mutation is associated

    with a better prognosis. PDNECs however are infrequently

    associated with MEN1[1]. Deletion of theVHLgene occurs in

    up to 25 % of sporadic WDNETs.

    Among sporadic PanNETs, inactivating somatic mutations

    of the DAXX(death-domain associated protein) and ATRX

    (alpha thalassemia/mental retardation syndrome X-linked)

    genes have been identified in 45 % of cases. DAXX and

    ATRX proteins are crucial for telomere maintenance [48],

    and their associated genes are implicated in chromatin remod-

    eling [39]. When mutated, DAXXand ATRXresult in immu-

    nohistochemical loss of their corresponding proteins.

    Approximately 15 % of PanNETs show genetic mutations

    in the mammalian target of rapamycin(mTOR) cell signal-

    ing pathway proteins [39]. The mTOR pathway alteration

    represents a unique therapeutic target for drugs like everoli-

    mus which are directed specifically at this pathway [49].

    Chromosomal gains and losses have also been identified in

    PanNETs, and multiple chromosomal abnormalities are asso-

    ciated with a worse prognosis [50].

    Endocr Pathol (2014) 25:6579 73

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    10/15

    Differential Diagnosis

    Differential diagnoses of PanNETs include all primary pan-

    creatic neoplasms with diffuse, cellular sheet-like proliferation

    as well as metastatic tumors.

    Acinar cell carcinoma is one such primary pancreatic tumor

    that is derived from exocrine acinar cells and shows acinar

    differentiation as evidenced by positivity for trypsin, chymo-trypsin, lipase, other pancreatic enzymes, and BCL10. Tumor

    cells have abundant, granular, zymogen-rich cytoplasmic gran-

    ules, coarse chromatin, and prominent, sometimes cherry red,

    central nucleoli. A subset of acinar cell carcinomas is morpho-

    logically almost indistinguishable from PanNETs. In addition,

    most acinar cell carcinomas focally express neuroendocrine

    markers either as scattered individual cells or large zones or

    even as a very-well-formed distinct component (mixed carci-

    nomas). The presence of exuberant mitotic activity in the setting

    of seemingly bland monotonous epithelial cells is more in

    keeping with acinar cell carcinoma than a differentiated

    PanNET, in which mitotic figures are often few and far be-tween. Additionally, acinar cell carcinomas show prominent

    central nucleoli, which are typically less prominent in

    PanNETs. Most helpful though is the neuroendocrine chroma-

    tin pattern. Morphologic distinction is facilitated by immuno-

    histochemistry. Acinar cell carcinoma is negative or only focal-

    ly positive for neuroendocrine markers and shows positivity for

    pancreatic enzymes, while NETs show opposite results.

    Pancreatoblastomas are morphologically similar to acinar cell

    carcinoma and may also be confused with PanNETs. However,

    immunohistochemistry as well as the finding of classical

    squamoid morules should facilitate distinction.

    Morphologic distinction of mixed acinarneuroendocrine

    carcinoma, an acinar cell neoplasm that shows over 30 %

    neuroendocrine differentiation, can be especially challenging

    both on morphology as well as immunohistochemistry as

    tumor cells stain with both acinar and neuroendocrine

    markers. The key to distinguishing this variant from PanNET

    is the mixed neuroendocrine and acinar immunophenotype as

    well as the distribution of the neuroendocrine component,

    which should not be significant in these mixed tumors.

    Another key differencel is SPN. Both PanNET and SPN can

    show solid sheet-like pattern and nested growth (Fig.9). Typ-

    ically, the nesting is more vague in SPNs, and both patterns

    blend, rather imperceptibly, with each other, creating a more

    mesenchymal-like appearance. Striking overlapping of tumor

    nuclei is more prominent in SPN and the presence of large

    cytoplasmic vacuoles also favors SPN. On close examination,

    one should note the absence of the salt and pepperchromatin

    of neuroendocrine tumors and the presence of fine, open,

    powdery chromatin along with longitudinal nuclear grooves,

    which are distinctive findings in SPN. In addition, the presence

    of PAS-positive, diastase-resistant hyaline globules is support-

    ive of SPN, although similar globules can occasionally be seen

    in PanNETs [51]. Immunohistochemistry is extremely helpful

    in making the diagnosis as SPNs, in contrast to PanNETs, show

    only focal or weak positivity for keratin and chromogranin and

    strongly express nuclear-catenin. It should be kept in mind

    that SPNs may express neuroendocrine markers CD56 and

    synaptophysin, sometimes diffusely. This is a potential source

    of misinterpretation of immunohistochemistry unless one pays

    close attention to the distinguishing morphologic features. Itshould also be noted that some PanNETs exhibit striking

    pseudopapillary pattern mimicking SPNs.

    The clear cell or lipid-rich variant of WDNET can poten-

    tially be misdiagnosed as metastatic clear-cell renal cell carci-

    noma (RCC) involving the pancreas. Metastasis of clear-cell

    RCC to the pancreas is a known phenomenon and is one of the

    most frequent tumors to metastasize to the pancreas [52].

    Additionally, the lipid-rich variant of WDNET often fails to

    show the classical nuclear features of neuroendocrine neo-

    plasms, making diagnosis more challenging (Fig.4). As men-

    tioned earlier, the oncocytic variant of WDNET may resemble

    hepatocellular carcinoma (HCC) and, if it first presents as livermetastasis, can be misdiagnosed as HCC.

    Plasmacytomas are known to involve the pancreas, and for

    WDNETs showing marked plasmacytoid features, distinction

    from these hematopoietic neoplasms can be especially challeng-

    ing. It is important to note that in plasmacytoma, the chromatin

    distribution has a clock-facepattern with a distinctive perinuclear

    Hof. In WDNET, the chromatin distribution is different and the

    perinuclear Hof is absent. Plasmacytomas stain positively for

    CD138 and show clonal immunoglobulin, light chain restriction,

    with negativity for keratin and neuroendocrine markers.

    The large-cell variant of poorly differentiated (G3) neuro-

    endocrine carcinoma resembles poorly differentiated adeno-

    carcinoma and may be misdiagnosed as such unless suspected

    on morphologic assessment. Immunohistochemical expres-

    sion of neuroendocrine markers in addition to keratin supports

    the former. The small-cell variant of PDNEC may resemble

    metastatic small-cell carcinoma of lung. TTF1 stain is not

    helpful in distinction as small-cell carcinoma is TTF1-

    positive in a variety of extra-pulmonary sites. Clinical infor-

    mation and a history of previous carcinoma are especially

    important in the accurate diagnosis of such cases.

    The crushed and molded tumor cells of small-cell PDNEC

    may resemble a high-grade lymphoma. Morphologic distinc-

    tion is often impossible and immunohistochemistry is required

    for diagnosis.

    Small, round, blue cell tumors may involve the pancreas

    primarily or secondarily. These include desmoplastic small,

    round-cell tumor (DRCT) [53 ] as well as primitive

    neuroectodermal tumors (PNETs) [54]. The latter also ex-

    presses keratin in addition to CD99, both of which are also

    expressed by WDNETs. However, the salt and pepperchro-

    matin distribution is absent in DRCT and PNETs, which also

    show characteristic molecular alterations (EWS-WT1 gene

    74 Endocr Pathol (2014) 25:6579

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    11/15

    fusion and t(11;22), respectively) that help in distinction.

    Merkel cell carcinoma may rarely metastasize to the pancreas

    and in one report was present with a synchronous PanNET(insulinoma) [55]. Awareness of a previous history of this

    disease should prompt appropriate immunohistochemical

    studies to exclude this differential.

    Because of their brisk mitotic activity and morphologic

    resemblance to neuroendocrine cells, acinar cell carcinoma

    may be mistaken for large-cell neuroendocrine carcinoma. A

    panel of immunohistochemical stains including both neuro-

    endocrine and acinar markers (trypsin, chymotrypsin) can

    help to distinguish the two.

    Cytology

    Fine needle aspiration (FNA) has been used in the cytologic

    diagnosis of primary and metastatic PanNETs for many years. It

    is typically performed by endoscopic ultrasound-guided FNA,

    which is successful at identifying NETs in up to 90 % of cases.

    The cytologic features of these tumors are distinctive. For

    ordinary, well-differentiated (G1 and 2) NETs, tumor cells are

    classically singly dispersed or clustered as bland-appearing

    monotonous cells with round to oval nuclei and variable cyto-

    plasm, which may or may not have distinctive plasmacytoid

    features (Fig.10). In addition, multiple cytoplasmic vacuoles

    may be seen. On Papanicolaou stain, tumor cells show the

    classical salt-and-pepper chromatin distribution, which sup-

    ports the diagnosis (Fig. 10). Pseudorosettes are visible not only

    on cellblock but also on smears. Immunohistochemical analysis

    of cellblock material often demonstrates tumor cell positivity

    for keratin as well as neuroendocrine markers.

    In poorly differentiated neuroendocrine carcinoma of small-

    cell type, the cytologic features are similar to the lung counter-

    part. Tumor cells are often crushed and small to intermediate in

    size with irregular nuclear borders, high nucleus-to-cytoplasm

    ratio, crushed artifact, and nuclear molding. There is extensive

    single-cell and background necrosis in addition to brisk mitotic

    activity seen both on smears and cellblock. Tumor cells expresspancytokeratin as well as neuroendocrine markers, which help

    to support the diagnosis. For large-cell neuroendocrine carcino-

    ma, tumor cells are usually large with variable cytoplasm as

    well as prominent central nucleoli. On cytology, these tumors

    may resemble a poorly differentiated adenocarcinoma and may

    not show the classical salt-and-pepper chromatin of WDNETs

    or the nuclear molding of their small-cell counterpart. If a

    neuroendocrine primary is not suspected at the time of evalua-

    tion, this diagnosis may be easily missed.

    The grading of PanNETs has been performed on FNA by

    immunolabelling cell blocks with ki-67 [56, 57]. One might

    argue that cytologic samples are notoriously hypocellular and

    therefore lack the requisite minimum of 500 tumor cells

    Fig. 9 Solid pseudo-papillary neoplasm of the pancreas mimicking a

    pancreatic neuroendocrine tumor.a There is an organoid arrangement of

    (stroma-poor) tumor cells with morphology similar to a well-differentiat-

    ed pancreatic neuroendocrine tumor (hematoxylin & eosin stain, 100

    magnification). b Tumor cells shownuclear and cytoplasmic positivityfor

    -catenin immunostain (shown on the right) in contrast to the acinar cells

    on the left, which do not express nuclear-catenin (200 magnification)

    Fig. 10 Fine needle aspiration of a well-differentiated pancreatic neuro-

    endocrine tumor showing singly dispersed, bland tumor cells with

    plasmacytoid morphology, oval nuclei, and salt and pepperchromatin

    (Papanicolaou stain, 400 magnification)

    Endocr Pathol (2014) 25:6579 75

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    12/15

    required for calculation. Additionally, since NETs are fraught

    with proliferative heterogeneity, is the index/grade generated by

    an FNA (or even a core biopsy) reliable? Few have adequately

    addressed this question, but there are studies that support grad-

    ing of these tumors on biopsy-generated samples despite

    intratumoral heterogeneity [58]. In a recent cytologic study,

    grading results obtained by manual counting of ki-67-positive

    cells correlated positively with indices obtained by automatedimage cytometer, as well as corresponding resections [56].

    These findings suggest that the grading of PanNETs on cyto-

    logic material is applicable, and in situations in which such

    samples are the only ones available for evaluation (particularly

    in patients with metastatic disease), they may present the only

    opportunity for classification and treatment stratification.

    Biologic Behavior

    Ordinary PanNETs (G1 and G2) are considered low-grade

    malignancies (with the exception of tumorlets described ear-lier), with an overall 10-year survival of 6070 %. In the

    previous (WHO-2004) classification system as well as earlier

    studies, an attempt was made to recognize a benign behav-

    iorcategory. However, after more recent studies with longer

    clinical follow-up, it became clear that even the incidentally

    detected and resectable PanNETs that are 95 %.

    The prognosis of the G3 category is more complicated,

    largely due to the fact that this category encompasses at least

    two distinct entities: ordinary PanNETs with high proliferative

    rate and true PDNECs (Fig. 11). Preliminary results from

    Basturk et al. indicate that G3 cases with ordinary PanNET

    morphology are clearly more aggressive (median survival 32

    months, 5-year survival 22 %) than G2 NETs (median survival

    63 months, 5-year survival 61 %) but less aggressive than

    PDNEC (median survival 15 months, 5-year survival 17 %)

    [10]. Similar differences in survival have been shown by

    others [10, 11, 59, 60]. This was also confirmed by a study

    from Europe which showed that for all gastrointestinal G3

    NECs survival times were longer when the ki-67 index was

    2055 versus over 55 %, and the latter group was also moreresponsive to platinum-based therapy [11]. Accordingly, we

    currently regard G3 NETs as two distinct groups: (1) well

    moderately differentiated PanNET with proliferative index of

    G3, which typically has ki-67 >20 and 50 %.

    Treatment Options

    It is generally agreed among experts that, if possible, all

    functioning PanNETs should be resected [61, 62], with theexception of patients with MEN1 and ZollingerEllison syn-

    drome or those with small (

  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    13/15

    resection (cryosurgery) and transplantation, and hepatic artery

    embolization or radiofrequency ablation for non-surgical

    candidates.

    The role of chemotherapy (or targeted therapy) has been

    debatable. Since PanNETs are slow-growing tumors, they

    often do not respond to conventional cytotoxic chemothera-

    peutic agents. Recently, targeted therapies have become a

    serious alternative consideration. For tumors with alterationsof the mTOR pathway, the drug everolimus has shown prom-

    ise as a therapeutic agent [63] and is currently in phase IV

    clinical trials (http://www.nanets.net/research/current-clinical-

    trials). Cabozantinib, a tyrosine kinase inhibitor, and sunitinib,

    a vascular endothelial growth factor inhibitor, have also

    shown similar promise (http://www.nanets.net/research/

    current-clinical-trials).

    For patients with PDNECs, cisplatin and etoposide-based

    regimens are recommended and patients often show 4070 %

    (albeit brief) response rate [61]. The recent demonstration of

    BCL2 overexpression by PDNECs [43] suggests that BCL2

    antagonists may prove useful in their treatment in a mannersimilar to their current use in small cell carcinoma of lung,

    which also expresses BCL2.

    Summary of Key Elements to Include in a Pathology

    Report

    Resection of a Primary Pancreatic Neuroendocrine Tumor

    Diagnosis and differentiation: well-differentiated PanNET

    versus poorly differentiated PanNEC

    Tumor grade: based on mitoses/10 HPF and ki-67 prolif-

    eration index (%)

    Tumor size

    Lymphvascular invasion

    Perineural invasion

    Large vessel invasion

    Margin status (for resections)

    TNM stage (if a resection); specify method used (CAP/

    AJCC vs ENETS)

    Comment: Tumor grade, mitotic count/10 HPF, ki-67 index

    (%), necrosis, neuroendocrine marker expression, cytokeratin

    19 expression (optional)

    Resection or Biopsy of Metastatic Tumor (Presumed)

    from Pancreas

    Diagnosis and differentiation: well-differentiated PanNET

    versus poorly differentiated PanNEC

    Margin status: for resections

    Comment: tumor grade [based on mitoses/10 HPF, ki-

    67 proliferation index (%)], additional (optional) stains

    (PDX-1, isl1)

    References

    1. Klimstra D, Arnold R, Capella C, Hruban R, Kloppel G, Komminoth

    P, Solcia E, Rindi G (2010) Neuroendocrine neoplasms of the pan-

    creas. In: Bosman F, Carneiro F, Hruban R, Theise N (eds) WHO

    classification of tumours of the digestive system, 3rd edn. IARC,

    Lyon, pp 322-326

    2. Vagefi PA, Razo O, Deshpande V, McGrath DJ, Lauwers GY, Thayer

    SP, Warshaw AL, Fernandez-Del Castillo C (2007) Evolving patternsin the detection and outcomesof pancreatic neuroendocrine neoplasms:

    the Massachusetts General Hospital experience from 1977 to 2005.

    Arch Surg 142 (4):347-354. doi:10.1001/archsurg.142.4.347

    3. Anlauf M, Schlenger R, Perren A, Bauersfeld J, Koch CA, Dralle H,

    Raffel A, Knoefel WT, Weihe E, Ruszniewski P, Couvelard A,

    Komminoth P, Heitz PU, Kloppel G (2006) Microadenomatosis of

    the endocrine pancreas in patients with and without the multiple

    endocrine neoplasia type 1 syndrome. Am J Surg Pathol 30 (5):

    560-574. doi:10.1097/01.pas.0000194044.01104.25

    4. Lubensky IA, Pack S, Ault D, Vortmeyer AO, Libutti SK, Choyke

    PL, Walther MM, Linehan WM, Zhuang Z (1998) Multiple neuro-

    endocrine tumors of the pancreas in von Hippel-Lindau disease

    patients: histopathological and molecular genetic analysis. Am J

    Pathol 153 (1):223-231. doi:10.1016/S0002-9440(10)65563-0

    5. Hoang MP, Hruban RH, Albores-Saavedra J (2001) Clear cell endo-

    crine pancreatic tumor mimicking renal cell carcinoma: a distinctive

    neoplasm of von HippelLindau disease. Am J Surg Pathol 25 (5):

    602-609

    6. Fryer E, Serra S, Chetty R (2012) Lipid-rich (clear cell) neuroen-

    docrine tumors of the pancreas in MEN I patients. Endocr Pathol 23

    (4):243-246. doi:10.1007/s12022-012-9221-z

    7. Whipple AO, Frantz VK (1935) Adenoma of islet cells with hyper-

    insulinism: a review. Ann Surg 101 (6):1299-1335

    8. Shi C, Siegelman SS, Kawamoto S, Wolfgang CL, Schulick RD,

    Maitra A, Hruban RH (2010) Pancreatic duct stenosis secondary to

    small endocrine neoplasms: a manifestation of serotonin production?

    Radiology 257 (1):107-114. doi:10.1148/radiol.10100046

    9. Kenney B, Singh G, Salem RR, Paterno F, Robert ME, Jain D (2011)

    Pseudointraductal papillary mucinous neoplasia caused by microscopic

    periductal endocrine tumors of the pancreas: a report of 3 cases. Hum

    Pathol 42 (7):1034-1041. doi:10.1016/j.humpath.2010.09.018

    10. Basturk O, Yang Z, Tang LH, Hruban R, McCall C, Adsay V,

    Krasinskas A, Jang KT, Bellizzi A, Shi C, Klimstra DS (2013)

    Increased (> 20 %) Ki67 proliferation index in morphologically well

    differentiated pancreatic neuroendocrine tumors (PanNETs) corre-

    lates with decreased overall survival (abstract). Modern Pathology

    26:423A-423A

    11. Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P,

    Dueland S, Hofsli E, Guren MG, Ohrling K, Birkemeyer E, Thiis-

    Evensen E, BiaginiM, Gronbaek H, Soveri LM, Olsen IH, Federspiel

    B, Assmus J, Janson ET, Knigge U (2013) Predictive and prognostic

    factors for treatment and survival in 305 patients with advanced

    gastrointestinal neuroendocrine carcinoma (WHO G3): the

    NORDIC NEC study. Annals of Oncology: Official Journal of theEuropean Society for Medical Oncology/ESMO 24 (1):152-160.

    doi:10.1093/annonc/mds276

    12. Hochwald SN, Zee S, Conlon KC, Colleoni R, Louie O, Brennan

    MF, Klimstra DS (2002) Prognostic factors in pancreatic endocrine

    neoplasms: an analysis of 136 cases with a proposal for low-grade

    and intermediate-grade groups. J Clin Oncol 20 (11):2633-2642

    13. Deshpande V, Fernandez-del Castillo C, Muzikansky A, Deshpande

    A, Zukerberg L, Warshaw AL, Lauwers GY (2004) Cytokeratin 19 is

    a powerful predictor of survival in pancreatic endocrine tumors.Am J

    Surg Pathol 28 (9):1145-1153

    14. Jain R, Fischer S, Serra S, Chetty R (2010)The use of Cytokeratin 19

    (CK19) immunohistochemistry in lesions of the pancreas,

    Endocr Pathol (2014) 25:6579 77

    http://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trialshttp://dx.doi.org/10.1001/archsurg.142.4.347http://dx.doi.org/10.1097/01.pas.0000194044.01104.25http://dx.doi.org/10.1016/S0002-9440(10)65563-0http://dx.doi.org/10.1007/s12022-012-9221-zhttp://dx.doi.org/10.1148/radiol.10100046http://dx.doi.org/10.1016/j.humpath.2010.09.018http://dx.doi.org/10.1093/annonc/mds276http://dx.doi.org/10.1093/annonc/mds276http://dx.doi.org/10.1016/j.humpath.2010.09.018http://dx.doi.org/10.1148/radiol.10100046http://dx.doi.org/10.1007/s12022-012-9221-zhttp://dx.doi.org/10.1016/S0002-9440(10)65563-0http://dx.doi.org/10.1097/01.pas.0000194044.01104.25http://dx.doi.org/10.1001/archsurg.142.4.347http://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trialshttp://www.nanets.net/research/current-clinical-trials
  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    14/15

    gastrointestinal tract, and liver. Appl Immunohistochem Mol

    Morphol 18 (1):9-15. doi:10.1097/PAI.0b013e3181ad36ea

    15. Schmitt AM, Anlauf M, Rousson V, Schmid S, Kofler A, Riniker F,

    Bauersfeld J, Barghorn A, Probst-Hensch NM, Moch H, Heitz PU,

    Kloeppel G, Komminoth P, Perren A (2007) WHO 2004 criteria and

    CK19are reliable prognostic markersin pancreatic endocrine tumors.

    Am J Surg Pathol 31 (11):1677-1682. doi: 10.1097/PAS.

    0b013e31805f675d

    16. Zhang L, Smyrk TC, Oliveira AM, Lohse CM, Zhang S, Johnson

    MR, Lloyd RV (2009) KIT is an independent prognostic marker forpancreatic endocrine tumors: a finding derived from analysis of islet

    cell differentiation markers. Am J Surg Pathol 33 (10):1562-1569.

    doi:10.1097/PAS.0b013e3181ac675b

    17. Tang LH, Gonen M, Hedvat C, Modlin IM, Klimstra DS (2012)

    Objective quantification of the Ki67 proliferative index in neuroen-

    docrine tumors of the gastroenteropancreatic system: a comparison of

    digital image analysis with manual methods. Am J Surg Pathol 36

    (12):1761-1770. doi:10.1097/PAS.0b013e318263207c

    18. Bagci P, Ohike N, Dursun N, Jang K, Tajin T, Basturk O, Konh SY,

    Ducato L, Reid M, Adsay V (2012) Comparative analysis of different

    methodologies for ki67 in pancreatic neuroendocrine tumors (ab-

    stract). Modern Pathol 25;441A-441A

    19. AJCC (2010) Cancer staging manual, 7th edn edn. Springer, New York

    20. Sobin L, Gospodarowicz M, Wittekind C (2009) UICC: TNM clas-

    sification of malignant tumors, 7th edn. Wiley-Blackwell, Oxford

    21. CAP (2013) College of american pathologists cancer protocols and

    checklists. College of American Pathologists. http://www.cap.org/.

    Accessed 04/10/2013

    22. Rindi G, Kloppel G, Alhman H, Caplin M, Couvelard A, de Herder

    WW, Erikssson B, Falchetti A, Falconi M, Komminoth P, Korner M,

    Lopes JM, McNicol AM, NilssonO, Perren A, Scarpa A, ScoazecJY,

    Wiedenmann B (2006) TNM staging of foregut (neuro)endocrine

    tumors: a consensus proposal including a grading system. Virchows

    Arch 449 (4):395-401. doi:10.1007/s00428-006-0250-1

    23. Adsay NV, Bagci P, Tajiri T, Oliva I, Ohike N, Balci S, Gonzalez RS,

    Basturk O, Jang KT, Roa JC (2012) Pathologic staging of pancreatic,

    ampullary, biliary, and gallbladder cancers: pitfalls and practical

    limitations of the current AJCC/UICC TNM staging system and

    opportunities for improvement. Seminars in Diagnostic Pathology

    29 (3):127-141. doi:10.1053/j.semdp.2012.08.010

    24. Saka B, Oliva I, Bandyopadhyay S, Basturk O, Balci S, Maithel S,

    Kooby D, Sarmiento J, Staley C, El-Rayes B, Choi H, Knight J,

    Krasinskas A, Adsay V (2014) Will the pT1 and pT2 pancreas cancer

    please stand up? Peripancreatic soft tissue is involved in most

    pancreatic ductal adenocarcinomas (PDAC), negating its value as a

    staging parameter and necessitating a new staging scheme (abstract).

    Modern Pathology 27(Supplement), in press

    25. Saka B, Balci S, Bagci P, Maithel S, Kooby D, Sarmiento J, Staley C,

    El-Rayes B, Choi H, Knight J, Goodman M, Krasinskas A, Adsay V

    (2014) Proposal for a new and prognostically valuable tumor size

    based T-stage for pancreatic adenocarcinoma (abstract). Modern

    Pathology 27(Supplement), in press

    26. Rindi G, Falconi M, Klersy C, Albarello L, Boninsegna L, Buchler

    MW, Capella C, Caplin M, Couvelard A, Doglioni C, Delle Fave G,Fischer L, Fusai G, de Herder WW, Jann H, Komminoth P, de Krijger

    RR, La Rosa S, LuongTV, Pape U, Perren A, Ruszniewski P, Scarpa A,

    Schmitt A, Solcia E, Wiedenmann B (2012) TNM staging of neoplasms

    of the endocrine pancreas: results from a large internationalcohort study.

    J Natl Cancer Inst 104 (10):764-777. doi:10.1093/jnci/djs208

    27. Rindi G, de Herder WW, O'Toole D, Wiedenmann B (2006)

    Consensus guidelines for the management of patients with digestive

    neuroendocrine tumors:why such guidelines and how we wentabout

    it. Neuroendocrinology 84 (3):155-157. doi:10.1159/000098006

    28. Liszka L, Pajak J, Mrowiec S, Zielinska-Pajak E, Golka D, Lampe P

    (2011) Discrepancies between two alternative staging systems

    (European Neuroendocrine Tumor Society 2006 and American Joint

    Committee on Cancer/Union for International Cancer Control 2010) of

    neuroendocrine neoplasms of the pancreas. A study of 50 cases. Pathol

    Res Pract 207 (4):220-224. doi:10.1016/j.prp.2011.01.008

    29. Anlauf M, Perren A, Kloppel G (2007) Endocrine precursor lesions

    and microadenomas of the duodenum and pancreas with and without

    MEN1: criteria, molecular concepts and clinical significance.

    Pathobiology 74 (5):279-284. doi:10.1159/000105810

    30. Laidlaw GF (1938) Nesidioblastoma, the islet tumor of the pancreas.

    Am J Pathol 14 (2):125-134 125

    31. Vance JE, Stoll RW, Kitabchi AE, Williams RH, Wood FC, Jr. (1969)Nesidioblastosis in familial endocrine adenomatosis. JAMA 207 (9):

    1679-1682

    32. Heitz PU, Kasper M, Polak JM, Kloppel G (1979) Pathology of the

    endocrine pancreas. J Histochem Cytochem 27 (10):1401-1402

    33. Kloppel G, Anlauf M, Raffel A, Perren A, Knoefel WT (2008) Adult

    diffuse nesidioblastosis: genetically or environmentally induced?

    Hum Pathol 39 (1):3-8. doi:10.1016/j.humpath.2007.09.010

    34. Perez-Montiel MD, Frankel WL, Suster S (2003) Neuroendocrine

    carcinomas of the pancreas with 'Rhabdoid' features. Am J Surg

    Pathol 27 (5):642-649

    35. Zee SY, Hochwald SN, Conlon KC, Brennan MF, Klimstra DS

    (2005) Pleomorphic pancreatic endocrine neoplasms: a variant com-

    monly confused with adenocarcinoma. Am J Surg Pathol 29 (9):

    1194-1200

    36. Garbrecht N, Anlauf M, Schmitt A, Henopp T, Sipos B, Raffel A,

    Eisenberger CF, Knoefel WT, Pavel M, Fottner C, Musholt TJ, Rinke

    A, Arnold R, Berndt U, Plockinger U, Wiedenmann B, Moch H,

    Heitz PU, Komminoth P, Perren A, Kloppel G (2008) Somatostatin-

    producing neuroendocrine tumors of the duodenum and pancreas:

    incidence, types, biological behavior, association with inherited syn-

    dromes, and functional activity. Endocr Relat Cancer 15 (1):229-241.

    doi:10.1677/ERC-07-0157

    37. McCall CM, Shi C, Klein AP, Konukiewitz B, Edil BH, Ellison TA,

    Wolfgang CL, Schulick RD, Kloppel G, Hruban RH (2012)

    Serotonin expression in pancreatic neuroendocrine tumors correlates

    with a trabecular histologic pattern and large duct involvement. Hum

    Pathol 43 (8):1169-1176. doi:10.1016/j.humpath.2011.09.014

    38. Basturk O, Hruban R, Adsay V, Krasinskas A, Jang KT, Liu X,

    Vakiani E, Zhang L, Frankel W, Giordano T, Bellizzi A, Chen JH,

    Deshpande V, Yang Z, Klimstra DS (2013) High-grade neuroendo-

    crine carcinomas of the pancreas: a clinicopathologic analysis of 60

    cases (abstract). Modern Pathology 26:422A-423A

    39. Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A,

    Schulick RD, Tang LH, Wolfgang CL, Choti MA, Velculescu VE,

    Diaz LA, Jr., Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos

    N (2011) DAXX/ATRX, MEN1, and mTOR pathway genes are

    frequently altered in pancreatic neuroendocrine tumors. Science

    331 (6021):1199-1203. doi:10.1126/science.1200609

    40. Agaimy A, Erlenbach-Wunsch K, Konukiewitz B, Schmitt AM,

    Rieker RJ, Vieth M, Kiesewetter F, Hartmann A, Zamboni G,

    Perren A, Kloppel G (2013) ISL1 expression is not restricted to

    pancreatic well-differentiated neuroendocrine neoplasms, but is also

    commonly found in well and poorly differentiated neuroendocrine

    neoplasms of extrapancreatic origin. Mod Pathol 26 (7):995-1003.doi:10.1038/modpathol.2013.40

    41. Graham RP, Shrestha B, Caron BL, Smyrk TC, Grogg KL, Lloyd RV,

    Zhang L (2013) Islet-1 is a sensitive but not entirely specific marker

    for pancreatic neuroendocrine neoplasms and their metastases. Am J

    Surg Pathol 37 (3):399-405. doi:10.1097/PAS.0b013e31826f042c

    42. Chan ES, Alexander J, Swanson PE,Jain D, Yeh MM (2012) PDX-1,

    CDX-2, TTF-1, and CK7: a reliable immunohistochemical panel for

    pancreatic neuroendocrine neoplasms. Am J Surg Pathol 36 (5):737-

    743. doi:10.1097/PAS.0b013e31824aba59

    43. Yachida S, Vakiani E, White CM, Zhong Y, Saunders T, Morgan R,

    de Wilde RF, Maitra A, Hicks J, Demarzo AM, Shi C, Sharma R,

    Laheru D, Edil BH, Wolfgang CL, Schulick RD, Hruban RH, Tang

    78 Endocr Pathol (2014) 25:6579

    http://dx.doi.org/10.1097/PAI.0b013e3181ad36eahttp://dx.doi.org/10.1097/PAS.0b013e31805f675dhttp://dx.doi.org/10.1097/PAS.0b013e31805f675dhttp://dx.doi.org/10.1097/PAS.0b013e3181ac675bhttp://dx.doi.org/10.1097/PAS.0b013e318263207chttp://www.cap.org/http://dx.doi.org/10.1007/s00428-006-0250-1http://dx.doi.org/10.1007/s00428-006-0250-1http://dx.doi.org/10.1053/j.semdp.2012.08.010http://dx.doi.org/10.1093/jnci/djs208http://dx.doi.org/10.1159/000098006http://dx.doi.org/10.1016/j.prp.2011.01.008http://dx.doi.org/10.1159/000105810http://dx.doi.org/10.1016/j.humpath.2007.09.010http://dx.doi.org/10.1677/ERC-07-0157http://dx.doi.org/10.1016/j.humpath.2011.09.014http://dx.doi.org/10.1126/science.1200609http://dx.doi.org/10.1038/modpathol.2013.40http://dx.doi.org/10.1097/PAS.0b013e31826f042chttp://dx.doi.org/10.1097/PAS.0b013e31824aba59http://dx.doi.org/10.1097/PAS.0b013e31824aba59http://dx.doi.org/10.1097/PAS.0b013e31826f042chttp://dx.doi.org/10.1038/modpathol.2013.40http://dx.doi.org/10.1126/science.1200609http://dx.doi.org/10.1016/j.humpath.2011.09.014http://dx.doi.org/10.1677/ERC-07-0157http://dx.doi.org/10.1016/j.humpath.2007.09.010http://dx.doi.org/10.1159/000105810http://dx.doi.org/10.1016/j.prp.2011.01.008http://dx.doi.org/10.1159/000098006http://dx.doi.org/10.1093/jnci/djs208http://dx.doi.org/10.1053/j.semdp.2012.08.010http://dx.doi.org/10.1007/s00428-006-0250-1http://www.cap.org/http://dx.doi.org/10.1097/PAS.0b013e318263207chttp://dx.doi.org/10.1097/PAS.0b013e3181ac675bhttp://dx.doi.org/10.1097/PAS.0b013e31805f675dhttp://dx.doi.org/10.1097/PAS.0b013e31805f675dhttp://dx.doi.org/10.1097/PAI.0b013e3181ad36ea
  • 8/10/2019 10.1007-s12022-013-9295-2.pdf

    15/15

    LH, Klimstra DS, Iacobuzio-Donahue CA (2012) Small cell and

    large cell neuroendocrine carcinomas of the pancreas are genetically

    similar and distinct from well-differentiated pancreatic neuroendo-

    crine tumors. Am J Surg Pathol 36 (2):173-184. doi:10.1097/PAS.

    0b013e3182417d36

    44. Gortz B, Roth J, Krahenmann A, de Krijger RR, Muletta-Feurer

    S, Rutimann K, Saremaslani P, Speel EJ, Heitz PU, Komminoth

    P (1999) Mutations and allelic deletions of the MEN1 gene are

    associated with a subset of sporadic endocrine pancreatic and

    neuroendocrine tumors and not restricted to foregut neoplasms.Am J Pathol 154 (2):429-436. doi:10.1016/S0002-9440(10)

    65289-3

    45. Hessman O, Lindberg D, SkogseidB, Carling T, Hellman P, Rastad J,

    Akerstrom G, Westin G (1998) Mutation of the multiple endocrine

    neoplasia type 1 gene in nonfamilial, malignant tumors of the endo-

    crine pancreas. Cancer Res 58 (3):377-379

    46. Toliat MR, Berger W, Ropers HH, Neuhaus P, Wiedenmann B (1997)

    Mutations in the MEN I gene in sporadic neuroendocrine tumours of

    gastroenteropancreatic system. Lancet 350 (9086):1223. doi:10.

    1016/S0140-6736(05)63453-8

    47. Wang EH, Ebrahimi SA, Wu AY, Kashefi C, Passaro E, Jr., Sawicki

    MP (1998) Mutation of the MENIN gene in sporadic pancreatic

    endocrine tumors. Cancer Res 58 (19):4417-4420

    48. Heaphy CM, de Wilde RF, Jiao Y, Klein AP, Edil BH, Shi C,

    Bettegowda C, Rodriguez FJ, Eberhart CG, Hebbar S, Offerhaus

    GJ, McLendon R, Rasheed BA, He Y, Yan H, Bigner DD, Oba-

    Shinjo SM, Marie SK, Riggins GJ, Kinzler KW, Vogelstein B,

    Hruban RH, Maitra A, Papadopoulos N, Meeker AK (2011)

    Altered telomeres in tumors with ATRX and DAXX mutations.

    Science 333 (6041):425. doi:10.1126/science.1207313

    49. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E,

    Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P,

    Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011)

    Everolimus for advanced pancreatic neuroendocrine tumors. N Engl

    J Med 364 (6):514-523. doi:10.1056/NEJMoa1009290

    50. Wood L, Adsay N, Hruban R (2013) Molecular pathology of pan-

    creatic cancer. In: Cheng L, Eble J (eds) Molecular surgical pathol-

    ogy. Springer, New York, pp 17-42

    51. Meriden Z, Shi C, Edil BH, Ellison T, Wolfgang CL, Cornish TC,

    Schulick RD, Hruban RH (2011) Hyaline globules in neuroendocrine

    and solid-pseudopapillary neoplasms of the pancreas: a clue to the

    diagnosis. Am J Surg Pathol 35 (7):981-988. doi:10.1097/PAS.

    0b013e31821a9a14

    52. Adsay NV, Andea A, Basturk O, Kilinc N, Nassat H, Cheng JD

    (2004) Secondary tumors of the pancreas: ana analysis of a surgical

    and autopsy database and review of the literature. Virchows Archiv

    444(6):527-535

    53. Bismar TA, Basturk O, Gerald WL, Schwarz K, Adsay NV (2004)

    Desmoplastic small cell tumor in the pancreas. Am J Surg Pathol 28

    (6):808-812

    54. Movahedi-Lankarani S, Hruban RH, Westra WH, Klimstra DS

    (2002) Primitive neuroectodermal tumors of the pancreas: a report

    of seven cases of a rare neoplasm. Am J Surg Pathol 26 (8):1040-

    1047

    55. Bachmann J, Kleeff J, Bergmann F, Shrikhande SV, Hartschuh W,

    Buchler MW, Friess H (2005) Pancreatic metastasis of Merkel cell

    carcinoma and concomitant insulinoma: case report and literature

    review. World J Surg Oncol 3:58. doi:10.1186/1477-7819-3-58

    56. Fung AD, Cohen C, Kavuri S, Lawson D, Gao X, Reid MD (2013)

    Phosphohistone h3 and ki-67 labeling indices in cytologic specimensfrom well-differentiated neuroendocrine tumors of the gastrointesti-

    nal tract and pancreas: a comparative analysis using automated image

    cytometry. Acta Cytol 57 (5):501-508. doi:10.1159/000351475

    57. Larghi A, Capurso G, Carnuccio A, Ricci R, Alfieri S, Galasso D,

    Lugli F, Bianchi A, Panzuto F, De Marinis L, Falconi M, Delle Fave

    G, Doglietto GB, Costamagna G, Rindi G (2012) Ki-67 grading of

    nonfunctioning pancreatic neuroendocrine tumors on histologic sam-

    ples obtained by EUS-guided fine-needle tissue acquisition: a pro-

    spective study. Gastrointest Endosc 76 (3):570-577. doi:10.1016/j.

    gie.2012.04.477

    58. Yang Z, Tang LH, Klimstra DS (2011) Effect of tumor heterogeneity

    on the assessment of Ki67 labeling index in well-differentiated neu-

    roendocrine tumors metastatic to the liver: implications for prognos-

    tic stratification. Am J Surg Pathol 35 (6):853-860. doi:10.1097/PAS.

    0b013e31821a0696

    59. Velayoudom-Cephise FL, Duvillard P, Foucan L, Hadoux, J,

    Chougnet CN, Leboulleux, S, Malka D, Guigay J, Goere D,

    Debaere T, Caramella C, Schlumberger M, Planchard D, Elias D,

    Ducreux M, Scoazec JY, Baudin E (2013) Are G3 ENETS neuroen-

    docrine neoplasms heterogeneous? Endocr Relat Cancer 20 (5):649-

    657. doi:10.1530/ERC-13-0027

    60. McCall CM, Shi C, Cornish TC, Klimstra DS, Tang LH, Basturk O,

    Mun LJ, Ellison TA, Wolfgang CL, Choti MA, Schulick RD, Edil

    BH, Hruban RH (2013) Grading of well-differentiated pancreatic

    neuroendocrine tumors is improved by the inclusion of both Ki67

    proliferative index and mitotic rate. Am J Surg Pathol 37 (11):1671-

    1677. doi:10.1097/PAS.0000000000000089

    61. Kulke MH, Anthony LB, Bushnell DL, de Herder WW, Goldsmith

    SJ, Klimstra DS, Marx SJ, Pasieka JL, Pommier RF, Yao JC, Jensen

    RT (2010) NANETS treatment guidelines: well-differentiated neuro-

    endocrine tumors of the stomach and pancreas. Pancreas 39 (6):735-

    752. doi:10.1097/MPA.0b013e3181ebb168

    62. Metz DC, Jensen RT (2008) Gastrointestinal neuroendocrine tumors:

    pancreatic endocrine tumors. Gastroenterology 135 (5):1469-1492.

    doi:10.1053/j.gastro.2008.05.047

    63. Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, Jacobs C,

    Mares JE, Landgraf AN, Rashid A, Meric-Bernstam F (2008) Efficacy

    of RAD001 (everolimus) and octreotide LAR in advanced low- to

    intermediate-grade neuroendocrine tumors: results of a phaseII study.J

    Clin Oncol 26 (26):4311-4318. doi:10.1200/JCO.2008.16.7858

    Endocr Pathol (2014) 25:6579 79

    http://dx.doi.org/10.1097/PAS.0b013e3182417d36http://dx.doi.org/10.1097/PAS.0b013e3182417d36http://dx.doi.org/10.1016/S0002-9440(10)65289-3http://dx.doi.org/10.1016/S0002-9440(10)65289-3http://dx.doi.org/10.1016/S0140-6736(05)63453-8http://dx.doi.org/10.1016/S0140-6736(05)63453-8http://dx.doi.org/10.1126/science.1207313http://dx.doi.org/10.1056/NEJMoa1009290http://dx.doi.org/10.1097/PAS.0b013e31821a9a14http://dx.doi.org/10.1097/PAS.0b013e31821a9a14http://dx.doi.org/10.1186/1477-7819-3-58http://dx.doi.org/10.1159/000351475http://dx.doi.org/10.1016/j.gie.2012.04.477http://dx.doi.org/10.1016/j.gie.2012.04.477http://dx.doi.org/10.1097/PAS.0b013e31821a0696http://dx.doi.org/10.1097/PAS.0b013e31821a0696http://dx.doi.org/10.1530/ERC-13-0027http://dx.doi.org/10.1097/PAS.0000000000000089http://dx.doi.org/10.1097/MPA.0b013e3181ebb168http://dx.doi.org/10.1053/j.gastro.2008.05.047http://dx.doi.org/10.1200/JCO.2008.16.7858http://dx.doi.org/10.1200/JCO.2008.16.7858http://dx.doi.org/10.1053/j.gastro.2008.05.047http://dx.doi.org/10.1097/MPA.0b013e3181ebb168http://dx.doi.org/10.1097/PAS.0000000000000089http://dx.doi.org/10.1530/ERC-13-0027http://dx.doi.org/10.1097/PAS.0b013e31821a0696http://dx.doi.org/10.1097/PAS.0b013e31821a0696http://dx.doi.org/10.1016/j.gie.2012.04.477http://dx.doi.org/10.1016/j.gie.2012.04.477http://dx.doi.org/10.1159/000351475http://dx.doi.org/10.1186/1477-7819-3-58http://dx.doi.org/10.1097/PAS.0b013e31821a9a14http://dx.doi.org/10.1097/PAS.0b013e31821a9a14http://dx.doi.org/10.1056/NEJMoa1009290http://dx.doi.org/10.1126/science.1207313http://dx.doi.org/10.1016/S0140-6736(05)63453-8http://dx.doi.org/10.1016/S0140-6736(05)63453-8http://dx.doi.org/10.1016/S0002-9440(10)65289-3http://dx.doi.org/10.1016/S0002-9440(10)65289-3http://dx.doi.org/10.1097/PAS.0b013e3182417d36http://dx.doi.org/10.1097/PAS.0b013e3182417d36