Download as pdf or txt
Download as pdf or txt
You are on page 1of 30

REVIEW ARTICLE

Assigning Site of Origin in Metastatic Neuroendocrine


Neoplasms: A Clinically Significant Application
of Diagnostic Immunohistochemistry
Andrew M. Bellizzi, MD*w

WDNETs exhibit a range of “indolent” biologic


Abstract: The neuroendocrine epithelial neoplasms (NENs) include potentials—from benign to widely metastatic and even-
well-differentiated neuroendocrine tumors (WDNETs) and poorly tually fatal. Within each of these 2 groups of tumors, there
differentiated neuroendocrine carcinomas (PDNECs). Whereas is substantial morphologic overlap. The PDNECs are
PDNECs are highly lethal, with localized Merkel cell carcinoma
composed of “small,” intermediate, or large cells, often
somewhat of an exception, WDNETs exhibit a range of “indolent”
biologic potentials—from benign to widely metastatic and even- exhibit diffuse growth patterns, and are highly proliferative.
tually fatal. Within each of these 2 groups there is substantial The WDNETs, regardless of site of origin, are charac-
morphologic overlap. In the metastatic setting, the site of origin of terized by a series of growth patterns collectively referred to
a WDNET has significant prognostic and therapeutic implications. as “organoid” and frequently demonstrate finely granular
In the skin, Merkel cell carcinoma must be distinguished from chromatin. The range of biologies referenced above for the
spread of a visceral PDNEC. This review intends to prove the thesis WDNETs can to a large extent be predicted based on
that determining the site of origin of a NEN is clinically vital and anatomic site. In addition, the therapeutic armamentarium
that diagnostic immunohistochemistry is well suited to the task. It for metastatic WDNETs is rapidly expanding, and the
will begin by reviewing current World Health Organization ter-
efficacy of individual agents is related to site of origin.
minology for the NENs, as well as an embryologic and histologic
pattern–based classification. It will present population-based data When faced with a metastatic WDNET of unknown
on the relative frequency and biology of WDNETs arising at var- origin (generally to the liver), an all-too-familiar diagnostic
ious anatomic sites, including the frequency of metastases of scenario, pathologists have long been content to levy a
unknown primary, and comment on limitations of contemporary diagnosis to the effect of “metastatic neuroendocrine
imaging techniques, as a means of defining the scope of the prob- tumor” and to move on with their day. This practice is no
lem. It will go on to discuss the therapeutic significance of site of longer sufficient. Patients and their treating surgical oncolo-
origin. The heart of this review is a synthesis of data compiled from gists, endocrinologists, and medical oncologists deserve
>100 manuscripts on the expression of individual markers in some attempt at assigning site of origin, as well as comment
WDNETs and PDNECs, as regards site of origin. These include
on the degree of proliferation. These parameters are of signi-
proteins that are considered “key markers” and others that are
either useful “secondary markers,” potentially very useful markers ficant prognostic and therapeutic importance. In the skin,
that need to be further vetted, or ones that are widely applied MCC must be distinguished from spread of a visceral
despite a lack of efficacy. It will conclude with my approach to the PDNEC. Most pathologists are familiar with the application
metastatic NEN of unknown origin. of cytokeratin (CK) 20 and thyroid transcription factor-1
(TTF-1) immunohistochemistry (IHC) in this setting. Fewer
Key Words: well-differentiated neuroendocrine tumor, poorly dif- are familiar with the frequency of TTF-1 expression in
ferentiated neuroendocrine carcinoma, carcinoid tumor, Merkel PDNECs, regardless of site of origin. At least 2 other
cell carcinoma, carcinoma of unknown primary, immunohis- markers are useful in this setting, including one that directly
tochemistry, transcription factor, CDX2, TTF-1, cytokeratin 20 reflects the etiopathogenesis of MCC.
(Adv Anat Pathol 2013;20:285–314) Regarding the WDNETs, a number of markers,
including several transcription factors, have emerged that
are very useful in assigning site of origin. Most of these
antibodies are “workhorses” in diagnostic IHC (eg, TTF-1,
CDX2), others are emerging “powerhouses” (eg, polyclonal
T he neuroendocrine epithelial neoplasms (NENs),
including well-differentiated neuroendocrine tumors
(WDNETs) and poorly differentiated neuroendocrine car-
PAX8), and a few others are “classics” [eg, progesterone
receptor (PR), S-100], perhaps with a “new trick up their
cinomas (PDNECs), are characterized by the expression of sleeves.” Pathologists are, in general, unfamiliar with this
keratin intermediate filaments and the production of pep- application of IHC or will make assumptions about the
tide hormones and/or biogenic amines. Whereas PDNECs expression pattern of a marker based on data from ade-
are biologically aggressive and highly lethal, with localized nocarcinoma (eg, they assume that CDX2 will be expressed
Merkel cell carcinoma (MCC) somewhat of an exception, by tumors throughout the intestine).
This review intends to prove the thesis that determin-
ing the site of origin of a NEN is clinically vital and that
From the *Department of Pathology, University of Iowa Hospitals and
diagnostic IHC is well suited to the task. It will begin by
Clinics; and wDepartment of Pathology, University of Iowa Carver reviewing current World Health Organization (WHO) ter-
College of Medicine, Iowa City, IA. minology as well as an embryology-based and a histologic
The author has no funding or conflicts of interest to disclose. pattern–based classification that I find useful in organizing
Reprints: Andrew M. Bellizzi, MD, Department of Pathology, Uni-
versity of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa
my thoughts about these lesions. It will present (1) pop-
City, IA 52242 (e-mail: andrew-bellizzi@uiowa.edu). ulation-based data on the relative frequency and biology of
Copyright r 2013 by Lippincott Williams & Wilkins WDNETs arising at various anatomic sites, as a framework

Adv Anat Pathol  Volume 20, Number 5, September 2013 www.anatomicpathology.com | 285
Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

for, in the metastatic setting, conceptualizing the pretest Ki-67 IHC has been shown to highlight PIs for TC and AC
probabilities of spread from various sites; (2) the frequency analogous to those seen with G1 and G2 GEP NETs,
of metastases of unknown primary in several recent series; respectively (reviewed in Rekhtman3). As has been pointed
and (3) the limitations of current imaging techniques, as a out by several authors, Ki-67 IHC is often a useful diag-
means of defining the scope of the problem. It will go on to nostic adjunct in these tumors, particularly in crushed small
discuss the contemporary therapeutic significance of site of biopsies in which mistaking a carcinoid tumor for small cell
origin. The heart of this review is a synthesis of data com- lung carcinoma (SCLC) is a genuine pitfall of serious
piled from >100 manuscripts on the expression of indivi- clinical significance.3–5
dual markers in WDNETs and PDNECs, as regards site of The PDNECs include SCLC and large cell neuro-
origin. These will include proteins I consider “key markers” endocrine carcinoma (LCNEC). To qualify as poorly dif-
and others that are either useful “secondary markers,” ferentiated, a pulmonary NEN must demonstrate at least
potentially very useful markers that need to be further vetted, 11 mitotic figures per 2 mm2, although examples usually
or ones that are widely applied despite a lack of efficacy in exhibit much higher mitotic rates. [It is noteworthy that the
this setting. It will conclude with my approach to the meta- current mitotic “floor” for a pulmonary PDNEC (Z11/
static NEN of unknown origin. 2 mm2) is lower than that for a GEP NEC (> 20/10 HPF).]
Compared with SCLC, LCNEC is more apt to demonstrate
TUMOR CLASSIFICATIONS an organoid (rather than diffuse) architecture, is composed
of larger cells with more abundant cytoplasm, typically
World Health Organization Classification contains prominent nucleoli, and may or may not exhibit
The WHO Classification of Tumours of the Digestive “neuroendocrine chromatin.” Because of substantial mor-
System presents a nomenclature for gastroenteropancreatic phologic overlap with large cell (undifferentiated) carcinoma, a
(GEP) NENs that supplants the WHO 2000 Classification.1 diagnosis of LCNEC formally requires the immuno-
Morphologically well-differentiated NENs are referred histochemical demonstration of expression of at least 1 gen-
to simply as “neuroendocrine tumors” (NETs), whereas eral neuroendocrine marker.2
poorly differentiated examples are classified as “neuro- Although WDNETs and PDNECs predominate in the
endocrine carcinomas” (NECs). The “well-differentiated GEP system and lung, respectively, similar-appearing
endocrine carcinoma” of WHO 2000 no longer exists, with tumors occur in every organ system. These have generally
the current classification of a NEN based on its appearance, been classified by appropriating the terminology used for
rather than whether or not it has metastasized. In this GEP or pulmonary tumors. As most of the relevant WHO
classification, the term “NET” implies a well-differentiated Blue Books are still in their third editions, the descriptor
tumor (although WDNET will be used often throughout “carcinoid tumor” (a term emphatically stricken from the
this review for clarity). record for GEP NETs) still abounds. At some sites
As discussed above, NETs are biologically hetero- WDNETs are all classified as well-differentiated NECs,
genous, and it is well recognized that a group of tumors based on the premise that all WDNETs are potentially
characterized by tumor necrosis, increased mitotic activity, malignant.6 These semantics should not interfere with the
and increased tumor fraction participating in the cell cycle simple facts that the NENs, regardless of anatomic site, can
[ie, increased proliferation index (PI); determined by the be separated into well-differentiated, generally indolent
percentage of tumor cells expressing Ki-67 by IHC] carve tumors and poorly differentiated, aggressive ones, and that,
out a “middle path” between less-proliferative NETs and for the well-differentiated tumors, evidence of increased
PDNECs. In WHO 2010, this is captured in the grade of a cell turnover suggests the possibility of more aggressive
NET. By definition, grade (G)1 tumors demonstrate <2 behavior. A couple tumors deserve special mention here.
mitotic figures per 10 high-power fields (HPF) and/or a Ki- MCC is a PDNEC of the skin, and medullary thyroid
67 index of r2%, whereas G2 is assigned on the basis of a carcinoma is defined as a “malignant tumor of the thyroid
mitotic count of 2 to 20/10 HPF and/or a Ki-67 index of gland showing C-cell differentiation.”7
3% to 20%. The NECs can be described as being of “large
cell” or “small cell type” (the WHO 2010 term “NEC” Williams and Sandler Classification
implies a poorly differentiated tumor, although PDNEC Based on observations of the histology, histochemical
will be used for clarity). NECs are G3, with a mitotic count features, biochemical content, and biology of WDNETs,
>20/10 HPF and/or a Ki-67 index of >20%. Williams and Sandler,8 in an article published in The Lancet
The most recent WHO Classification of pulmonary in 1963, proposed that tumors be grouped into those that
NENs is contained in the 2004 WHO Classification of are foregut, midgut, and hindgut derived. The familiar
Tumours: Pathology and Genetics of Tumours of the Lung, “midgut carcinoid” is characterized by a nested archi-
Pleura, Thymus and Heart.2 In this system, the term “carcinoid tecture, often contains brightly eosinophilic cytoplasmic
tumor” persists. WDNETs of the lung are classified as “typical granules that are argentaffin positive (ie, they reduce silver
carcinoid” (TC) or “atypical carcinoid” (AC) on the basis of salts to metallic silver), has a high 5-hydroxytrypamine
mitotic rate and the presence or absence of tumor necrosis. (serotonin) content, and is frequently associated with car-
TCs possess well-differentiated histology, demonstrate <2 cinoid syndrome. Foregut-derived and hindgut-derived
mitotic figures per 2 mm2 (generally related as 10 HPF but tumors share several characteristics that distinguish them
actually closer to 8 HPF on many modern microscopes), lack from midgut ones. They have a tendency toward trabecular
necrosis, and measure Z0.5 cm. A diagnosis of AC is rendered architecture, are nonargentaffin, and contain little, if any,
when a morphologically similar tumor contains 2 to 10 mitotic serotonin. Compared to hindgut tumors, foregut ones are
figures per 2 mm2 and/or necrosis. more apt to be associated with carcinoid syndrome and
It is noteworthy that Ki-67 PI is not presently a often secrete 5-hydroxytryptophan.
component of the classification of pulmonary NENs Transcription factor expression is tightly regulated in
(although it may be part of the next one). Despite this fact, time and space and drives embryonic patterning. Thus, the

286 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

FIGURE 1. Architectural patterns in well-differentiated neuroendocrine tumors: Soga and Tazawa classification. A, Type A tumors are
nested; this histology is typical of ileal tumors. B, Type B tumors grow in ribbon-like fashion, as in this rectal tumor. C, Type C tumors are
composed of tubular, acinar, or rosette-like structures. This example was located in the ampulla, and it had been initially diagnosed on
biopsy as an adenocarcinoma. Note, also, the 2 psammomatous calcifications in the upper portion of this image. D, Type D tumors have
“diffuse” architecture. This appendiceal tumor, which was metastatic at presentation, had type A architecture elsewhere, and, in fact,
mixed patterns are often seen (hematoxylin and eosin, each 200).

Williams and Sandler classification provides an intuitively and mixed) and correlated them with site of origin (Figs. 1A–
appealing framework upon which to hang the observation D). Type A tumors are composed of solid nests (ie, insular).
that jejunal, ileal, and appendiceal WDNETs, all midgut Type B tumors are trabecular or “ribbon-like,” with the
derived, cluster together in terms of transcription factor cords of cells having a tendency to anastomose. Type C
expression and that pancreaticoduodenal tumors, foregut tumors contain tubular, acinar, or rosette-like structures (ie,
derived and arising from intimately developmentally related pseudoglandular). Type D tumors exhibit “lower or atypical
organs, would, by transcription factor expression patterns, be differentiation” (ie, “diffuse” or “undifferentiated”). Mixed
essentially indistinguishable. Similarly, the classification may tumors exhibit any combination of patterns A to D. In their
be useful to predict the expression profiles of cecal and other series, type A histology was typical of “midgut carcinoids,”
colonic tumors, for which little data exist. However, it is foregut-derived tumors tended to be type B, and hindgut
insufficient to explain all observations—for example, why the tumors were apt to be mixed.
transcription factor signature of pancreaticoduodenal tumors In the Armed Forces Institute of Pathology (AFIP)
seems to have more in common with hindgut-derived rectal series of 167 jejunoileal WDNETs, 93% were type A.10 The
tumors than with fellow foregut-derived gastric ones. association with type A histology is less strong in the
appendix, where Burke et al11 described, among 86 pure
Soga and Tazawa Classification WDNETs, 50% type A and 24% mixed tumors, as well as
Soga and Tazawa,9 in a manuscript published in Cancer 20% “tubular” carcinoids and 6% “clear cell” carcinoids.
in 1971 (based on their observations in 62 tumors), described While the type A, mixed, and clear cell tumors were
5 architectural patterns in “carcinoid tumors” (types A to D argentaffin positive and produced serotonin [characteristics

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 287


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

of enterochromaffin (EC)-cell WDNETs], the tubular car-


cinoids reacted with antibodies to glucagon. The latter TABLE 1. Site of Origin and Outcome in 35,825 Well-
Differentiated Neuroendocrine Tumors From the Surveillance,
tumor type may be related to rare appendiceal WDNETs Epidemiology, and End Results Registry (1973 to 2004)
composed of L cells (enteroglucagon cells), which express
glucagon-like peptides and peptide YY; these minute, Relative Distant Median
indolent tumors are distinctly trabecular (type B).12 Small, Frequency Metastasis Survival
incidentally discovered rectal WDNETs are also charac- Site of Origin (%)* (%) (mo)
teristically composed of L cells and demonstrate a type B Foregut
growth pattern.13 Type C histology is typical of somatos- Lung 27 28 193
tatin-producing D-cell tumors of the duodenum.14 Gar- Thymus 0.4 31 77
brecht et al15 found that, among 82 non-multiple endocrine Stomach 6.0 15 124
neoplasia type 1 (MEN1)-related duodenal NENs, 26% Duodenum 3.8 9 99
predominantly or exclusively expressed somatostatin; Pancreas 6.4 64 42
Liver 0.8 28 23
among these, 60% demonstrated at least a component of Midgut
type C growth. Also of note, 71% of these D-cell tumors Jejunum/ileum 13 30 88
occurred in the ampulla and 58% contained psammoma Appendix 3.0 12 NR
bodies. A type C component was noted in only 22% of Cecum 3.2 44 83
pancreatic D-cell tumors, which were much less common Colon 4.0 32 121
than in the duodenum (4%; 21/541). Outside of the jeju- Hindgut
noileum (type A) and rectum (type B), WDNETs are often Rectum 17 5 240
mixed.9,16 Other/unknown 15 NA NS
primary
Data from Yao et al.17
FREQUENCY, BIOLOGY, AND IMAGING *Data in this column are from the period between 2000 and 2004.
OF WELL-DIFFERENTIATED NEUROENDOCRINE NA indicates not applicable; NR, not reached; NS, not stated.
TUMORS
Population-based Data Regarding Relative
Frequency and Biology
In 2008, Yao et al17 published an analysis of the Notable among these studies is one from Kirshbom
epidemiology of WDNETs based on 35,825 tumors reported et al20 that reported on the clinical and biochemical char-
to the Surveillance, Epidemiology, and End Results (SEER) acteristics of 143 metastatic WDNETs of unknown primary
Program from 1973 to 2004. This study expanded on pre- (among >750 total WDNETs) seen by J.M. Feldman at
vious reports of US population–based cohorts published in Duke University Medical Center/Durham Veterans Affairs
1975 and 2003.18,19 Data from this study regarding relative Hospital over a nearly 30-year period. Patients with
tumor frequency, proportion presenting with distant meta- unknown primary tumors had high levels of urine 5-
stasis, and median overall survival—segregated by anatomic hydroxyindoleacetic acid (main serotonin metabolite) and
site—are summarized in Table 1. The 5 most frequent pri- urine, platelet, and serum serotonin levels on par with
mary sites included lung (27%), rectum (15%), jejunoileum those seen in patients with metastatic midgut WDNETs
(13%), pancreas (6.4%), and stomach (6%), accounting for
two thirds of all tumors. Among these, pancreatic tumors
most often presented with distant metastasis (64%) and were
associated with the shortest median survival (42 mo). By TABLE 2. Proportion of All WDNETs Accounted for by Metastases
comparison, jejunoileal tumors were metastatic half as often From Unknown Primaries in Recent Series
(30%) and associated with twice as long overall survival Frequency
(88 mo). Lung tumors were metastatic at a rate of 28% but of Total
were associated with long survival (193 mo), only exceeded Unknown Number
by rectal (240 mo) and appendiceal (median survival not Primaries of
reached) tumors. A significant weakness of the SEER data References (%) WDNETs Comments
set is that it relies on the reporting of tumors to the registry, Kirshbom 19 > 750 US single-center cohort
with the authors acknowledging that “many small, benign- et al20
appearing tumors likely are excluded.” This may explain why Yao et al17 13 35,825 US population–based
appendiceal tumors, for example, perceived as relatively cohort
common among WDNETs, represented only 3%. Pape et al21 10.5 399 German single-center
cohort; only
Frequency and Biology of Metastatic Tumors of gastroenteropancreatic
Lombard- 11.5 668 French multicenter
Unknown Origin Bohas cohort; only
Data from recent studies reporting the proportion of et al22 gastroenteropancreatic
metastatic tumors of unknown origin among all WDNETs Garcia- 9 907 Spanish population–based
are summarized in Table 2. The frequency has ranged from Carbonero cohort; only
9% (population-based cohort)23 to 19% (single-center, et al23 gastroenteropancreatic
referral-based cohort).20 The median of these studies is Catena et al24 11 750 Italian single-center
11%. In the SEER series, the frequency of tumors of cohort
“other/unknown” origin was exceeded only by that of WDNET indicates well-differentiated neuroendocrine tumor.
bronchopulmonary and rectal primaries.17

288 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

(and greater than those seen in patients with locoregional promise for superior spatial resolution, decreased patient
midgut tumors and much greater than patients with foregut exposure to radiation, and shorter scanning times.35,36
or hindgut tumors). The survival curves for unknown pri- A common response to the suggestion that patholo-
mary and metastatic midgut tumors were also quite similar, gists should participate in assigning the site of origin of a
with 10-year survivals of 22% and 28%, respectively, and metastatic WDNET of unknown primary is that we should
convergence of survival around 15 to 20 years. These “let radiology do it.” In the UCSF experience discussed
findings led the authors to suggest that most WDNETs of above, although CT successfully detected pancreatic
unknown origin may represent metastases from midgut WDNETs (pancNETs) in every case, the sensitivities of CT
primaries. and SRI (presumably OctreoScan) for detecting tubal gut
A retrospective analysis of the University of Cal- primaries were only 34.6% and 26.2%, respectively.25
ifornia, San Francisco (UCSF) experience with liver Savelli et al37 found that among 36 patients presenting with
metastases from WDNETs lends additional support to a metastatic NET of unknown primary (among 428 GEP
Kirshbom and colleagues’ hypothesis.25 Wang and colleagues NETs; 8.4%), OctreoScan successfully localized the pri-
identified 123 patients diagnosed between 1993 and 2008, mary in 39% (9 midgut, 3 pancreas, 1 cecum, 1 colon).
in whom hepatic metastasis was the initial disease pre- More recently, Prasad et al35 reported that DOTANOC
sentation in 64% (n = 79). Among 71 patients presenting PET/CT identified a primary site in 59% of 59 patients (14
with metastatic disease, adequate medical records, and in jejunoileum, 16 pancreas, 2 colorectum, 2 lung, 1 para-
whom a workup was undertaken, the primary tumor was ganglioma). Thus, traditional SRI may be useful in
successfully localized [making use of various combinations assigning the primary site in less than half of cases. Newer
of computed tomography (CT), somatostatin receptor PET tracers appear more sensitive, but they are not widely
imaging (SRI), positron emission tomography (PET), available. An additional consideration is cost. OctreoScan
magnetic resonance imaging, and upper and lower endos- is available at most US hospitals with a Nuclear Medicine
copy] in 56 (79%): pancreas 27, small intestine 17, colon 7, section, is charged at about $8000, and demonstrates a
lung 2, stomach 1, kidney 1, presacral 1. The remaining 15 sensitivity of 20 mm (the mean size of the occult small
patients underwent surgical exploration to detect an occult intestinal primaries detected by surgical exploration in
primary, which successfully identified 12 ileal and 1 jejunal Wang and colleagues’ series was 13.8 mm). In contrast,
tumor (13/15; 87%). DOTA scans, with a sensitivity of 4 mm, are, at present,
available at only 4 US centers, through the Food and Drug
Imaging of Well-Differentiated Neuroendocrine Administration’s Investigational New Drug program (T.M.
Tumors O’Dorisio, written personal communication). Diagnostic
In addition to conventional imaging modalities, IHC is nearly universally available and is much less
WDNETs can be visualized using nuclear medicine tech- expensive than either of these imaging techniques (or a
niques that take advantage of distinctive biochemical high-resolution CT for that matter); the Medicare Fee
properties of this class of tumors, including increased glu- Schedule for Current Procedural Terminology code 88342
cose metabolism, uptake of biogenic amines, and frequent is around $100.38
expression of somatostatin receptors (SSTRs). The first
of these is the basis of PET imaging with the radiotracer
18
F-fluoro-2-deoxy-D-glucose, which is more pertinent to THERAPEUTIC SIGNIFICANCE OF DETERMINING
the highly metabolically active PDNECs. The uptake of THE SITE OF ORIGIN IN WELL-DIFFERENTIATED
radioiodine-labeled metaiodobenzylguanidine (MIBG) by NEUROENDOCRINE TUMORS
the majority of WDNETs is the basis of the MIBG scan, as Key therapeutic determinants in WDNETs include
well as MIBG-radionuclide therapy. Although this modal- tumor site and size, anatomic extent of disease (local,
ity has been overtaken by SRI in NENs, MIBG-radio- regional, metastatic), presence of a functional syndrome,
nuclide therapy remains a mainstay of the treatment of and, in metastatic disease not amenable to surgery with
advanced pheochromocytoma/paraganglioma.26 curative intent, site of origin of the primary tumor. Ther-
Somatostatin is an inhibitory peptide hormone pro- apeutic options include endoscopic removal (for select
duced in regions of the brain and by D cells in the stomach, gastroduodenal and rectal tumors), surgery, ablative/
intestine, and pancreas. In the gastrointestinal (GI) tract, locoregional techniques (for liver metastases), antisecretory
somatostatin suppresses the release of several hormones treatment (for functioning tumors), and antiproliferative
including gastrin, cholecystokinin, vasoactive intestinal treatment. In patients with locoregional disease, the tumor
peptide, insulin, and glucagon. It mediates its effects is generally excised. Metastatic disease to the liver,
through SSTRs, which exist as 6 subtypes (SSTR1, 2A, 2B, depending on the pattern and extent of spread, is also
3 to 5). Frequent SSTR expression by NENs is the basis of potentially resectable.39–42 Adjuvant therapy following an
the OctreoScan, in which the somatostatin analogue R0 or R1 resection is not standard of care.43 Patients with
octreotide is coupled to 111In through the chelator dieth- diffuse hepatic involvement or extrahepatic disease are
ylene triamine pentaacetic acid. Of the SSTR subtypes, candidates for antiproliferative therapy. Even patients with
a positive scan has been shown to most closely relate to advanced metastatic disease may have their primary tumor
SSTR2A expression,27,28 which can be detected immuno- resected; this is particularly the case for jejunoileal tumors,
histochemically in 60% to 90% of NENs (see below).29–34 which have a tendency to cause obstruction, bleeding, and
Newer radiotracers couple octreotide or octreotate to ischemia (due to mesenteric vascular elastosis).
68
Ga via 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetra- Antiproliferative treatment options are very different
acetic acid (DOTA) (eg, DOTATOC, DOTANOC, in metastatic midgut and pancNETs (Table 3). Classes of
DOTATATE). These are used with PET and hybrid PET/ agents include those also used in antisecretory therapy,
CT imaging (as opposed to planar imaging or single systemic chemotherapeutics, small molecular inhibitors,
photon–emission CT with the 111In compound) and hold and peptide receptor–targeted radiotherapeutics. The

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 289


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

“SCLC” chemotherapy (ie, cisplatin or carboplatin and


TABLE 3. Options for Antiproliferative Medical Therapy in etoposide).57–59
Metastatic Neuroendocrine Tumors
Applicable in
Agent Mechanism Midgut Pancreatic KEY MARKERS IN DETERMINING THE SITE OF
ORIGIN OF A WELL-DIFFERENTIATED
Octreotide Somatostatin analogue + +
Interferon-a Immune activation + + NEUROENDOCRINE TUMOR
Streptozocin Alkylating agent +
Temozolomide Alkylating agent +
Caudal type Homeobox 2 (CDX2)
Everolimus mTOR inhibitor +/ + CDX2, one of 3 human homologues of the Drosophila
Sunitinib Tyrosine kinase inhibitor + gene Caudal, is a homeodomain-containing transcription
factor critical in gut development and patterning and in the
maintenance of its phenotype.60,61 It is normally expressed
by epithelial cells of the tubal gut distal to the stomach.62
somatostatin analogue octreotide is the mainstay of anti- CDX2 expression can also be found in pancreatic cen-
secretory therapy in functioning tumors (including patients troacinar and intercalated and intralobular duct cells and in
with carcinoid syndrome); interferon-a is a second-line scattered cells in the larger ducts.63 Downstream targets
agent due to its unfavorable side-effect profile. These agents include MUC2, sucrose-isomaltase, and lactase. Induction
also have weak antiproliferative activity and are utilized in of CDX2 expression is characteristic of gastric intestinal
advanced metastatic disease of midgut origin.44,45 Metastatic metaplasia64 and Barrett’s esophagus.65 Cdx2 homozygous–
pancNETs often respond to streptozocin-based systemic null mouse embryos fail to implant, as Cdx2 expression is
chemotherapy.46 Alternatively, temozolomide, another alky- required for the development of trophoectoderm. In a con-
lating agent, may be used.47 Intriguingly, in 1 retrospective ditional knockout (that overcomes the implantation block)
study, immunohistochemically detectable deficiency of the the tubal gut, which is lined by squamous instead of colum-
DNA-repair enzyme O(6)-methylguanine DNA methyl- nar epithelium, ends in a blind pouch at the cecum.66
transferase, observed in half of pancreatic and in no jeju- Diagnostic pathologists routinely use CDX2 as a
noileal tumors, predicted treatment response.48 The response marker of intestinal-type adenocarcinomas. Aside from
rate of midgut tumors to both of these agents has been shown colorectal and appendiceal tumors, in which expression is
to be poor.48,49 detected in most cases (> 90%), it is often, although var-
There is surging clinical interest in directed biological iably, reported in tumors arising elsewhere in the gut and in
therapy. Small-molecule inhibitors have been increasingly other tumors with intestinal-type differentiation (eg, muci-
evaluated in clinical trials. The mTOR inhibitor everolimus nous ovarian tumors).63,67–69 Two early IHC surveys of
was recently shown in a phase 3 randomized control trial to CDX2 expression reported positivity in a significant fraction
prolong progression-free survival in patients with advanced of intestinal WDNETs.63,68 Subsequently, CDX2 expression
pancNETs by 6.4 months (vs. best supportive care).50 has been shown to be highly sensitive and fairly specific for a
A similar trial in advanced midgut tumors fell just short of midgut origin (ie, jejunoileum or appendix). Nearly all of the
reaching statistical significance, but given the paucity of data regarding CDX2 expression in WDNETs are based on
therapeutic alternatives, everolimus may be considered.51 staining with the mouse monoclonal antibody (MoAb)
A phase 3 trial of the multiple receptor tyrosine kinase CDX2-88. A recent study demonstrated high sensitivity and
inhibitor sunitinib (inhibits platelet-derived growth factor superior specificity for a jejunoileal origin (appendiceal
receptors, vascular endothelial growth factor receptors, and tumors were not studied) with the rabbit MoAb EPR2764Y.70
KIT, among others) in advanced pancNETs was similarly Data from 14 studies assessing CDX2 expression
positive,52 while efficacy was not demonstrated in midgut in primary and metastatic WDNETs are summarized
tumors.53 Finally, although peptide receptor–targeted ther- in Table 4. Expression has been detected in 90% (177/197)
apy (eg, 90Y-DOTATOC) holds promise in both advanced of primary and 91% of metastatic (129/142) jejunoileal
midgut and pancreatic tumors, it is not widely available in the tumors (Figs. 2A, B). Similarly, CDX2 positivity has been
United States.54,55 noted in 93% (67/72) of appendiceal primaries; only a
handful of metastases have been tested (83%; 5/6). CDX2
expression has been detected in 31% (14/45) of duodenal,
THERAPEUTIC SIGNIFICANCE OF DETERMINING 14% (10/74) of gastric, and 29% (32/110) of rectal pri-
THE SITE OF ORIGIN IN POORLY maries. A large number of pancreatic tumors have been
DIFFERENTIATED NEUROENDOCRINE assessed, with CDX2 expression found in 16% (66/415) of
CARCINOMAS primary and 18% (15/84) of metastatic tumors. Only 3%
In the skin, the critical distinction is between MCC and (8/233) of pulmonary carcinoids have been positive. The
metastatic visceral PDNEC. Clinically localized MCC is specificity of CDX2 in this diagnostic context is probably
treated with excision and sentinel lymph node biopsy.56 A increased by taking quantity and intensity of staining into
positive sentinel lymph node biopsy prompts completion account. Whereas midgut tumors generally demonstrate
lymphadenectomy and/or radiation therapy. The primary diffuse, strong staining, expression in the foregut and
tumor bed may also be treated with radiation therapy, hindgut GEP tumors is often weak and patchy.
especially in patients considered at high risk for local recur- One additional anatomic site deserves special mention
rence (eg, positive margins, large tumor size, lymphovascular here. WDNETs of the ovary often arise in the background
space invasion). At other potentially metastatic sites (eg, of a teratoma and are divided into insular, trabecular,
liver), determination of the site of origin of a PDNEC strumal, and mucinous types. Rabban et al83 found that 5
is mainly of academic interest, as all metastatic tumors, of 7 (71%) ovarian WDNETs with an insular growth pat-
regardless of origin, are treated with platinum-based tern expressed CDX2, while none of 8 (0%) tumors with a

290 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


r
Adv Anat Pathol

TABLE 4. CDX2 Expression in Well-Differentiated Neuroendocrine Tumors


n (%)
References Antibody Lung Stomach Duodenum Pancreas Jejunum/Ileum Appendix Cecum Colon Rectum
Moskaluk et al63 CDX2-88 P: 2/7 (29) P: 0/3 (0) P: NE P: 4/14 (29) P: ND P: ND P: NE P: NE P: 7/9 (78)
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Barbareschi et al71 CDX2-88 P: 0/30 (0) P: 5/5 (100) P: 4/4 (100) P (F): 0/13 (0) P: 14/14 (100) P: 6/6 (100) P: NE P: 0/1 (0) P: 7/9 (78)
M: 0/1 (0) M: 1/1 (100) M: 1/1 (100) P (NF): 14/48 (29) M: 6/6 (100) M: NE M: NE M: NE M: NE
M: 1/6 (17)

2013 Lippincott Williams & Wilkins


Erickson et al72 CDX2-88 P: 3/10 (30) P: 0/10 (0) P: NE P: 1/8 (13) P: 53/53 (100) P: NE P: NE P: NE P: 3/6 (50)
M: 1/5 (20) M: NE P: NE M: 0/2 (0) M: 53/54 (98) M: NE M: NE M: NE M: NE
La Rosa et al73 CDX2-88 P: 0/7 (0) P: 0/6 (0) P: 3/7 (43) P: 5/24 (21) P: 12/12 (100) P: 5/5 (100) P: 2/2 (100) P: NE P: 0/14 (0)
M: NE M: 0/3 (0) M: 0/1 (0) M: NE M: 6/6 (100) M: NE M: 2/2 (100) M: NE M: 0/1 (1)
Saqi et al74 CDX2-88 P: 0/15 (0) P: 1/6 (17) P: 3/4 (75) P: 6/20 (30) P: 6/6 (100) P: 6/7 (86) P: NE P: NE P: 6/18 (33)
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Jaffee et al75 CDX2-88 P: 0/10 (0) P: 0/8 (0) P: NE P: NE P: 7/7 (100) P: 4/4 (100) P: NE P: NE P: 2/7 (29)
Volume 20, Number 5, September 2013

M: NE M: NE M: NE M: NE M: 4/4 (100) M: NE M: NE M: 1/1 (100) M: NE


Lin et al76 CDX2-88 P: 2/30 (7) P: 3/20 (15) P: NE P: 5/30 (17) P: 13/14 (93) P: 11/11 (100) P: NE P: 3/4 (75) P: 4/11 (36)
M: 0/2 (0) M: 0/1 (0) M: NE M: 0/1 (0) M: 5/5 (100) M: 2/2 (100) M: NE M: 2/2 (100) M: NE
Schmitt et al77 CDX2-88 P: 0/31 (0) P: 1/2 (50) P: 1/2 (50) P: 18/87 (21) P: 16/31 (52) P: 19/22 (86) P: NE P: 0/2 (0) P: 0/1 (0)
M: NE M: 1/1 (100) M: 2/2 (50) M: 1/18 (5) M: 11/18 (61) M: 3/4 (75) M: NE M: 1/2 (50) M: NE
Srivastava and Hornick78 CDX2-88 P: 0/20 (0) P: 0/5 (0) P: 0/5 (0) P: 3/31 (10) P: 21/21 (100) P: 11/11 (100) P: NE P: NE P: 0/12 (0)
M: NE M: NE M: NE M: 4/8 (50) M: 9/10 (90) M: NE M: NE M: NE M: NE
Hermann et al79 AMT28 P: NE P: NE P: 1/14 (7) P: 5/36 (16) P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Chan et al80 CDX2-88 P: 0/29 (0) P: NE P: NE P: 1/25 (4) P: 15/16 (94) P: 5/6 (83) P: 0/1 (0) P: 1/1 (100) P: 1/2 (50)
M: NE M: NE M: 0/2 (0) M: 2/5 (40) M: 2/2 (100) M: NE M: 2/2 (100) M: 1/1 (100) M: NE
Denby et al81 CDX2-88 P: NE P: NE P: NE P: NE P: NE P: NE P: NE P: NE P: NE
M: NE M: ND M: ND M: 6/16 (38) M: ND M: NE M: ND M: ND M: ND
Koo et al70 EPR2764Y P: 0/31 (0) P: 0/9 (0) P: NE P: 0/33 (0) P: 20/23 (87) P: NE P: NE P: NE P: 0/14 (0)
M: 0/5 (0) M: NE M: 0/1 (0) M: 1/28 (4) M: 33/37 (89) M: NE M: NE M: 1/1 (100) M: 0/1 (0)
Graham et al82 AMT28 P: NE P: NE P: 2/9 (22) P: 4/46 (9) P: NE P: NE P: NE P: 0/8 (0) P: 2/7 (29)
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Total P: 7/220 (3) P: 10/74 (14) P: 14/45 (31) P: 66/415 (16) P: 177/197 (90) P: 67/72 (93) P: 2/3 (67) P: 4/16 (25) P: 32/110 (29)
M: 1/13 (8) M: 2/6 (33) M: 3/7 (43) M: 15/84 (18) M: 129/142 (91) M: 5/6 (83) M: 4/4 (100) M: 6/7 (86) M: 0/2 (0)
F indicates functioning; M, metastatic; ND, not distinguishable; NE, not evaluated; NF, nonfunctioning; P, primary.

www.anatomicpathology.com |
291
Neuroendocrine Neoplasms: Assigning Site of Origin
Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

FIGURE 2. CDX2 expression in midgut well-differentiated neuroendocrine tumors (WDNET). A, Ileal tumor with predominantly type A
and focally type C histology. Note, also, the eosinophilic cytoplasmic granularity typical of enterochromaffin-cell tumors (hematoxylin
and eosin, 200). This patient had a concurrent ampullary adenocarcinoma. B, Two lymph nodes in the Whipple specimen contained
metastatic WDNET morphologically similar to that in the ileum. Diffuse, strong CDX2 expression suggests spread from the ileum
(immunoperoxidase, 200).

trabecular growth pattern did. Thus, in the ovary, CDX2 is TTF-1 expression is typical, while some studies have also
not helpful in distinguishing a primary insular WDNET demonstrated SPT24 positivity in occasional tumors in
from a midgut metastasis; interestingly, these 2 tumor types which TTF-1 expression is considered unusual.101,102 For
are essentially histologically identical. Also of note, tumors example, Compérat et al101 found SPT24 and 8G7G3/1
in this study did not express TTF-1. positivity in 84% versus 65% of 86 lung adenocarcinomas
and 10% versus 0% of 41 lung metastases of colorectal
Thyroid Transcription Factor-1 origin, respectively.
Initially identified based on its ability to bind to the In 21 studies using the 8G7G3/1 clone, TTF-1
thyroglobulin (TG) gene promoter,84 TTF-1 (also known as expression has been detected in 32% of 557 lung carcinoid
NK2 homeobox 1) is the best known of several human NKX tumors (Table 5 and Figs. 3A–C). Including 1 additional
homeodomain–containing transcription factors.85,86 It is study in which a small number of TCs were examined with
critical to lung, thyroid, and brain (diencephalon) develop- a rabbit polyclonal antibody, rates of positivity in individ-
ment, and it is expressed by type II pneumocytes, a ual studies have ranged from 0% to 95%, with a median of
subpopulation of nonciliated bronchiolar epithelial cells, 32%. ACs (mean 32%; median 40%) are possibly some-
follicular and parafollicular (C) cells, and neuroblasts in areas what more likely to be positive than TCs (mean 28%;
of the developing diencephalon including the hypothalamus median 21%). Spindle cell carcinoids frequently express
and neurohypophysis.87–89 In addition to TG, TTF-1 pro- TTF-1 (mean and median 69%). Thirteen of 23 (57%)
motes the expression of thyroperoxidase, thyrotropin recep- metastases have been positive. In contrast, TTF-1 expres-
tor, surfactant proteins, and Clara cell secretory protein. sion by a GEP WDNET is exceptional, reported to date in
A mouse knockout is athyroid, lung development distal to 0.6% of 708 primary tumors and 0% of 220 metastases. In
the lobar bronchi fails to take place, the ventral forebrain is 3 studies comparing SPT24 with 8G7G3/1, SPT24 was
abnormally formed, and the pituitary is missing.90 more frequently expressed in lung carcinoids than was
TTF-1 is most commonly used in diagnostic pathology 8G7G3/1. For example, Matoso et al102 found 61%
as a marker of lung adenocarcinoma and thyroid (SPT24) versus 17% (8G7G3/1) of 23 TCs to be positive. In
tumors.91–95 In the setting of a WDNET, TTF-1 has been 1 study also evaluating GEP tumors, this increased sensi-
found to be a variably sensitive, although incredibly spe- tivity was not at the expense of specificity, with La Rosa
cific, marker of lung origin. Although unlikely to present as et al112 not detecting SPT24 positivity in any (0%) of 103
a metastasis from an occult primary, expression is also tumors. The huge range of positivity (0% to 95%) cannot
detected in nearly all medullary thyroid carcinomas, which be explained by differing antibody clones (it preceded the
may additionally mark for calcitonin and carcinoembryonic first reports of SPT24 expression in this class of tumors),
antigen.89,96–99 Nearly all studies of TTF-1 immunoex- use or nonuse of heat-induced epitope retrieval (use of
pression in NENs have used the same mouse MoAb which is described in most of these studies), or differing
8G7G3/1 initially described by Holzinger et al100 in 1996, retrieval solutions (although a variety are described, citrate
which was produced against full-length recombinant rat has been most frequently used and has been associated with
protein. A few more recent studies have also used the both extremes of this range).
mouse MoAb SPT24, raised against a 123 amino acid
sequence from the N-terminal region of human TTF-1. The Paired Box Gene 8 (PAX8)
distinction is not inconsequential, with SPT24 generally The transcription factor PAX8, 1 of 9 paired box
demonstrating increased sensitivity in tumor types in which genes, is essential for thyroid development,115,116

292 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

participates with PAX2 in kidney development,117,118 and Insulin Gene Enhancer Binding Protein Isl-1
was found, through microarray-based gene expression (Islet 1)
profiling, to be highly expressed in surface ovarian carci- The homeodomain-containing transcription factor Islet 1
nomas.119 As such, detection of PAX8 expression has was initially isolated based on its ability to bind the insulin
emerged as an important diagnostic tool to identify carci- gene enhancer.140 In the adult rat, Islet 1 expression was
nomas of the thyroid, Müllerian system, and kidney.120–122 detected in a, b, and d cells of the islets of Langerhans, cells in
As application of PAX8 IHC became more prevalent, a few the anterior and intermediate lobes of the pituitary, paraf-
groups recognized strong expression in islets of Langerhans ollicular (C) cells, chromaffin cells and some neurons in the
and pancNETs and reported on the usefulness of PAX8 adrenal medulla, some sensory neurons in the dorsal root
expression to suggest the pancreatic origin of a WDNET ganglia, and subsets of motor neurons and central nervous
(Table 6 and Figs. 4A, B). system nuclei involved in autonomic and endocrine control.141
In 7 studies using a commercially available poly- In the developing embryo, Islet 1 is expressed in mesenchymal
clonal PAX8 antibody, expression has been detected in 145 cells of the dorsal pancreatic bud, as well as islet cells. An Islet
of 209 (69%) primary and 46 of 84 (55%) metastatic pan- 1 knockout mouse is characterized by failed exocrine devel-
cNETs. Although only a few duodenal (14 primaries) and opment from the dorsal but not the ventral pancreatic bud and
relatively few rectal (48 primaries) tumors have been stud- a complete absence of differentiated islet cells.142 Given its
ied, PAX8 expression (detected by the same antibody) importance in pancreatic, and in particular islet, development,
appears typical of these sites as well (79% and 58%, Islet 1 expression has been explored as a marker of pancNETs.
respectively). PAX8 positivity has not been noted in 85 As with polyclonal PAX8, in addition to expression in this
primary and 67 metastatic ileal tumors, and it has ranged group of tumors, Islet 1 positivity is also typical of duodenal
from 8% to 17% in tumors from the lung, stomach, and and rectal WDNETs (Table 6).
appendix. Also of note, neither PAX8, PAX2, CDX2, nor In a total of 5 studies investigating nearly 700
TTF-1 were found to be expressed in a series of 9 renal WDNETs, Islet 1 expression has been detected in 209 of 255
WDNETs.135 (82%) primary and 47 of 67 (70%) metastatic pancNETs.
Interestingly, up to 2010, the pancreatic developmental Similarly, 92% of 25 duodenal and 86% of 22 rectal pri-
biology literature was quiet on PAX8. Instead, PAX4 and maries have been positive. Although only a handful of
PAX6 had been shown critical to islet development, with metastases from the duodenum and rectum have been ana-
the PAX4 knockout mouse lacking mature b and d cells,136 lyzed, the results are similar. Islet 1 expression is uncommon
the PAX6 knockout lacking a-cells,137,138 and the combined to rare at other sites. Twelve percent (12%) of 94 pulmonary
PAX4/PAX6 knockout lacking any pancreatic neuro- and 17% of 42 appendiceal primaries have been positive,
endocrine cells.137 The most frequently used PAX8 anti- whereas the few metastases studied have not. Islet 1 expres-
body is polyclonal, raised against an N-terminal peptide sion has not been described in a gastric tumor, and although
that is highly conserved across the PAXs. It has been 3% of 69 primary ileal tumors have been positive, all 68
demonstrated that PAX8 mRNA is expressed at very low metastases studied to date have been negative. A recent study
levels in human islets (relative to PAX6 and PAX4), that a has highlighted frequent expression in medullary thyroid
MoAb directed against the divergent C-terminal region carcinoma (9/9; 100%).131 In the same study, Islet 1 expres-
does not react with non-neoplastic islets or pancNETs, and sion was also seen in most PDNECs including MCCs and
that the polyclonal PAX8 antibody cross-reacts with SCLCs, as well as neuroblastomas and paragangliomas/
PAX6.129 pheochromocytomas. (Of note, this lack of “anatomic site
The authors of the above-referenced study suggest that specificity” for a transcription factor in PDNECs has a
their results “cast doubts on the value of Pax8 as a pan- precedent; see directly below.) Compared with PAX8, Islet 1
creatic neuroendocrine tumor marker.”129 In a letter pub- boasts superior sensitivity for a pancreaticoduodenal or rectal
lished in the American Journal of Surgical Pathology, the primary. Its principal disadvantage is that it is a “unitasker”
same group warns of the potential for “flawed diag- (ie, it has no other defined diagnostic application at this time).
nostics.”139 Another group reports on the development of a
PAX8 MoAb, which they suggest “offers a significant
advantage by simplifying interpretation and enabling a KEY MARKERS IN DETERMINING THE SITE OF
more confident and accurate diagnosis.”130 In my opinion, ORIGIN OF A POORLY DIFFERENTIATED
abandoning the polyclonal PAX8 antibody in this context NEUROENDOCRINE CARCINOMA
is akin to throwing out the proverbial baby with the
bathwater. As a GI pathologist, I embrace polyclonal car- Thyroid Transcription Factor-1
cinoembryonic antigen IHC’s cross-reactivity with biliary Although most familiar as a marker of lung adeno-
glycoprotein to highlight the bile canaliculi diagnostic of carcinoma, the first reports of TTF-1 expression in human
hepatocellular differentiation. Am I to divert my eyes from tumors actually found more frequent expression in
granular cytoplasmic staining for TTF-1, suggesting the pulmonary small cell carcinomas than in adenocarcinomas.
possibility, again, of hepatocellular differentiation (another In their initial description of the 8G7G3/1 MoAb, Hol-
cross-reactivity, this time with carbamoyl phosphate synthe- zinger et al100 found 5 of 6 (83%) SCLCs and 7 of 11 (64%)
tase I)? Even if polyclonal PAX8 does cross-react with PAX6, adenocarcinomas to be positive, whereas Fabbro et al,103
the antibody remains an excellent marker of pancreatic (and using a rabbit polyclonal antibody, reported TTF-1
probably duodenal and rectal) WDNETs, better, in fact, than expression in 10 of 10 (100%) SCLCs and only 3 of 11
PAX6 itself [in Lorenzo et al’s129 study, while polyclonal (27%) adenocarcinomas. TTF-1 is also expressed, although
PAX8 highlighted 7 of 9 (78%) pancNETs, a PAX6 MoAb much less frequently, by pulmonary LCNECs. Although
was positive in only 3 (33%)]. It can be argued that this cross- the reported rates have been highly variable, TTF-1 is also
reactivity adds value above and beyond that seen with the expressed by at least a significant minority of extrapulmo-
recently developed PAX8 monoclonal. nary PDNECs. However, expression by a cutaneous

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 293


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

TABLE 5. TTF-1 Expression in Well-Differentiated Neuroendocrine Tumors


n (%)
References Antibody Pulmonary Tumors Gastroenteropancreatic Tumors
Fabbro et al103 Rabbit polyclonal TC 0/8 (0) NE
Folpe et al104 8G7G3/1 TC 18/51 (35) NE
AC 9/9 (100)
Agoff et al98 NR NE GE 0/49 (0)
P 0/15 (0)
Kaufmann and Dietel96 8G7G3/1 TC 4/9 (44) GEP 1/19 (5)
AC 2/3 (67)
Oliveira et al97 8G7G3/1 TC 16/17 (94) GE 0/26 (0)
AC 3/3 (100) P 0/10 (0)
MC 8/10 (80) Met GE 0/24 (0)
Cai et al105 8G7G3/1 C 11/16 (69) GE 1/46 (2)
MC 2/2 (100) P 0/12 (0)
Met GE 0/3 (0)
Met P 0/3 (0)
Sturm et al106 8G7G3/1 TC 0/27 (0) NE
AC 0/23 (0)
Zamecnik and Kodet107 8G7G3/1 TC 0/6 (0) NE
Chang et al108 8G7G3/1 TC 0/8 (0) NE
AC 0/3 (0)
Du et al109 8G7G3/1 TC 10/36 (28) GEP 0/17 (0)
AC 5/17 (29)
(SC) 12/16 (75)
Jerome Marson et al110 8G7G3/1 C 6/23 (26) NE
Pelosi et al4 8G7G3/1 TC 1/5 (25) NE
AC 0/2 (0)
Saqi et al74 8G7G3/1 TC 6/12 (50) GE 0/40 (0)
AC 2/3 (67) P 0/19 (0)
Hiroshima et al111 8G7G3/1 TC 0/7 (0) NE
AC 0/2 (0)
Lin et al76 8G7G3/1 TC 8/22 (36) GE 0/60 (0)
AC 5/8 (63) P 0/30 (0)
(SC) 8/13 (61) Met GE 0/10 (0)
MC 2/2 (100) Met P 0/1 (0)
Schmitt et al77 8G7G3/1 C 16/31 (52) GE 1/63 (2)
P 1/87 (1)
Met GE 0/18 (0)
Met P 0/28 (0)
Srivastava and Hornick78 8G7G3/1 C 7/20 (35) GE 0/54 (0)
P 0/31 (0)
Met GE 0/10 (0)
Met P 0/8 (0)
Matoso et al102 8G7G3/1 TC 4/23 (17) NE
SPT24 TC 14/23 (61)
La Rosa et al112 8G7G3/1 TC 4/41 (10) GE 0/60 (0)
AC 0/10 (0) P 0/26 (0)
(PC) 4/12 (33) Met GE 0/14 (0)
MC 0/4 (0) Met P 0/3 (0)
SPT24 TC 15/41 (37) GE 0/60 (0)
AC 2/10 (20) P 0/26 (0)
(PC) 9/12 (75) Met GE 0/14 (0)
MC 1/4 (25) Met P 0/3 (0)
Tsuta et al113 8G7G3/1 SC 9/13 (69) NE
Chan et al80 8G7G3/1 TC 11/24 (46) GE 0/26 (0)
AC 1/2 (50) P 0/25 (0)
Met GE 0/8 (0)
Met P 0/5 (0)
Denby et al81 8G7G3/1 NE Met GE 0/18 (0)
Met P 0/16 (0)
Koo et al70 8G7G3/1 C 16/31 (52) GE 0/46 (0)
MC 1/5 (20) P 0/33 (0)
Met GE 0/40 (0)
Met P 0/28 (0)
Masai et al114 8G7G3/1 C 2/50 (4) NE
SPT24 C 12/51 (24)
Range (median) (%) C 0-95 (32) GEP 0-5 (0)
TC 0-94 (21) GE 0-2 (0)

294 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

TABLE 5. (continued)
n (%)
References Antibody Pulmonary Tumors Gastroenteropancreatic Tumors
AC 0-100 (40) P 0-1 (0)
SC 61-75 (69) Met GE 0 (0)
PC 33 (33) Met P 0 (0)
MC 0-100 (80)
Totals C 176/565 (31) GEP 4/708 (0.6)
TC 82/296 (28) GE 2/410 (0.5)
AC 27/85 (32) P 1/262 (0.4)
SC 29/42 (69) Met GE 0/131(0)
PC 4/12 (33) Met P 0/89 (0)
MC 13/23 (57)
AC indicates atypical carcinoid; C, primary carcinoid (encompassing typical and atypical); GE, gastroenteric (tubal gut); GEP, gastroenteropancreatic;
MC, metastatic carcinoid; Met, metastatic; NE, not evaluated; P, pancreatic; PC, peripheral carcinoid; SC, spindle cell carcinoid; TC, typical carcinoid.

PDNEC (ie, MCC) is exceptional. As such, TTF-1 IHC is GI tract adenocarcinomas, and a human colon cancer cell
an essential component of the small panel of stains useful line, which they had designated protein IT.204 Several years
in distinguishing visceral from cutaneous tumors, with later, protein IT was proven to be a keratin and designated
expression effectively ruling out the diagnosis of MCC. CK20.205 CK20 is normally expressed by gastric foveolar
Data from 57 studies examining TTF-1 expression in epithelium, intestinal epithelium, urothelial umbrella cells,
PDNECs are summarized in Table 7. Expression has been and by Merkel cells in the skin. Diagnostically, it is gen-
detected in 698 of 846 (83%) SCLCs and 101 of 282 (36%) erally used as a marker of intestinal adenocarcinomas, and
pulmonary LCNECs; the median reported rates for these 2 it is expressed by a smaller number of gastroesophageal;
tumor types are 86% and 41%, respectively (Figs. 5A, B). pancreatobiliary; mucinous ovarian and lung; and urothe-
Two recent studies have compared the performance of the lial carcinomas.186,194 Germane to this discussion, CK20
8G7G3/1 and SPT24 clones, and, as with lung carcinoids, expression is seen in the vast majority of MCCs, while
positivity is more frequent with SPT24. For example, Masai expression is unusual in SCLC. CK20 is similarly uncom-
et al114 detected TTF-1 expression in 86% versus 68% of mon in extrapulmonary PDNECs, with the exception of
SCLCs and 47% versus 23% of pulmonary LCNECs. TTF- those arising in the major salivary glands, which have been
1 positivity has been reported in 200 of 550 (36%) extrap- described as “Merkel cell–like.”
ulmonary PDNECs; rates in individual studies have ranged CK20 expression has been noted in 416 of 472 (88%)
from 0% to 83%, with a median of 33%. TTF-1 expression MCCs, with rates ranging from 67% to 100% in 24 studies
has been reported in only 2 of 260 (0.8%) MCCs, each as a (median 91%) (Table 7). Expression is typically perinuclear/
single occurrence in 2 of 15 studies. dot-like but may also be diffuse (Fig. 6). CK20 positivity has
been reported in 18 of 373 (5%) SCLCs and 1 of 10 (10%)
Cytokeratin 20 pulmonary LCNECs. Eleven of these 18 were seen in 1 study
Using a combination of 2-dimensional gel electro- (11/33; 33%).145 Excluding this outlier study, the rate is 2%
phoresis, immunoblotting, and tryptic peptide mapping, in (7/340); the median frequency in 19 studies is 0%. CK20
1985 Moll and Franke203 identified a distinct cytoskeletal expression has been found in 19 of 30 (63%) major salivary
protein of molecular weight 46,000 in 9 of 9 (100%) MCCs gland tumors and 20 of 331 (6%) nonmajor salivary gland
and in none of 8 (0%) SCLCs. The protein colocalized with ones. In the largest series of major salivary gland small cell
one they had previously described in human intestinal cells, carcinomas (SCNECs), CK20 expression was noted in 11 of

FIGURE 3. TTF-1 expression in bronchopulmonary carcinoid tumors. A, Liver metastasis from unknown primary composed of epi-
thelioid and spindled cells. Mitotic activity is inconspicuous (hematoxylin and eosin, 400). B, Ki-67 immunohistochemistry (IHC)
reveals a proliferation index of 4.2% (immunoperoxidase, 400). C, 8G7G3/1 IHC demonstrates modest, although fairly diffuse, TTF-1
expression, which in a WDNET suggests a bronchopulmonary origin; spindle cell carcinoids are more apt to be positive (immuno-
peroxidase, 600). This case was seen in consultation; the referring pathologist’s impression was metastatic small cell carcinoma.

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 295


TABLE 6. PAX8, Islet 1, and PDX1 Expression in Well-Differentiated Neuroendocrine Tumors
Bellizzi

n (%)
Jejunum/
References Antibody (Vendor) Lung Stomach Duodenum Pancreas Ileum Appendix Cecum Colon Rectum
Polyclonal PAX8
Long et al123 Polyclonal (Proteintech) P: 0/20 (0) P: 1/5 (20) P: 5/5 (100)
P: 42/63 (67) P: 0/31 (0) P: 4/19 (21) P: NE P: NE P: 11/13 (85)
M: NE M: NE M: NE M: 9/18 (50) M: 0/16 (0) M: NE M: NE M: NE M: NE
Sangoi et al124 Polyclonal (Proteintech) P: 0/21 (0) P: 2/20 (10) P: 6/8 (75)
P: 49/66 (74) P: 0/31 (0) P: 1/11 (9) P: NE P: NE P: 5/17 (29)
M: 0/2 (0) M: NE M: 0/2 (0)
M: 20/31 (65) M: 0/11 (0) M: NE M: NE M: NE M: 0/1 (0)
Ozcan et al125 Polyclonal (Proteintech) P: NE P: ND P: ND P: 6/17 (35) P: ND P: ND P: ND P: ND P: ND
M: NE M: ND M: ND M: ND M: ND M: ND M: ND M: ND M: ND
Haynes126 Polyclonal (Proteintech) P: 0/12 (0) P: NE P: 0/1 (0)
P: 12/18 (67) P: NE P: NE P: NE P: NE P: 0/1 (0)
M: 0/1 (0) M: NE M: 0/1 (0)
M: 2/7 (29) M: 0/3 (0) M: NE M: NE M: NE M: NE

296 | www.anatomicpathology.com
Laury et al127 Polyclonal (Proteintech) P: 0/2 P: NE P: NE P: NE P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Tacha128 Polyclonal (Biocare Medical) P: 0/7 P: NE P: NE P: 0/3 (0) P: NE P: NE P: NE P: NE P: 1/3 (33)
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Lorenzo et al129 Polyclonal (Proteintech) P: NE P: NE P: NE P: 7/9 (78) P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
ab53490 (Abcam) P: 0/9 (0)
M: NE
Koo et al70 Polyclonal (Proteintech) P: 7/31 (23) P: 2/9 (22) P: NE P: 29/33 (88) P: 0/23 (0) P: NE P: NE P: NE P: 11/14 (79)
M: 0/5 (5) M: NE M: 0/1 (0) M: 15/28 (54) M: 0/37 (0) M: NE M: NE M: 0/1 (0) M: 0/1 (0)
Tacha et al130 BC12 (Biocare Medical) P: 0/16 (0) P: 0/3 (0) P: ND P: NE P: ND P: 0/2 (0) P: ND P: ND P: 0/2 (0)
M: NE M: NE M: NE P: NE M: NE M: NE M: NE M: NE M: NE
Total Polyclonal P: 7/93 (8) P: 5/34 (15) P: 11/14 (79) P: 145/209 (69) P: 0/85 (0) P: 5/30 (17) P: ND P: NE P: 28/48 (58)
M: 0/8 (0) M: ND M: 0/4 (0) M: 46/84 (55) M: 0/67 (0) M: ND M: ND M: 0/1 (0) M: 0/2 (0)
Monoclonal P: 0/16 (0) P: 0/3 (0) P: ND P: 0/9 (0) P: ND P: 0/2 (0) P: ND P: ND P: 0/2 (0)
M: NE M: NE M: ND M: NE M: NE M: NE M: NE M: NE M: NE
Islet 1
Schmitt et al77 40.3A4 P: 5/31 (16) P: 0/2 (0) P: 1/2 (50) P: 58/84 (69) P: 1/30 (3) P: 4/20 (20) P: NE P: 0/2 (0) P: 0/1 (0)
(DSHB) M: NE M: 0/1 M: 2/2 (100) M: 12/18 (67) M: 0/17 (0) M: 0/4 (0) M: NE M: 0/2 (0) M: NE
Adv Anat Pathol

Hermann et al79 C-terminal portion P: NE P: NE P: 14/14 (100) P: 32/36 (89) P: NE P: NE P: NE P: NE P: NE




(DSHB) M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE

r
Koo et al70 40.3A4 (DSHB) P: 2/31 (6) P: 0/9 (0) P: NE P: 27/33 (82) P: 0/23 (0) P: NE P: NE P: NE P: 12/14 (86)
M: 0/5 (0) M: NE M: 0/1 (0) M: 19/28 (68) M: 0/37 (0) M: NE M: NE M: 0/1 (0) M: 1/1 (100)
Graham et al82 1H9 (Abcam) P: 0/17 (0) P: 0/10 (0) P: 8/9 (89) P: 92/102 (90) P: 1/16 (6) P: 3/22 (14) P: NE P: 3/8 (38) P: 7/7 (100)
M: 0/2 (0) M: NE M: NE M: 16/21 (76) M: 0/14 (0) M: NE M: 0/1 (0) M: NE M: 2/2 (100)
Agaimy et al131 1H9 (Abcam) P: 4/15 (27) P: NE P: NE P: NE P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE
Total P: 11/94 (12) P: 0/21 (0) P: 23/25 (92) P: 209/255 (82) P: 2/69 (3) P: 7/42 (17) P: NE P: 3/10 (30) P: 19/22 (86)
M: 0/7 (0) M: 0/1 (0) M: 2/3 (67) M: 47/67 (70) M: 0/68 (0) M: 0/4 (0) M: 0/1 (0) M: 0/3 (0) M: 3/3 (100)
PDX1
Srivastava and Hornick78 Polyclonal (Santa Cruz) P: 0/20 (0) P: 3/5 (60) P: 4/5 (80) P/M: 11/39 (28)* P: 0/21 (0) P: 6/11 (55) P: NE P: NE P: 2/12 (17)
M: NE M: NE M: NE M: 0/10 (0) M: NE M: NE M: NE M: NE
Fendrichet al132w Polyclonal (Chemicon) P: NE P: NE P: 0/15 (0) P: 8/8 (100) P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: 0/3 (0) M: 5/5 (100) M: NE M: NE M: NE M: NE M: NE
Zhang et al133 Polyclonal (Abcam) P: NE P: NE P: NE P: 47/97 (48) P: NE P: NE P: NE P: NE P: NE
M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE M: NE

2013 Lippincott Williams & Wilkins


Volume 20, Number 5, September 2013
Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

15 (73%) tumors; TTF-1 expression was restricted to 3 of the

P: 2/14 (14)
4 CK20-negative cases.157
P: 0/2 (0)
M: NE

M: NE

M: NE
P: NE

P: NE
P: NE

OTHER MARKERS OF INTEREST IN WELL-


DIFFERENTIATED NEUROENDOCRINE TUMORS
M: 0/1 (0)

M: 0/1 (0)
P: 0/1 (0)

P: 0/1 (0)

Cytokeratins 7 and 20
M: NE
P: NE

P: NE
P: NE

A good deal of effort has been put forth to define the


expression of various keratin types in carcinomas from
diverse anatomic sites, with the goal of applying these data
M: 0/2 (0)

M: 0/2 (0)
P: 0/1 (0)

P: 0/1 (0)

to questions of tumor type (eg, adenocarcinoma vs. squ-


M: NE
P: NE

P: NE
P: NE

amous cell carcinoma) and, especially, site of origin. I have


seen pathologists attempt to extrapolate the findings from
what principally has been the study of non-neuroendocrine
P: 7/17 (41)
P: 1/6 (17)

carcinomas to WDNETs. In fact, there is actually a relative


M: NE

M: NE

M: NE

M: NE

paucity of data on the expression of individual keratins in


P: NE

P: NE

these tumors, with CK7 and CK20, given their wide clinical
application, having received the most attention (there is a
separate literature on the role of CK19 as a prognostic
marker in pancNETs). Although only up to a few hundred
M: 0/12 (0)
P: 0/16 (0)

P: 0/37 (0)
M: 0/2 (0)

tumors have been studied, the results have been fairly


M: NE

M: NE
P: NE

P: NE

consistent, such that conclusions can be drawn (Table 8).


CK7 is frequently, although not invariably, expressed
by carcinoid tumors of the lung. In 5 studies including a
total of 87 tumors, frequency of expression in individual
P: 114/210 (54)
M: 10/10 (100)
P: 18/36 (50)

P: 23/44 (52)

P: 18/25 (72)

studies has ranged from 4% to 66% (median 30%); overall,


M: 5/5 (100)

40% of pulmonary WDNETs have expressed CK7.


M: NE

M: NE

Expression is less frequent in both tubal gut (11/131; 8%)


and pancNETs (8/37; 22%), with the caveat that relatively
few pancreatic tumors have been examined. Although CK7
expression is more common in lung tumors, given more
P: 12/14 (86)

P: 16/34 (56)
M: 2/2 (100)

M: 2/5 (40)

frequent metastasis from ileal and pancreatic primaries,


CK7 positivity at a metastatic site probably would be most
M: NE

M: NE
P: NE

P: NE

often seen with spread from the pancreas. CK7 IHC is


neither sensitive nor specific enough to be useful in this
diagnostic application.
P: 3/5 (60)

CK20 is expressed by around a quarter of tubal gut


M: NE

M: NE

M: NE

M: NE
P: NE

P: NE

P: 3/29 (10) P: NE

WDNETs, perhaps slightly less often in pancNETs, and


almost never in carcinoid tumors of the lung. In 7 studies
M indicates metastatic; ND, not distinguishable; NE, not evaluated; P, primary.

including a total of 114 tubal gut tumors, frequency of


expression in individual studies has ranged from 15% to
P: 3/49 (6)

100% (median 24%); overall, expression has been noted in


M: NE

M: NE

M: NE

M: NE
P: NE

P: NE

24%. Of note, the 2 earliest studies describing CK20 pos-


itivity in 100% and 86% of 2 and 7 tumors, respectively,
reported expression in rare cells only.184,187 For pancNETs,
frequency of expression in 4 studies has ranged from 12%
to 33% (median 17%); in total, 9 of 49 (18%) tumors have
*Primary tumors and metastases were not distinguished.
Polyclonal (Santa Cruz)
267712 (R&D Systems)

267712 (R&D Systems)

been CK20 positive. Chan et al80 recently reported CK20


expression in 1 of 29 (3%) bronchopulmonary carcinoids; 56
tumors in 4 previous studies had all been negative. Although
wStudy restricted to (functioning) gastrinomas.

relatively insensitive as a marker of GEP WDNETs, CK20


expression argues against a pulmonary origin.
Neuroendocrine Secretory Protein 55
Neuroendocrine secretory protein 55 (NESP55) is 1 of 5
main transcripts of the GNAS locus. It is a member of the
granin family (like the more familiar chromogranin) and is
found in dense-core secretory granules in the adrenal medulla,
pituitary, and several parts of the brain.209 A few studies have
Hermann et al79

suggested that NESP55 expression in a WDNET is in keeping


Park et al134

Chan et al80

with a pancreatic origin. Jakobsen et al210 found NESP55


expression in 14 of 25 (56%) pancNETs and 0 of 15 (0%) ileal
Total

WDNETs. NESP55 expression was also detected in non-


neoplastic islets, most frequently in b cells. Also of note, 19 of
19 (100%) pheochromocytomas were positive. Similarly,

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 297


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

FIGURE 4. Polyclonal PAX8 expression in pancreatic neuroendocrine tumors. A, Interface of a primary pancreatic tumor with trabecular
architecture and non-neoplastic pancreas (hematoxylin and eosin, 200). B, Polyclonal PAX8 immunostain highlights both the tumor
and adjacent non-neoplastic islets of Langerhans (immunoperoxidase,  200).

Srivastava et al211 described NESP55 expression in 14 of 19 duodenum, PDX1 has been explored as a marker of
(74%) pancNETs, 10 of 10 (100%) pheochromocytomas, and WDNETs of these organs (Table 6).
0 of 11 (0%) ileal WDNETs. In addition, there was no staining There are very little data regarding PDX1 expression
in 4 gastric tumors and focal staining (< 5% of cells) in 1 of 4 outside of the pancreas—where it has been demonstrated in
rectal (25%) and 1 of 15 (7%) lung tumors. Srivastava and 114 of 220 (54%) primary tumors and 10 of 10 (100%)
Hornick subsequently extended this finding in a larger group metastases (data from 1 study are excluded from these
of tumors, reporting expression in 16 of 39 (41%) pancNETs totals because primaries and metastases were not reported
and no expression in gastric (0/5), duodenal (0/5), ileal (0/31), separately). Although in 3 studies PDX1 was expressed by
or appendiceal tumors (0/11). As in the prior study, expression most duodenal tumors [16/19 (84%) primaries and 2/2
was noted in a rare rectal (1 of 12; 8%) and lung (1 of 20; 5%) (100%) metastasis],78–80 in a single study that only looked
tumor.78 Recently, Denby et al81 reported NESP55 expression at functioning gastrinomas, PDX1 was not expressed at all
in 5 of 16 (31%) and 0 of 16 (0%) metastatic pancreatic and (15 primaries and 3 metastases).132 Hermann et al79 ach-
tubal gut WDNETs, respectively. ieved a very different result, with PDX1 expression noted in
Of note, all 4 of these studies made use of a polyclonal 10 of 10 (100%) gastrin-expressing duodenal tumors [5 of
rabbit antibody gifted by the same Austrian investigator. An these patients had Zollinger-Ellison syndrome (ZES)].
internet search identified several commercial sources of anti- PDX1 expression appears to be relatively uncommon in
NESP55, including at least 1 claiming to have been tested in lung primaries (3/49; 6%) and, importantly, has not been
IHC of formalin-fixed, paraffin-embedded tissue. Although observed in 37 jejunoileal primaries and 12 metastases
the specificity of these commercial antibodies for pancNETs (0%). There are almost no data from the stomach (3/5
would have to be verified, NESP55 represents an attractive primaries; 60%) and little data from the appendix (7/17
option in this setting. A drawback of this marker is its rela- primaries; 41%) or rectum (2/14 primaries; 14%). PDX1
tively limited clinical utility beyond this specific application. may have some role as a marker of pancreaticoduodenal
WDNETs. It may be less frequently positive in rectal
Pancreatic and Duodenal Homeobox 1 tumors than the other similar markers PAX8 and Islet 1,
Pancreatic and duodenal homeobox 1 (PDX1) is although additional data would be welcomed.
another homeodomain-containing transcription factor, ini-
tially described based on its ability to bind to and trans- Peptide Hormones
activate the insulin and somatostatin gene promoters.212,213 In Commercial antibodies to a wide variety of peptide
the developing embryo, PDX1 is expressed in the dorsal and hormones are available. A partial list of those normally
ventral pancreatic buds and the intervening duodenum; in the expressed in the GEP system is provided in Table 9. Aside
pancreas, expression is gradually extinguished in acinar and from gastrin, which is often used in gastric mucosal biopsies
ductal epithelium, such that, by around birth, PDX1 is rel- as part of the workup of a suspected case of autoimmune
atively islet-restricted.214 PDX1 knockout mice demonstrate atrophic gastritis, these are generally considered of limited
severe pancreatic hypoplasia, duodenal malformation, clinical utility. As such, most laboratories will only carry a
absence of Brunner glands, and decreased numbers of duo- few of these. Of note, functional syndromes are defined
denal enteroendocrine cells.214,215 In the adult, PDX1 based on the presence of characteristic symptoms, rather
expression has been noted in islets of Langerhans, cen- than predominant or even exclusive immunohistochemical
troacinar cells, rare intralobular and interlobular ductal cells, expression of a peptide hormone. Numerous studies have
and duodenal epithelium, and not in pancreatic acinar demonstrated that WDNETs, regardless of site of origin,
parenchyma.134 Given its importance in pancreaticoduodenal tend to express >1 hormone.14,216–222 Furthermore, meta-
development and its continued expression in islets and the stases may produce fewer or even additional peptides than

298 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

TABLE 7. TTF-1 and CK20 Expression in Poorly Differentiated TABLE 7. (continued)


Neuroendocrine Carcinomas
n (%)
n (%)
Extrapulmonary
Extrapulmonary References Pulmonary Merkel Cell [Site]
References Pulmonary Merkel Cell [Site]
Hiroshima SC: 13/23 (57) NE NE
TTF-1 et al111 LC: 4/17 (24)
Holzinger SC: 5/6 (83) NE NE Yun et al169 NE NE SC: 15/21 (71)
et al100 [esophagus]
Fabbro SC: 10/10 NE NE Carlson SC: 20/22 (91) NE SC: 0/7 (0)
et al103 (100) et al170 [ovary,
Di Loreto SC: 38/41 (93) NE NE hypercalcemic-
et al143 type]*
Folpe et al104 SC: 20/21 (95) NE NE SC: 1/2 (50)
LC: 6/8 (75) [ovary, lung-
Byrd-Gloster SC: 35/36 (97) 0/21 (0) NE type]
et al144 SC: 3/8 (38)
Hanly et al145 SC: 28/33 (85) 0/21 (0) NE [uterine cervix]
Agoff et al98 NE 0/6 (0) SC: 7/16 (44) El Demellawy NE NE SC: 0/10 (0)
Kaufmann SC: 30/37 (81) 0/16 (0) SC: 12/15 (80) et al171 [colon]
and Dietel96 LC: 2/4 (50) LC: 1/4 (25) Alijo Serrano NE NE SC: 11/44 (25)
Ordóñez146 SC: 27/28 (96) 0/18 (0) SC: 4/36 (11) et al172 LC: 2/2 (100)
Cheuk et al147 SC: 43/52 (83) 0/23 (0) SC: 21/50 (42) [bladder]
Shin et al148 NE NE SC: 2/10 (20) Lin et al76 SC: 28/30 (93) 0/3 (0) NE
[breast] Kargi et al173 SC: 28/28 NE NE
Leech et al149 SC: 10/10 0/11 (0) NE (100)
(100) Lu et al174 NE NE SC: 9/15 (60)
Sturm et al150 LC: 18/44 (41) NE NE [esophagus]
Chhieng SC: 2/5 (40) NE NE Wang and NE NE SC: 23/44 (52)
et al151 Epstein175 [prostate]
Zamecnik and SC: 8/9 (89) NE NE Ralston SC: 43/59 (73) 1/30 (3) NE
Kodet107 et al176
Sturm et al106 SC: 47/55 (85) NE NE McCluggage NE NE SC: 11/13 (85)
LC: 31/64 (48) et al177 LC: 4/8 (50)
Wu et al152 SC: 20/23 (87) NE NE [uterine cervix]
Myong153 SC: 14/16 (88) NE NE Li et al178 NE NE SC: 9/42 (21)
Yamamoto NE NE SC: 2/6 (33) [tubal gut]
154
et al [esophagus] Lewis et al179 NE 0/12 (0) HG: 5/14 (36)
Yang et al155 SC: 9/9 (100) 0/22 (0) NE [head and neck
Liu156 SC: 6/8 (75) NE NE mucosa]
Chang et al108 SC: 19/36 (53) NE NE La Rosa SC: 14/19 (73) 0/4 (0) HG: 5/41 (12)
157
Nagao et al NE NE SC: 3/15 (20) et al112 LC: 4/13 (31) [clone [clone 8G7G3/
[major salivary [clone 8G7G3/1] 1]
gland] 8G7G3/1]
Du et al109 LC: 6/16 (38) NE LC: 0/3 (0) SC: 16/19 (84) 0/4(0) HG: 8/41 (20)
Jerome SC: 24/30 (80) NE NE LC: 10/13 (77) [clone [clone SPT24]
Marson LC: 6/10 (60) [clone SPT24]
et al110 SPT24]
Soriano NE NE SC: 2/10 (20) Sidiropoulos SC: 23/30 (77) 1/40 (3) NE
et al158 [bladder] et al180
McCluggage NE NE SC: 0/15 [ovary, Fernández- NE NE SC: 1/6 (16)
et al159 hypercalcemic- Aceñero [bladder]
type]* et al181
Liu and SC: 6/6 (100) NE NE Thompson NE NE SC: 0/7 (0)
Farhood160 et al182 [bladder]
Zhang et al161 SC: 26/28 (93) NE NE Quinn et al183 NE NE SC: 9/25 (36)
Pelosi et al4 SC: 6/9 (67) NE NE LC: 0/2 (0)
Nassar et al162 NE NE HG: 0/9 (0) Masai et al114 SC: 46/68 (68) NE NE
[ampulla] LC: 24/106
Llombart NE 0/20 (0) NE (23) [clone
et al163 8G7G3/1]
Jones et al164 NE NE SC: 17/44 (39) SC: 59/69 (86)
[bladder] LC: 50/106
Wu et al165 SC: 15/16 (94) NE NE (47) [clone
Bobos et al166 SC: 11/13 (85) 0/13 (0) SPT24]
Yao et al167 NE NE SC: 15/18 (83) Denby et al81 SC: 12/17 (71) NE SC: 6/10 (60)
[prostate] [metastatic] [metastatic]
Kalhor SC: 12/13 (96) NE NE
et al168

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 299


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

TABLE 7. (continued) TABLE 7. (continued)


n (%) n (%)
Extrapulmonary Extrapulmonary
References Pulmonary Merkel Cell [Site] References Pulmonary Merkel Cell [Site]
Range SC: 40-100 0-3 (0) 0-83 (33) Ishida et al199 NE NE SC: 0/10 (0)
(median) (86) [uterine cervix]
(%) LC: 23-75 (41) Nassar et al162 NE NE HG: 3/8 (38)
Total SC: 698/846 2/260 (0.8) 200/550 (36) [ampulla]
(83) Llombart NE 15/20 (75) NE
LC: 101/282 et al163
(36) Jones et al164 NE NE SC: 0/44 (0)
Using 8G7G3/1 [bladder]
clone data Acebo et al200 NE 8/9 (89) NE
from Masai Bobos et al166 SC: 1/13 (8) 13/13 (100) NE
and La Yao et al167 NE NE SC: 2/18 (11)
Rosa [prostate]
CK20 El Demellawy NE NE SC: 2/10 (20)
Moll et al184 SC: 3/15 (20) 15/15 (100) NE et al171 [colon]
[rare cells] Alijo Serrano NE NE SC: 3/44 (7)
Brinkschmidt NE 17/18 (94) NE et al172 LC: 0/2 (0)
et al185 [bladder]
Wang et al186 SC: 0/11 (0) NE NE Saeb-Lima NE 7/7 (100) NE
Miettinen187 SC: 0/6 (0) 5/5 (100) NE et al201
Chan et al188 SC: 1/37 (3) 33/34 (97) SC: 4/52 (8) McCluggage NE NE SC: 3/13 (23)
[overall] et al177 LC: 1/8 (13)
including 3/5 (60) [uterine cervix]
[major salivary Lewis et al179 NE 11/12 (92) HG: 3/14 (21)
gland] [head and neck
Schmidt SC: 0/18 (0) 43/56 (77) NE mucosa]
et al189 Sidiropoulos SC: 0/30 (0) 35/40 (88) NE
Metz et al190 SC: 0/22 (0) 6/6 (100) NE et al180
Scott and SC: 0/6 (0) 9/10 (90) NE Fernández- NE NE SC: 0/6 (0)
Helm191 Aceñero LC: NE [bladder]
Byrd-Gloster SC: 1/36 (3) 16/21 (76) NE et al181
et al144 Chernock NE NE SC: 4/5 (80)
Hanly et al145 SC: 11/33 (33) 20/21 (95) NE et al202 LC: 1/2 (50)
Nicholson SC: 0/5 (0) 18/27 (67) NE [parotid]
et al192 Range 0-33 (0) 67-100 (91) 0-38 (3)
Ordóñez146 SC: 1/28 (4) 16/18 (89) SC: 0/36 (0) (median) [nonmajor
[overall] (%) salivary gland]
including 0/2 (0) 0-73 (60) [major
[salivary gland] salivary gland]
Shin et al193 NE NE SC: 0/9 (0)[breast] Total 19/383 (5) 416/472 (88) 20/331 (6)
Chu et al194 SC: 0/7 (0) 7/9 (78) NE [nonmajor
Jensen et al195 NE 23/26 (88) NE salivary gland]
Cheuk et al147 SC: 0/52 (0) 23/23 (100) SC: 2/50 (4) 19/30 (63) [major
[overall] salivary gland]
including 0/1 (0)
[parotid] *Data for small cell carcinoma of the ovary, hypercalcemic-type not
Leech et al149 SC: 0/10 (0) 10/11 (91) NE included in the total as this tumor type of uncertain histogenesis is non-
neuroendocrine.
Chhieng SC: 0/5 (0) NE NE HG indicates high-grade neuroendocrine carcinoma (ie, small cell and
et al151 large cell neuroendocrine carcinomas not distinguished); LC, large cell
Kende et al 196 NE NE SC: 0/2 (0) neuroendocrine carcinoma; NE, not evaluated; SC, small cell neuro-
LC: 0/2 (0) endocrine carcinoma.
M: 0/1 (0) [tubal
gut]
Mott et al197 NE 16/18 (89) NE
Yang et al155 SC: 0/9 (0) 19/22 (86) NE
Nagao et al157 NE NE SC: 11/15 (73)
matched primary tumors.219 I have used peptide hormone
[major salivary IHC in the workup of a WDNET in 3 contexts: (1) to
gland] distinguish pancreatic neuroendocrine (pseudo)hyperplasia,
Jerome SC: 0/30 (0) NE NE which should express some combination of insulin, gluca-
Marson LC: 1/10 (10) gon, somatostatin, and pancreatic polypeptide, from a
et al110 microadenoma (defined as a nonfunctioning pancNET
Soriano NE NE SC: 0/10 (0) <0.5 cm), which tends to express a single hormone; (2) in
et al158 [bladder] operated patients with multiple simultaneous tumors and a
Tanaka NE 31/31 (100) NE functional syndrome (MEN1-ZES), to help determine, for
et al198
example, which of the resected tumors (if any) is the source
of the syndrome; (3) in patients with metachronous tumors,

300 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

FIGURE 5. TTF-1 expression in small cell carcinoma (SCNEC). A, Metastatic SCNEC to the liver (hematoxylin and eosin,  400). B, The
tumor expresses TTF-1. Note, also, cross-reactivity with carbamoyl phosphate synthetase I, accounting for the granular, cytoplasmic
staining in hepatocytes at the lower right (immunoperoxidase, 400). TTF-1 expression is typical of small cell lung carcinoma (B85%)
but is seen often enough in extrapulmonary SCNEC (B35%) that it is not useful in assigning the site of origin; Merkel cell carcinoma,
however, is negative.

in which the first was known to express a dominant hor- of 96 (58%) pancNETs, 0 of 29 (0%) tubal gut WDNETs,
mone (Figs. 7A–D). and 1 of 15 (7%) lung TCs.224 Tempering enthusiasm
for the marker, expression was much more frequent in
Progesterone Receptor benign tumors (72%) than in malignant ones (31%; defined
Progesterone receptor IHC is a standard-of-care pre- in this study by the presence of local invasion or metastasis),
dictive marker in breast cancer and is widely used as part of a
panel to help determine the site of origin of a carcinoma of
unknown primary. In the setting of a WDNET, PR
expression has been proposed as indicative of pancreatic TABLE 8. CK7 and CK20 Expression in Well-Differentiated
origin. Doglioni et al223 reported PR expression in 40% to Neuroendocrine Tumors
75% of non-neoplastic islet cells, especially prevalent n (%)
in a cells, and 7 of 18 (39%) pancNETs. Estrogen receptor
(ER) expression was not detected. In a follow-up study References Lung Gastrointestinal Pancreatic
from the same group, PR expression was detected in 56 CK7
Broers et al206 3/10 (30) NE NE
Chu et al194 2/9 (22) 2/15 (13) NE
Cai et al105 10/16 5/46 (11) 6/12 (50)
(63)
Jerome Marson et al110 1/23 (4) NE NE
Alsaad et al207 NE 0/25 (0)* NE
Jiang et al208 NE 2/19 (11)* NE
Chan et al80 19/29 2/26 (8) 2/25 (8)
(66)
Range (median) 4-66 (30) 0-13 (11) 8-50 (29)
Total 35/87 11/131 (8) 8/37 (22)
(40)
CK20
Moll et al184 NE 2/2 (100)w 1/5 (20)
Miettinen187 0/8 (0) 6/7 (86)w 1/7 (14)
Chu et al194 0/9 (0) 1/15 (7) NE
Cai et al105 0/16 (0) 11/46 (24) 4/12 (33)
Jerome Marson et al110 0/23 (0) NE NE
Alsaad et al207 NE 4/25 (16)* NE
Jiang et al208 NE 6/19 (32)* NE
Chan et al80 1/29 (3) 4/26 (15) 3/25 (12)
Range (median) 0-1 (0) 15-100 (24) 12-33 (17)
Total 1/85 (1) 30/114 (24) 9/49 (18)
FIGURE 6. CK20 expression is seen in B90% of Merkel cell *Appendix only.
carcinomas; expression is typically perinuclear/dot-like (immuno- wOnly rare cells positive.
peroxidase, 600). In contrast, CK20 expression by visceral NE indicates not evaluated.
SCNECs (with the exception of parotid tumors) is unusual.

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 301


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

identified. Zhao et al225 found (focal) PR and ER expres-


TABLE 9. Commercially Available Antibodies to Peptide sion in 1 of 42 (2%) ovarian carcinoid tumors. Sica et al226
Hormones
studied PR and ER expression in pulmonary NENs and
Gastrin found more frequent PR expression than did the Milanese
Insulin group: 11 of 42 (26%) TCs and 2 of 7 (29%) ACs. In
Glucagon addition, ER expression was seen in over half the carci-
Somatostatin
noids. Given the wide availability of PR IHC and its
Pancreatic polypeptide
Gastric inhibitory peptide
potential application as a secondary marker in assigning the
Motilin site of origin in a WDNET, PR expression in these tumors
Secretin warrants additional investigation.
Cholecystokinin
Vasoactive intestinal polypeptide
Glicentin Prostatic Acid Phosphatase
Peptide YY
Prostatic acid phosphatase (PrAP) is often used along
with prostate-specific antigen to support a diagnosis of
and expression was detected in only 1 of 7 (14%) liver prostate cancer, either in the workup of a carcinoma of
metastases. unknown primary or in the setting of a high-grade tumor in
This finding has apparently received relatively little the prostate or bladder for which urothelial carcinoma also
attention, as only 2 other relevant published studies were enters the differential diagnosis. The frequent expression of

FIGURE 7. Peptide hormone immunohistochemistry. A, A 60-year-old woman with symptomatic hypoglycemia was found to have a
1.2 cm pancreatic tumor, which is composed of polygonal cells exhibiting a trabecular growth pattern and with prominent peliosis
(hematoxylin and eosin, 400). B, The tumor expresses insulin (immunoperoxidase,  400). C, Preoperative staging workup also
demonstrated a small lung tumor, which was subsequently resected. It is composed of somewhat fusiform cells, exhibiting trabecular to
pseudoglandular growth (hematoxylin and eosin, 400). D, This tumor does not express insulin (immunoperoxidase,  400). Given
subtly distinct morphologies and disparate insulin expression, these tumors were interpreted as separate primaries. Of note, insulinomas
are rarely metastatic at presentation.

302 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

FIGURE 8. S-100 immunohistochemistry. A, Metastasis from the appendiceal tumor depicted in Figure 1D. Note the nested growth and
eosinophilic cytoplasmic granularity typical of midgut well-differentiated neuroendocrine tumors (hematoxylin and eosin, 200).
B, S-100 immunohistochemistry performed on the metastatic focus highlights sustentacular cells (immunoperoxidase,  400). These
were also noted in the primary tumor. The presence of sustentacular cells has been suggested as useful in distinguishing appendiceal
from ileal tumors, although there are little to no data in the metastatic setting.

PrAP by rectal WDNETs (leading to an incorrect diagnosis recently, using the more specific marker SOX10, Tsuta
of prostate cancer in a small, crushed rectal biopsy) repre- et al237 detected sustentacular cells in 51 of 78 (65%)
sents a “classic pitfall” in diagnostic IHC. In the AFIP pulmonary carcinoids (more frequently in spindle cell/
series of 81 rectal (and 3 sigmoid) WDNETs, PrAP peripheral tumors) and 0 of 35 (0%) pulmonary PDNECs.
expression was detected in 82%.222 One important study casts doubt on the appendiceal spe-
PrAP expression is not restricted to rectal tumors, cificity of sustentacular cells in midgut WDNETs, with
although at most other sites it appears more infrequent. In Al-Khafaji et al238 finding S-100-positive sustentacular cells
the AFIP series of 51 jejunoileal WDNETs, PrAP was in 3 of the 11 (27%) jejunoileal WDNETs they studied.
expressed in 10 (20%).10 In an earlier study, the same group I recently encountered an appendiceal WDNET
reported expression in 15% of nonrectal WDNETs (includ- metastatic to the uterus, in which sustentacular cells were
ing gastric, small intestinal, and appendiceal tumors).227 demonstrable in both the primary and metastatic tumor
Azumi et al228 found even less frequent expression in 28 (Figs. 8A, B). That sustentacular cells were present in the
foregut-derived and midgut-derived tumors (focal, weak metastasis is intriguing, and the possibility that S-100 IHC
expression in 1 lung tumor). In contrast, frequent expression might have some usefulness in assigning the site of origin of
has been reported in insulinomas229,230 and strumal carci- a WDNET is considered, although, obviously an appendi-
noids,231–233 although this finding has not been well vetted in ceal tumor presenting as a metastatic WDNET of unknown
these tumor types. Again, given wide availability and appa- primary would be very unusual. S-100 could also be
rent differential expression across sites of origin, PrAP may potentially useful in distinguishing a tumor arising at the
have some use as a secondary marker in this context. base of the appendix from an ileocecal one.

S-100 Somatostatin Receptor 2A


The detection of S-100 protein expression, including as As discussed above, most NENs, even poorly differ-
a marker of melanocytic and Schwannian differentiation, is entiated examples, express SSTRs. Of these, the 2A subtype
a pillar of diagnostic IHC. S-100 is also expressed by sus- is the most relevant, as the somatostatin analogues used
tentacular cells (eg, in pheochromocytoma/paraganglioma), clinically (SRI, octreotide/lanreotide therapy, and peptide
relevant to this discussion. The presence of S-100-positive receptor radionuclide therapy) have the greatest affinity for
sustentacular cells has been touted as characteristic of an this receptor. SSTR2A IHC may have a role in selecting
appendiceal WDNET, especially in the differential diag- patients for somatostatin analogue therapy in instances in
nosis of an ileal tumor, which otherwise would stain sim- which SRI is unavailable or is negative (particularly in
ilarly for the markers already discussed. For example, small tumors).45 Until recently, application was limited by
Lundqvist and Wilander234 described this pattern of S-100 the fact that the best characterized antibody, which is
expression of 11 of 12 (92%) appendiceal tumors and not in polyclonal, was not commercially available. A recently
small intestinal (0/11) or cecal (0/10) tumors. Similarly, developed, commercially available MoAb (clone UMB-1)
Moyana and Satkunam235 found sustentacular cells in 8 of compares favorably to this antibody and gold standard
8 (100%) appendiceal WDNETs and 0 of 8 (0%) jejunoileal receptor autoradiography. Körner et al239 recently reported
ones. Sustentacular cells are also characteristic, although optimization of a staining protocol with this antibody.
less frequently seen, in bronchopulmonary carcinoids. Heat-induced epitope retrieval in a pressure cooker with
Barbareschi et al236 found them in 18 of 46 (39%) tumors; citrate buffer was shown to perform best. Quality and

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 303


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

intensity of staining are evaluated, with only membranous OTHER MARKERS OF INTEREST IN POORLY
staining considered truly positive. Staining is either faint DIFFERENTIATED NEUROENDOCRINE
(1 +), strong but with incomplete membrane staining (2 +), CARCINOMAS
or strong with complete membrane staining (3 +). The
group recommended that staining of >10% of tumor cells Merkel Cell Polyomavirus Large T Antigen
(of any intensity) be considered positive; staining of 1% to Drs Yuan Chang and Patrick Moore, having pre-
10% of tumors cells be considered inconclusive, with the viously discovered the human herpesvirus that causes
caveat that 1 + staining in this setting is likely negative; Kaposi sarcoma, hypothesized that, given its epidemiology
and that no tumor staining be considered negative. The (ie, associations with older age and immune suppression),
>10% cutoff was 86% sensitive, 95% specific, and had a MCC might also be attributable to an infectious agent.
95% positive predictive value for high receptor levels by Utilizing a technique they dubbed “digital transcriptome
autoradiography. Of 27 tumors with no staining, 96% had subtraction,” which entailed (1) sequencing tens of millions
no or low receptor levels by autoradiography. In the future, of bases from cDNA libraries of 4 MCCs, (2) aligning those
alternative somatostatin analogue–based therapies may be sequences with National Center for Biotechnology Infor-
developed that preferentially target other or a wider range mation reference sequences, and (3) looking for homology
of SSTRs. with infectious agents in the rare nonaligned sequences,
they discovered a single sequence with high homology to
known polyomaviruses.246 They subsequently identified the
Vesicular Monoamine Transporter 2 virus, which they termed Merkel cell polyomavirus
Vesicular monoamine transporters (VMATs) are (MCPyV) in 8 of 10 (80%) tumor samples. MCPyV is
integral membrane proteins involved in loading biogenic clonally integrated into tumor DNA and is associated with
amines into synaptic vesicles. Two human isoforms exist, characteristic mutations in the large T antigen (LT Ag),
with VMAT2 expression in the tubal gut relatively which abolish the virus’s ability to effectively replicate while
restricted to the histamine-producing, EC-like (ECL) cells maintaining LT Ag’s oncogenic properties.247 To date,
of gastric corpus mucosa.240 In contrast, only rare VMAT1- MCPyV has been detected by polymerase chain reaction by
expressing neuroendocrine cells are found in the stomach, B20 groups analyzing B600 MCCs, generally in the 80%
while the majority of neuroendocrine cells in the small and range initially reported by Chang and Moore. A notable
large intestine are VMAT1 positive. As such, VMAT2 has outlier was a cohort of Australian tumors, in which virus
been advocated as a marker of ECL-cell gastric WDNETs. was detected in only 5 of 21 (24%).248
VMAT2 IHC is considered a considerable improvement Chang and Moore’s group developed a MoAb to
over histamine IHC, which is practically limited by the low residues 116 to 129 of the LT Ag (clone CM2B4), which has
histamine content of ECL cells and its rapid dissolution in emerged as a fairly sensitive and highly specific marker of
liquid fixatives. MCC. In 8 studies including 313 pure MCCs, LT Ag
Rindi et al241 reported diffuse, strong VMAT2 expression has been detected in 60% (Table 10). As with the
expression in 16 of 16 (100%) ECL-cell tumors, the pres- polymerase chain reaction data, there is a single outlier,
ence of only rare VMAT2-positive cells in 12 of 21 (57%) again an Australian study in which only 19 of 89 (21%)
EC-cell (ie, serotonergic) tumors and 9 of 11 (82%) panc- tumors were positive.253 Excluding this study, 168 of 224
NETs, and absent expression (0%) in 5 gastrin, 4 soma- (75%) tumors have been CM2B4 positive. About 5% of
tostatin, and 3 enteroglucagon-expressing tumors. Like MCCs are associated with actinic keratosis, squamous cell
NESP55, VMAT2 expression is also characteristic of carcinoma in situ, a component of squamous cell carcinoma
pheochromocytoma [6 of 6 (100%) in this series; of note, or basal cell carcinoma, or have foci of intratumoral squ-
both VMAT 1 and 2 are highly expressed in the adrenal amous differentiation (so-called “combined MCC”). Inter-
medulla]. Similarly, Jakobsen and colleagues found estingly, these tumors have been uniformly negative for LT
VMAT2 expression in 11 of 12 (92%) gastric WDNETs Ag (0/43; 0%). These findings suggest that, although most
and less frequent and generally focal expression in appen- MCCs are MCPyV related, a subset including the combined
diceal (4/21; 19%), rectal (2/22; 9%), and pancreatic (8/24; tumors and some pure tumors are not, and are instead more
33%) tumors. VMAT2 expression was also detected in 34 likely ultraviolet radiation–associated (explaining enrich-
of 52 (65%) ileal tumors, and in 7 (13%) it was diffuse. ment in the Australian cohorts). Nearly 400 non-MCCs,
Anlauf and colleagues performed a focused investigation of including a large number of PDNECs (predominantly
non-neoplastic pancreas and pancNETs. In the non-neo- pulmonary but also CK20-positive tumors from the sali-
plastic pancreas, VMAT2 expression was restricted to b vary gland and uterine cervix) and hematolymphoid tumors
cells. It was also seen in 15 of 44 (34%) insulinomas and have been studied. Overall, 7 of 387 tumors (2%) have been
12 of 48 (25%) other functioning and nonfunctioning positive, including 6 of 23 (26%) SCLCs from 1 study.254
tumors.242 These studies serve to highlight high sensitivity Outside of this 1 study, CM2B4 IHC has been extremely
but lack of specificity for an ECL-cell primary (although specific, with only 1 other group reporting a single positive
diffuse, strong expression may be moderately specific). In case (of 74 SCNECs; 1.3%).253
fact, a recent study has suggested VMAT2 be regarded as a The CM2B4 clone is now commercially available from
“general neuroendocrine marker.”243 And unfortunately, Santa Cruz Biotechnology (Santa Cruz, CA). It would
although high-level expression is typical of the ECL-cell appear useful to support a diagnosis of MCC in the up to
tumors driven by hypergastrinemic states (ie, those arising 10% of CK20-negative tumors and to argue against a
in autoimmune atrophic gastritis and combined MEN1- diagnosis of MCC in other CK20-positive SCNECs (espe-
ZES), it is also usually encountered in sporadic gastric cially primary parotid tumors). CM2B4 negativity appears
WDNETs, as well.241,244,245 Given the evidence, VMAT2 too frequent for it to exclude a diagnosis of MCC (espe-
expression appears to be more scientifically interesting than cially in heavily sun-exposed populations). Rodig et al256
diagnostically useful. recently described a new MoAb (Ab3) raised against the

304 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

TABLE 10. Merkel Cell Polyomavirus Large T Antigen Expression (CM2B4 Immunohistochemistry) in Merkel Cell Carcinoma and Other
Tumors
n (%)
References Country Pure MCC Combined MCC Other Tumors [Type]
Shuda et al249 Spain 21/36 (58) NE 0/146 (0) [142 hematolymphoid neoplasms,
4 PDNEC lung]
Busam et al250 USA 37/49 (76) 0/9 (0) 0/26 (0) [PDNEC lung]
Reisinger et al251 USA 15/20 (75) NE 0/12 (0) [nonmelanoma skin cancers in
MCC patients]
McCluggage et al177 Northern Ireland NE NE 0/4 (0) [PDNEC uterine cervix]
Chernock et al202 USA NE NE 0/7 (0) [PDNEC parotid]
Kuwamoto et al252 Japan 21/22 (95) 0/4 (0) NE
Paik et al253 Australia 19/89 (21) 0/15 (0) 1/74 (1.3)* [SCNEC, predominantly lung]
Jung et al254 South Korea 11/13 (85) NE 6/23 (26) [SCNEC lung]
Ly et al255 Canada 17/27 (63) 0/15 (0) 0/87 (0) [including 15 each of melanoma,
SCC, PDNEC lung, GI NET]
Rodig et al256 USA 46/57 (81) NE 0/8 (0) [SCNEC lung]
Range (median) (%) 21-95 (76) 0 (0) 0-26 (0)
Total 187/313 (60) 0/43 (0) 7/387 (2)
[all cases]
168/224 (75)
[non-Australian]
*The 1 positive non-Merkel cell tumor was a presumed lung primary.
GI NET indicates gastrointestinal neuroendocrine tumor; MCC, Merkel cell carcinoma; NE, not evaluated; PDNEC, poorly differentiated neuroendocrine
carcinoma; SCC, squamous cell carcinoma; SCNEC, small cell neuroendocrine carcinoma.

N-terminal 260 residues of the LT Ag with improved sen- Gaffey et al262 failed to detect NF in any (0%) of 19 color-
sitivity. Ab3 IHC was positive in 56 of 58 (97%) MCCs and ectal tumors, as did Shin et al193 and Zamboni et al263 in 9
0 of 18 (0%) GI and lung PDNECs, while CM2B4 pos- and 3 breast and ampullary tumors, respectively; most
itivity was noted in only 81% of the same MCCs. Ab3’s recently, Lewis et al179 described expression in 1 of 14 (7%)
performance in an Australian group of tumors would be of head and neck mucosal-based tumors.
interest. The antibody is not yet commercially available. Like MCPyV IHC, detection of NF may be useful
to support a diagnosis of MCC. Individual laboratories
Neurofilament would need to verify a lack of significant expression in SCLC.
Neurofilaments (NFs) are 1 of 6 types of intermediate Additional data on NF expression in extrapulmonary
filaments. NF IHC has applications in neuropathology and PDNECs, some of which are clearly positive, are of interest.
soft tissue pathology, in which it is used to highlight axons.
NF IHC has been suggested as a marker of MCC, espe-
cially in its differential diagnosis with SCLC. In the 5 APPROACH TO A SUSPECTED METASTATIC NEN
largest series directly comparing NF expression in MCC An algorithmic approach to a suspected metastatic
and SCLC, expression has been noted in 67 of 93 (72%) WDNET, with an emphasis on assigning site of origin, is
and 0 of 143 (0%) tumors, respectively.166,189,190,257,258 presented in Figure 9. The first considerations are to dis-
Expression is typically perinuclear/dot-like. tinguish WDNET from other non-neuroendocrine tumor
As a note of caution, NF expression has been detected by types (eg, adenocarcinoma, squamous cell carcinoma) and
some investigators in SCLC, and it can be seen in a subset of from PDNEC. As illustrated in Figure 1C, WDNETs, espe-
extrapulmonary SCNECs. In 1983, Lehto et al259 first described cially those with type C histology, are occasionally mistaken
detection of NF expression in SCLCs (6/6, 100%; using a for adenocarcinomas. I have a very low threshold for per-
rabbit polyclonal antibody) and actually suggested its utility in forming IHC for broad-spectrum keratins (eg, AE1/AE3)
distinguishing this tumor type from other lung tumors (all 22 of and general neuroendocrine markers (eg, synaptophysin and
which were negative). In a follow-up study, van Muijen et al260 chromogranin). As discussed above and illustrated in
failed to detect NF expression with 2 different MoAbs in a Figure 3, WDNETs are sometimes mistaken for PDNECs,
series of 9 SCLCs. The largest study to detect NF expression in especially in crushed small biopsies. Ki-67 IHC guards
SCLC was reported by Shy et al,261 who found 69% of 67 against this pitfall and allows for assignment of WHO G1 or
tumors to be positive (utilizing an MoAb from Immunotec). G2 (with the caveats that this grading scheme only formally
These discrepant results may, at least in part, be attributable to applies to GEP tumors and that I have sometimes seen grade
differences in the antibodies used to study NF expression. discordance between metastases and matched primaries, with
Nagao et al157 detected NF in 6 of 15 (40%) SCNECs of the metastases generally of higher grade). For tumors with a
the major salivary glands, and McCluggage et al177 found NF PI > 20% an alternative diagnosis (eg, PDNEC) should be
in 7 of 21 (33%) PDNECs of the uterine cervix. Interestingly, considered, although tumors with well-differentiated histol-
at both of these sites primary PDNECs with a MCC-like ogy but a PI in the G3 range have been described.131,264
phenotype (ie, CK20 positive) are not uncommon, and while There are very limited data on transcription factor IHC in
Nagao and colleagues’ NF-positive cases coexpressed CK20, these rare tumors, although in 1 recent report Islet 1 was
McCluggage and colleagues’ did not. Otherwise, there are expressed in 5 of 7 (71%) well-differentiated/G3 pancreatic
little data on NF expression in extrapulmonary PDNECs. NENs versus only 1 of 13 (8%) pancreatic PDNECs.131

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 305


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

FIGURE 9. IHC workup of a metastatic WDNET of unknown origin. IHC indicates immunohistochemistry; NE, neuroendocrine; PI,
proliferation index; pPAX8, polyclonal PAX8; PR, progesterone receptor; PrAP, prostatic acid phosphatase; WDNET, well-differentiated
neuroendocrine tumor.

Once the diagnosis of metastatic WDNET is secured, results for TTF-1 and polyclonal PAX8 (or Islet 1) is strong
attention can turn to assigning site of origin. Any available support for a midgut origin, while TTF-1 positivity, with
clinical information, especially the results of imaging studies, is negative results for the other markers, strongly supports a
welcome. If the site of origin is “occult” at presentation, given lung origin. Polyclonal PAX8 (or Islet 1) positivity is less
multiple lines of evidence, including the Duke biochemical and specific, as it may be seen with pancreaticoduodenal or
survival data and the UCSF surgical exploration experience, rectal tumors; it also may be accompanied by CDX2
I believe a jejunoileal primary to be most likely. I examine the expression, which is typically weak and patchy. Given
tumor with this thought in mind, seeking out characteristic WDNET biology, though, I interpret polyclonal PAX8 (or
histologic features including the presence of a monomorphous Islet 1)-positive tumors to be likely pancreatic in origin.
population of round (rather than spindled) cells, perhaps Upper and lower endoscopy can be pursued to exclude
punctuated by an occasional larger nucleus; moderate (rather duodenal and rectal primaries, respectively. The final,
than voluminous) amounts of cytoplasm, ideally with scattered “triple-negative” pattern (although CDX2 may actually be
eosinophilic cytoplasmic granules; and disposition in a type A “weak, patchy positive”) is noninformative. I report it as
pattern (although I have certainly seen types B, C, D, and “unusual for a midgut tumor (although it may be seen in up
mixed patterns in metastases from midgut primaries). to 10%) and in keeping with, although not ‘diagnostic of,’ a
For a metastatic WDNET of unknown origin, I per- pancreatic origin” (as up to half of the metastases from the
form CDX2, TTF-1, and polyclonal PAX8 IHC. For pancreas are polyclonal PAX8 negative—an argument for
tumors with a known or suspected origin, I may use 1 or bringing Islet 1 IHC online). For the final 2 patterns,
more markers to confirm the clinical impression (eg, only additional IHC is of interest, and I have occasionally
CDX2 in a suspected midgut metastasis). I do not have Islet attempted PR or PrAP IHC in the setting of a non-
1 IHC in my laboratory at this time, although in the diagnostic immunophenotype. Finally, it is not unlikely
algorithm a positive reaction with the polyclonal PAX8 that additional markers will emerge in the near future that
antibody or for Islet 1 are considered equivalent. I have will further inform the assignment of site of origin.
divided the results of this core panel into 4 patterns. The As with the well-differentiated tumors, the first step in
presence of diffuse, strong CDX2 expression, with negative working up a PDNEC is to distinguish it from other tumor

306 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

types. This is especially important in the liver, where of the Lung, Pleura, Thymus and Heart. 3rd ed. Lyon: IARC
metastatic adenocarcinomas are so frequent. Adenocarci- Press; 2004:188–195.
nomas often contain minor populations of neuroendocrine 7. Matias-Guiu X, DeLellis R, Moley JF, et al. Medullary
cells (on the order of 1% to 5% of total cells), and the thyroid carcinoma. In: DeLellis RA, Lloyd RV, Heitz PU,
Eng C, eds. World Health Organization Classification of
results of synaptophysin and chromogranin IHC are Tumours Pathology and Genetics of Tumours of Endocrine
sometimes “overinterpreted” in this setting. Occasional Organs. 3rd ed. Lyon: IARC Press; 2004:86–91.
poorly differentiated tumors will have extensive neuro- 8. Williams ED, Sandler M. The classification of carcinoid
endocrine marker expression (usually synaptophysin rather tum ours. Lancet. 1963;1:238–239.
than chromogranin), while lacking the histologic features of 9. Soga J, Tazawa K. Pathologic analysis of carcinoids.
a PDNEC. These have been referred to as “non–small cell Histologic reevaluation of 62 cases. Cancer. 1971;28:990–998.
carcinomas with (occult) neuroendocrine differentiation.” It 10. Burke AP, Thomas RM, Elsayed AM, et al. Carcinoids of the
is important to distinguish these from true PDNECs, as jejunum and ileum: an immunohistochemical and clinicopa-
they are unlikely to respond to “small cell” chemotherapy. thologic study of 167 cases. Cancer. 1997;79:1086–1093.
11. Burke AP, Sobin LH, Federspiel BH, et al. Appendiceal
Although the histomorphology of SCNEC and LCNEC carcinoids: correlation of histology and immunohistochemis-
differs, a common feature, and one that generally dis- try. Mod Pathol. 1989;2:630–637.
tinguishes them from other high-grade carcinomas, is 12. Iwafuchi M, Watanabe H, Ajioka Y, et al. Immunohisto-
monomorphism. chemical and ultrastructural studies of twelve argentaffin and
In the skin, once the diagnosis of a PDNEC is made, six argyrophil carcinoids of the appendix vermiformis. Hum
the key distinction is between MCC and metastatic visceral Pathol. 1990;21:773–780.
PDNEC. This is usually easily accomplished with the mini- 13. Fiocca R, Rindi G, Capella C, et al. Glucagon, glicentin,
IHC panel of CK20 and TTF-1. CK20 positivity essentially proglucagon, PYY, PP and proPP-icosapeptide immunoreac-
confirms a diagnosis of MCC, while TTF-1 positivity tivities of rectal carcinoid tumors and related non-tumor cells.
Regul Pept. 1987;17:9–29.
refutes it. Given negativity for both markers, correlation
14. Burke AP, Federspiel BH, Sobin LH, et al. Carcinoids of the
with imaging studies is recommended, since, if a PDNEC is duodenum. A histologic and immunohistochemical study of
metastatic to the skin, it is apt to be detectable elsewhere. 65 tumors. Am J Surg Pathol. 1989;13:828–837.
NF and MCPyV (CM2B4) IHC may also be helpful in this 15. Garbrecht N, Anlauf M, Schmitt A, et al. Somatostatin-
setting. The latter of these may also be useful in the head producing neuroendocrine tumors of the duodenum and
and neck, as parotid gland tumors are often CK20 positive pancreas: incidence, types, biological behavior, association
but MCPyV negative. with inherited syndromes, and functional activity. Endocr
IHC is not useful in assigning the site of origin of a Relat Cancer. 2008;15:229–241.
visceral PDNEC. Pathologists often infer a lung origin 16. Johnson LA, Lavin P, Moertel CG, et al. Carcinoids: the
association of histologic growth pattern and survival. Cancer.
based on TTF-1 expression, but expression is also seen in
1983;51:882–889.
around 35% of extrapulmonary tumors. In fact, given a 17. Yao JC, Hassan M, Phan A, et al. One hundred years after
morphologic impression of SCNEC and negative chromo- “carcinoid”: epidemiology of and prognostic factors for
granin and synaptophysin immunostains, I have occasionally neuroendocrine tumors in 35,825 cases in the United States.
used TTF-1 expression to support the tumor diagnosis. J Clin Oncol. 2008;26:3063–3072.
Regardless of the site of origin, metastatic PDNECs are 18. Godwin JD II. Carcinoid tumors. An analysis of 2,837 cases.
generally treated with “small cell” chemotherapy. Cancer. 1975;36:560–569.
19. Modlin IM, Lye KD, Kidd MA. 5-decade analysis of 13,715
carcinoid tumors. Cancer. 2003;97:934–959.
REFERENCES
20. Kirshbom PM, Kherani AR, Onaitis MW, et al. Carcinoids
1. Rindi G, Arnold R, Bosman FT, et al. Nomenclature and of unknown origin: comparative analysis with foregut,
classification of neuroendocrine neoplams of the digestive midgut, and hindgut carcinoids. Surgery. 1998;124:
system. In: Bosman FT, Carneiro F, Hruban RH, Theise ND, 1063–1070.
eds. World Health Organization Classification of Tumours 21. Pape UF, Berndt U, Muller-Nordhorn J, et al. Prognostic
WHO Classification of Tumours of the Digestive System. factors of long-term outcome in gastroenteropancreatic neuro-
4th ed. Lyon: IARC; 2010:13–14. endocrine tumours. Endocr Relat Cancer. 2008;15:1083–1097.
2. Travis WD. The concept of pulmonary neuroendocrine 22. Lombard-Bohas C, Mitry E, O’Toole D, et al. Thirteen-month
tumours. In: Travis WD, Brambilla E, Muller-Hermelink registration of patients with gastroenteropancreatic endocrine
HK, Harris CC, eds. World Health Organization Classifica- tumours in France. Neuroendocrinology. 2009;89:217–222.
tion of Tumours Pathology and Genetics of Tumours of the 23. Garcia-Carbonero R, Capdevila J, Crespo-Herrero G, et al.
Lung, Pleura, Thymus and Heart. 3rd ed. Lyon: IARC Press; Incidence, patterns of care and prognostic factors for
2004:19–20. outcome of gastroenteropancreatic neuroendocrine tumors
3. Rekhtman N. Neuroendocrine tumors of the lung: an update. (GEP-NETs): results from the National Cancer Registry of
Arch Pathol Lab Med. 2010;134:1628–1638. Spain (RGETNE). Ann Oncol. 2010;21:1794–1803.
4. Pelosi G, Rodriguez J, Viale G, et al. Typical and atypical 24. Catena L, Bichisao E, Milione M, et al. Neuroendocrine
pulmonary carcinoid tumor overdiagnosed as small-cell tumors of unknown primary site: gold dust or misdiagnosed
carcinoma on biopsy specimens: a major pitfall in the neoplasms? Tumori. 2011;97:564–567.
management of lung cancer patients. Am J Surg Pathol. 25. Wang SC, Parekh JR, Zuraek MB, et al. Identification of
2005;29:179–187. unknown primary tumors in patients with neuroendocrine
5. Aslan DL, Gulbahce HE, Pambuccian SE, et al. Ki-67 liver metastases. Arch Surg. 2010;145:276–280.
immunoreactivity in the differential diagnosis of pulmonary 26. Bomanji JB, Papathanasiou ND. (1)(1)(1)In-DTPA(0)-
neuroendocrine neoplasms in specimens with extensive crush octreotide (Octreoscan), (1)(3)(1)I-MIBG and other agents
artifact. Am J Clin Pathol. 2005;123:874–878. for radionuclide therapy of NETs. Eur J Nucl Med Mol
6. Marx A, Shimosato Y, Kuo TT, et al. Thymic neuro- Imaging. 2012;39(suppl 1):S113–S125.
endocrine tumours. In: Travis WD, Brambilla E, Muller- 27. John M, Meyerhof W, Richter D, et al. Positive somatostatin
Hermelink HK, Harris CC, eds. World Health Organization receptor scintigraphy correlates with the presence of soma-
Classification of Tumours Pathology and Genetics of Tumours tostatin receptor subtype 2. Gut. 1996;38:33–39.

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 307


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

28. Janson ET, Stridsberg M, Gobl A, et al. Determination 45. Pavel M, Baudin E, Couvelard A, et al. ENETS Consensus
of somatostatin receptor subtype 2 in carcinoid tumors Guidelines for the management of patients with liver and
by immunohistochemical investigation with somatostatin other distant metastases from neuroendocrine neoplasms of
receptor subtype 2 antibodies. Cancer Res. 1998;58: foregut, midgut, hindgut, and unknown primary. Neuro-
2375–2378. endocrinology. 2012;95:157–176.
29. Kimura N, Pilichowska M, Date F, et al. Immuno- 46. Moertel CG, Lefkopoulo M, Lipsitz S, et al. Streptozocin-
histochemical expression of somatostatin type 2A receptor doxorubicin, streptozocin-fluorouracil or chlorozotocin in the
in neuroendocrine tumors. Clin Cancer Res. 1999;5: treatment of advanced islet-cell carcinoma. N Engl J Med.
3483–3487. 1992;326:519–523.
30. Papotti M, Bongiovanni M, Volante M, et al. Expression of 47. Strosberg JR, Fine RL, Choi J, et al. First-line chemotherapy
somatostatin receptor types 1-5 in 81 cases of gastrointestinal with capecitabine and temozolomide in patients with meta-
and pancreatic endocrine tumors. A correlative immunohis- static pancreatic endocrine carcinomas. Cancer. 2011;117:
tochemical and reverse-transcriptase polymerase chain reac- 268–275.
tion analysis. Virchows Arch. 2002;440:461–475. 48. Kulke MH, Hornick JL, Frauenhoffer C, et al. O6-
31. Ono K, Suzuki T, Miki Y, et al. Somatostatin receptor methylguanine DNA methyltransferase deficiency and
subtypes in human non-functioning neuroendocrine tumors response to temozolomide-based therapy in patients with
and effects of somatostatin analogue SOM230 on cell neuroendocrine tumors. Clin Cancer Res. 2009;15:338–345.
proliferation in cell line NCI-H727. Anticancer Res. 49. Sun W, Lipsitz S, Catalano P, et al. Phase II/III study of
2007;27(4B):2231–2239. doxorubicin with fluorouracil compared with streptozocin
32. Zamora V, Cabanne A, Salanova R, et al. Immunohisto- with fluorouracil or dacarbazine in the treatment of advanced
chemical expression of somatostatin receptors in digestive carcinoid tumors: Eastern Cooperative Oncology Group
endocrine tumours. Dig Liv Dis. 2010;42:220–225. Study E1281. J Clin Oncol. 2005;23:4897–4904.
33. Sclafani F, Carnaghi C, Di Tommaso L, et al. Detection of 50. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced
somatostatin receptor subtypes 2 and 5 by somatostatin pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:
receptor scintigraphy and immunohistochemistry: clinical 514–523.
implications in the diagnostic and therapeutic management 51. Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus
of gastroenteropancreatic neuroendocrine tumors. Tumori. octreotide long-acting repeatable for the treatment of
2011;97:620–628. advanced neuroendocrine tumours associated with carcinoid
34. Schmid HA, Lambertini C, van Vugt HH, et al. Monoclonal syndrome (RADIANT-2): a randomised, placebo-controlled,
antibodies against the human somatostatin receptor subtypes phase 3 study. Lancet. 2011;378:2005–2012.
1-5: development and immunohistochemical application 52. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for
in neuroendocrine tumors. Neuroendocrinology. 2012;95: the treatment of pancreatic neuroendocrine tumors. N Engl J
232–247. Med. 2011;364:501–513.
35. Prasad V, Ambrosini V, Hommann M, et al. Detection of 53. Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib
unknown primary neuroendocrine tumours (CUP-NET) in patients with advanced neuroendocrine tumors. J Clin
using (68)Ga-DOTA-NOC receptor PET/CT. Eur J Nucl Oncol. 2008;26:3403–3410.
Med Mol Imaging. 2010;37:67–77. 54. Kwekkeboom DJ, Krenning EP, Lebtahi R, et al. ENETS
36. Krausz Y, Freedman N, Rubinstein R, et al. 68Ga-DOTA- Consensus Guidelines for the Standards of Care in Neuro-
NOC PET/CT imaging of neuroendocrine tumors: compar- endocrine Tumors: peptide receptor radionuclide therapy
ison with (1)(1)(1)In-DTPA-octreotide (OctreoScan(R)). Mol with radiolabeled somatostatin analogs. Neuroendocrinology.
Imaging Biol. 2011;13:583–593. 2009;90:220–226.
37. Savelli G, Lucignani G, Seregni E, et al. Feasibility of 55. Imhof A, Brunner P, Marincek N, et al. Response, survival,
somatostatin receptor scintigraphy in the detection of occult and long-term toxicity after therapy with the radiolabeled
primary gastro-entero-pancreatic (GEP) neuroendocrine somatostatin analogue [90Y-DOTA]-TOC in metastasized
tumours. Nucl Med Commun. 2004;25:445–449. neuroendocrine cancers. J Clin Oncol. 2011;29:2416–2423.
38. Medicare physician fee schedule. 2013. Available at: http:// 56. Merkel cell carcinoma. 2012. Available at: http://www.nccn.
www.cms.gov/apps/physician-fee-schedule/overview.aspx. org/professionals/physician_gls/pdf/mcc.pdf. Accessed Feb-
Accessed April 7, 2013. ruary 5, 2013.
39. Sarmiento JM, Heywood G, Rubin J, et al. Surgical treat- 57. Moertel CG, Kvols LK, O’Connell MJ, et al. Treatment of
ment of neuroendocrine metastases to the liver: a plea neuroendocrine carcinomas with combined etoposide and
for resection to increase survival. J Am Coll Surg. 2003;197: cisplatin. Evidence of major therapeutic activity in the ana-
29–37. plastic variants of these neoplasms. Cancer. 1991;68:227–232.
40. Elias D, Lasser P, Ducreux M, et al. Liver resection (and 58. Strosberg JR, Coppola D, Klimstra DS, et al. The NANETS
associated extrahepatic resections) for metastatic well-differ- consensus guidelines for the diagnosis and management of
entiated endocrine tumors: a 15-year single center poorly differentiated (high-grade) extrapulmonary neuro-
prospective study. Surgery. 2003;133:375–382. endocrine carcinomas. Pancreas. 2010;39:799–800.
41. Chamberlain RS, Canes D, Brown KT, et al. Hepatic 59. Neuroendocrine tumors. 2013. Available at: http://www.nccn.
neuroendocrine metastases: does intervention alter outcomes? org/professionals/physician_gls/pdf/neuroendocrine.pdf. Accessed
J Am Coll Surg. 2000;190:432–445. April 30, 2013.
42. Chen H, Hardacre JM, Uzar A, et al. Isolated liver metastases 60. James R, Kazenwadel J. Homeobox gene expression in the
from neuroendocrine tumors: does resection prolong survival? intestinal epithelium of adult mice. J Biol Chem. 1991;266:
J Am Coll Surg. 1998;187:88–92; discussion-3. 3246–3251.
43. Maire F, Hammel P, Kianmanesh R, et al. Is adjuvant 61. Guo RJ, Suh ER, Lynch JP. The role of Cdx proteins in
therapy with streptozotocin and 5-fluorouracil useful after intestinal development and cancer. Cancer Biol Ther.
resection of liver metastases from digestive endocrine tumors? 2004;3:593–601.
Surgery. 2009;145:69–75. 62. Silberg DG, Swain GP, Suh ER, et al. Cdx1 and cdx2
44. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo- expression during intestinal development. Gastroenterology.
controlled, double-blind, prospective, randomized study on 2000;119:961–971.
the effect of octreotide LAR in the control of tumor growth in 63. Moskaluk CA, Zhang H, Powell SM, et al. Cdx2 protein
patients with metastatic neuroendocrine midgut tumors: a expression in normal and malignant human tissues: an
report from the PROMID Study Group. J Clin Oncol. immunohistochemical survey using tissue microarrays. Mod
2009;27:4656–4663. Pathol. 2003;16:913–919.

308 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

64. Bai YQ, Yamamoto H, Akiyama Y, et al. Ectopic expression 83. Rabban JT, Lerwill MF, McCluggage WG, et al. Primary
of homeodomain protein CDX2 in intestinal metaplasia and ovarian carcinoid tumors may express CDX-2: a potential
carcinomas of the stomach. Cancer Lett. 2002;176:47–55. pitfall in distinction from metastatic intestinal carcinoid
65. Phillips RW, Frierson HF Jr, Moskaluk CA. Cdx2 as a tumors involving the ovary. Int J Gynecol Pathol.
marker of epithelial intestinal differentiation in the esoph- 2009;28:41–48.
agus. Am J Surg Pathol. 2003;27:1442–1447. 84. Civitareale D, Lonigro R, Sinclair AJ, et al. A thyroid-specific
66. Gao N, White P, Kaestner KH. Establishment of intestinal nuclear protein essential for tissue-specific expression of the
identity and epithelial-mesenchymal signaling by Cdx2. Dev thyroglobulin promoter. EMBO J. 1989;8:2537–2542.
Cell. 2009;16:588–599. 85. Guazzi S, Price M, De Felice M, et al. Thyroid nuclear
67. Barbareschi M, Murer B, Colby TV, et al. CDX-2 homeobox factor 1 (TTF-1) contains a homeodomain and displays
gene expression is a reliable marker of colorectal adenocarci- a novel DNA binding specificity. EMBO J. 1990;9:
noma metastases to the lungs. Am J Surg Pathol. 2003;27: 3631–3639.
141–149. 86. Stanfel MN, Moses KA, Schwartz RJ, et al. Regulation of
68. Werling RW, Yaziji H, Bacchi CE, et al. CDX2, a highly organ development by the NKX-homeodomain factors: an
sensitive and specific marker of adenocarcinomas of intestinal NKX code. Cell Mol Biol. 2005;suppl 51:OL785–OL799.
origin: an immunohistochemical survey of 476 primary and 87. Lazzaro D, M Price, de Felice M, et al. The transcription
metastatic carcinomas. Am J Surg Pathol. 2003;27:303–310. factor TTF-1 is expressed at the onset of thyroid and lung
69. Borrisholt M, Nielsen S, Vyberg M. Demonstration of CDX2 morphogenesis and in restricted regions of the foetal brain.
is highly antibody dependant. Appl Immunohistochem Mol Development. 1991;113:1093–1104.
Morphol. 2013;21:64–72. 88. Ikeda K, Clark JC, Shaw-White JR, et al. Gene structure and
70. Koo J, Mertens RB, Mirocha JM, et al. Value of Islet 1 and expression of human thyroid transcription factor-1 in
PAX8 in identifying metastatic neuroendocrine tumors of respiratory epithelial cells. J Biol Chem. 1995;270:8108–8114.
pancreatic origin. Mod Pathol. 2012;25:893–901. 89. Katoh R, Miyagi E, Nakamura N, et al. Expression of
71. Barbareschi M, Roldo C, Zamboni G, et al. CDX-2 homeo- thyroid transcription factor-1 (TTF-1) in human C cells
box gene product expression in neuroendocrine tumors: its and medullary thyroid carcinomas. Hum Pathol. 2000;31:
role as a marker of intestinal neuroendocrine tumors. Am J 386–393.
Surg Pathol. 2004;28:1169–1176. 90. Kimura S, Hara Y, Pineau T, et al. The T/ebp null mouse:
72. Erickson LA, Papouchado B, Dimashkieh H, et al. Cdx2 as a thyroid-specific enhancer-binding protein is essential for the
marker for neuroendocrine tumors of unknown primary sites. organogenesis of the thyroid, lung, ventral forebrain, and
Endocr Pathol. 2004;15:247–252. pituitary. Genes Dev. 1996;10:60–69.
73. La Rosa S, Rigoli E, Uccella S, et al. CDX2 as a marker of 91. Bejarano PA, Baughman RP, Biddinger PW, et al. Surfactant
intestinal EC-cells and related well-differentiated endocrine proteins and thyroid transcription factor-1 in pulmonary and
tumors. Virchows Arch. 2004;445:248–254. breast carcinomas. Mod Pathol. 1996;9:445–452.
74. Saqi A, Alexis D, Remotti F, et al. Usefulness of CDX2 and 92. Kaufmann O, Dietel M. Thyroid transcription factor-1 is the
TTF-1 in differentiating gastrointestinal from pulmonary superior immunohistochemical marker for pulmonary adeno-
carcinoids. Am J Clin Pathol. 2005;123:394–404. carcinomas and large cell carcinomas compared to surfactant
75. Jaffee IM, Rahmani M, Singhal MG, et al. Expression of the proteins A and B. Histopathology. 2000;36:8–16.
intestinal transcription factor CDX2 in carcinoid tumors is a 93. Katoh R, Kawaoi A, Miyagi E, et al. Thyroid transcription
marker of midgut origin. Arch Pathol Lab Med. 2006;130: factor-1 in normal, hyperplastic, and neoplastic follicular
1522–1526. thyroid cells examined by immunohistochemistry and non-
76. Lin X, Saad RS, Luckasevic TM, et al. Diagnostic value of radioactive in situ hybridization. Mod Pathol. 2000;13:
CDX-2 and TTF-1 expressions in separating metastatic 570–576.
neuroendocrine neoplasms of unknown origin. Appl Immu- 94. Bejarano PA, Nikiforov YE, Swenson ES, et al. Thyroid
nohistochem Mol Morphol. 2007;15:407–414. transcription factor-1, thyroglobulin, cytokeratin 7, and
77. Schmitt AM, Riniker F, Anlauf M, et al. Islet 1 (Isl1) cytokeratin 20 in thyroid neoplasms. Appl Immunohistochem
expression is a reliable marker for pancreatic endocrine Mol Morphol. 2000;8:189–194.
tumors and their metastases. Am J Surg Pathol. 2008;32: 95. Ordóñez NG. Thyroid transcription factor-1 is a marker
420–425. of lung and thyroid carcinomas. Adv Anat Pathol. 2000;7:
78. Srivastava A, Hornick JL. Immunohistochemical staining for 123–127.
CDX-2, PDX-1, NESP-55, and TTF-1 can help distinguish 96. Kaufmann O, Dietel M. Expression of thyroid transcription
gastrointestinal carcinoid tumors from pancreatic endocrine factor-1 in pulmonary and extrapulmonary small cell
and pulmonary carcinoid tumors. Am J Surg Pathol. carcinomas and other neuroendocrine carcinomas of various
2009;33:626–632. primary sites. Histopathology. 2000;36:415–420.
79. Hermann G, Konukiewitz B, Schmitt A, et al. Hormonally 97. Oliveira AM, Tazelaar HD, Myers JL, et al. Thyroid
defined pancreatic and duodenal neuroendocrine tumors transcription factor-1 distinguishes metastatic pulmonary
differ in their transcription factor signatures: expression of from well-differentiated neuroendocrine tumors of
ISL1, PDX1, NGN3, and CDX2. Virchows Arch. 2011;459: other sites. Am J Surg Pathol. 2001;25:815–819.
147–154. 98. Agoff SN, Lamps LW, Philip AT, et al. Thyroid transcription
80. Chan ES, Alexander J, Swanson PE, et al. PDX-1, CDX-2, factor-1 is expressed in extrapulmonary small cell carcinomas
TTF-1, and CK7: a reliable immunohistochemical panel for but not in other extrapulmonary neuroendocrine tumors.
pancreatic neuroendocrine neoplasms. Am J Surg Pathol. Mod Pathol. 2000;13:238–242.
2012;36:737–743. 99. Hirsch MS, Faquin WC, Krane JF. Thyroid transcription
81. Denby KS, Briones AJ, Bourne PA, et al. IMP3, NESP55, factor-1, but not p53, is helpful in distinguishing moderately
TTF-1 and CDX2 serve as an immunohistochemical panel in differentiated neuroendocrine carcinoma of the larynx from
the distinction among small-cell carcinoma, gastrointestinal medullary carcinoma of the thyroid. Mod Pathol.
carcinoid, and pancreatic endocrine tumor metastasized to 2004;17:631–636.
the liver. Appl Immunohistochem Mol Morphol. 2012;20: 100. Holzinger A, Dingle S, Bejarano PA, et al. Monoclonal
573–579. antibody to thyroid transcription factor-1: production,
82. Graham RP, Shrestha B, Caron BL, et al. Islet-1 is a sensitive characterization, and usefulness in tumor diagnosis. Hybrid-
but not entirely specific marker for pancreatic neuroendocrine oma. 1996;15:49–53.
neoplasms and their metastases. Am J Surg Pathol. 2013; 101. Compérat E, Zhang F, Perrotin C, et al. Variable sensitivity
37:399–405. and specificity of TTF-1 antibodies in lung metastatic

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 309


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

adenocarcinoma of colorectal origin. Mod Pathol. 2005;18: 122. Tong GX, Yu WM, Beaubier NT, et al. Expression of PAX8
1371–1376. in normal and neoplastic renal tissues: an immuno-
102. Matoso A, Singh K, Jacob R, et al. Comparison of thyroid histochemical study. Mod Pathol. 2009;22:1218–1227.
transcription factor-1 expression by 2 monoclonal antibodies 123. Long KB, Srivastava A, Hirsch MS, et al. PAX8 Expression
in pulmonary and nonpulmonary primary tumors. Appl in well-differentiated pancreatic endocrine tumors: correla-
Immunohistochem Mol Morphol. 2010;18:142–149. tion with clinicopathologic features and comparison with
103. Fabbro D, Di Loreto C, Stamerra O, et al. TTF-1 gene gastrointestinal and pulmonary carcinoid tumors. Am J Surg
expression in human lung tumours. Eur J Cancer. 1996;32A: Pathol. 2010;34:723–729.
512–517. 124. Sangoi AR, Ohgami RS, Pai RK, et al. PAX8 expression
104. Folpe AL, Gown AM, Lamps LW, et al. Thyroid tran- reliably distinguishes pancreatic well-differentiated neuro-
scription factor-1: immunohistochemical evaluation in endocrine tumors from ileal and pulmonary well-differ-
pulmonary neuroendocrine tumors. Mod Pathol. 1999;12:5–8. entiated neuroendocrine tumors and pancreatic acinar cell
105. Cai YC, Banner B, Glickman J, et al. Cytokeratin 7 and 20 carcinoma. Mod Pathol. 2011;24:412–424.
and thyroid transcription factor 1 can help distinguish 125. Ozcan A, Shen SS, Hamilton C, et al. PAX 8 expression in
pulmonary from gastrointestinal carcinoid and pancreatic non-neoplastic tissues, primary tumors, and metastatic
endocrine tumors. Hum Pathol. 2001;32:1087–1093. tumors: a comprehensive immunohistochemical study. Mod
106. Sturm N, Rossi G, Lantuejoul S, et al. Expression of thyroid Pathol. 2011;24:751–764.
transcription factor-1 in the spectrum of neuroendocrine cell 126. Haynes CM, Sangoi AR, Pai RK. PAX8 is expressed in
lung proliferations with special interest in carcinoids. Hum pancreatic well-differentiated neuroendocrine tumors and in
Pathol. 2002;33:175–182. extrapancreatic poorly differentiated neuroendocrine carci-
107. Zamecnik J, Kodet R. Value of thyroid transcription factor-1 nomas in fine-needle aspiration biopsy specimens. Cancer
and surfactant apoprotein A in the differential diagnosis of Cytopathol. 2011;119:193–201.
pulmonary carcinomas: a study of 109 cases. Virchows Arch. 127. Laury AR, Perets R, Piao H, et al. A comprehensive analysis
2002;440:353–361. of PAX8 expression in human epithelial tumors. Am J Surg
108. Chang YL, Lee YC, Liao WY, et al. The utility and limitation of Pathol. 2011;35:816–826.
thyroid transcription factor-1 protein in primary and metastatic 128. Tacha D, Zhou D, Cheng L. Expression of PAX8 in normal and
pulmonary neoplasms. Lung Cancer. 2004;44:149–157. neoplastic tissues: a comprehensive immunohistochemical study.
109. Du EZ, Goldstraw P, Zacharias J, et al. TTF-1 expression is Appl Immunohistochem Mol Morphol. 2011;19:293–299.
specific for lung primary in typical and atypical carcinoids: 129. Lorenzo PI, Jimenez Moreno CM, Delgado I, et al. Immu-
TTF-1-positive carcinoids are predominantly in peripheral nohistochemical assessment of Pax8 expression during
location. Hum Pathol. 2004;35:825–831. pancreatic islet development and in human neuroendocrine
110. Jerome Marson V, Mazieres J, Groussard O, et al. Expression tumors. Histochem Cell Biol. 2011;136:595–607.
of TTF-1 and cytokeratins in primary and secondary 130. Tacha D, Qi W, Zhou D, et al. PAX8 mouse monoclonal
epithelial lung tumours: correlation with histological type antibody [BC12] recognizes a restricted epitope and is highly
and grade. Histopathology. 2004;45:125–134. sensitive in renal cell and ovarian cancers but does not cross-
111. Hiroshima K, Iyoda A, Shida T, et al. Distinction of react with b cells and tumors of pancreatic origin. Appl
pulmonary large cell neuroendocrine carcinoma from small Immunohistochem Mol Morphol. 2013;21:59–63.
cell lung carcinoma: a morphological, immunohistochemical, 131. Agaimy A, Erlenbach-Wunsch K, Konukiewitz B, et al. ISL1
and molecular analysis. Mod Pathol. 2006;19:1358–1368. expression is not restricted to pancreatic well-differentiated
112. La Rosa S, Chiaravalli AM, Placidi C, et al. TTF1 expression neuroendocrine neoplasms, but is also commonly found in
in normal lung neuroendocrine cells and related tumors: well and poorly differentiated neuroendocrine neoplasms of
immunohistochemical study comparing two different mono- extrapancreatic origin. Mod Pathol. 2013;26:995–1003.
clonal antibodies. Virchows Arch. 2010;457:497–507. 132. Fendrich V, Ramerth R, Waldmann J, et al. Sonic hedgehog
113. Tsuta K, Kalhor N, Wistuba II, et al. Clinicopathological and and pancreatic-duodenal homeobox 1 expression distinguish
immunohistochemical analysis of spindle-cell carcinoid between duodenal and pancreatic gastrinomas. Endocr Relat
tumour of the lung. Histopathology. 2011;59:526–536. Cancer. 2009;16:613–622.
114. Masai K, Tsuta K, Kawago M, et al. Expression of squamous 133. Zhang L, Smyrk TC, Oliveira AM, et al. KIT is an
cell carcinoma markers and adenocarcinoma markers in independent prognostic marker for pancreatic endocrine
primary pulmonary neuroendocrine carcinomas. Appl Immu- tumors: a finding derived from analysis of islet cell differ-
nohistochem Mol Morphol. 2013;21:292–297. entiation markers. Am J Surg Pathol. 2009;33:1562–1569.
115. Macchia PE, Lapi P, Krude H, et al. PAX8 mutations 134. Park JY, Hong SM, Klimstra DS, et al. Pdx1 expression in
associated with congenital hypothyroidism caused by thyroid pancreatic precursor lesions and neoplasms. Appl Immuno-
dysgenesis. Nat Genet. 1998;19:83–86. histochem Mol Morphol. 2011;19:444–449.
116. Mansouri A, Chowdhury K, Gruss P. Follicular cells of the 135. Jeung JA, Cao D, Selli BW, et al. Primary renal carcinoid
thyroid gland require Pax8 gene function. Nat Genet. tumors: clinicopathologic features of 9 cases with emphasis on
1998;19:87–90. novel immunohistochemical findings. Hum Pathol. 2011;42:
117. Plachov D, Chowdhury K, Walther C, et al. Pax8 a murine 1554–1561.
paired box gene expressed in the developing excretory system 136. Sosa-Pineda B, Chowdhury K, Torres M, et al. The Pax4 gene
and thyroid gland. Development. 1990;110:643–651. is essential for differentiation of insulin-producing beta
118. Bouchard M, Souabni A, Mandler M, et al. Nephric lineage cells in the mammalian pancreas. Nature. 1997;386:399–402.
specification by Pax2 and Pax8. Genes Dev. 2002;16: 137. St-Onge L, Sosa-Pineda B, Chowdhury K, et al. Pax6 is
2958–2970. required for differentiation of glucagon-producing alpha-cells
119. Schaner ME, Ross DT, Ciaravino G, et al. Gene expression in mouse pancreas. Nature. 1997;387:406–409.
patterns in ovarian carcinomas. Mol Biol Cell. 2003;14: 138. Sander M, Neubuser A, Kalamaras J, et al. Genetic analysis
4376–4386. reveals that PAX6 is required for normal transcription of
120. Nonaka D, Tang Y, Chiriboga L, et al. Diagnostic utility pancreatic hormone genes and islet development. Genes Dev.
of thyroid transcription factors Pax8 and TTF-2 1997;11:1662–1673.
(FoxE1) in thyroid epithelial neoplasms. Mod Pathol. 2008; 139. Moreno CM, Lorenzo PI, Delgado I, et al. Pax8 detection
21:192–200. in well-differentiated pancreatic endocrine tumors: how
121. Nonaka D, Chiriboga L, Soslow RA. Expression of pax8 as a reliable is it? Am J Surg Pathol. 2011;35:1906–1908.
useful marker in distinguishing ovarian carcinomas from 140. Karlsson O, Thor S, Norberg T, et al. Insulin gene enhancer
mammary carcinomas. Am J Surg Pathol. 2008;32:1566–1571. binding protein Isl-1 is a member of a novel class of proteins

310 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

containing both a homeo- and a Cys-His domain. Nature. carcinoma of hypercalcemic type. Int J Gynecol Pathol.
1990;344:879–882. 2004;23:330–336.
141. Thor S, Ericson J, Brannstrom T, et al. The homeodomain 160. Liu J, Farhood A. Thyroid transcription factor-1 immuno-
LIM protein Isl-1 is expressed in subsets of neurons and cytochemical staining of pleural fluid cytocentrifuge prepara-
endocrine cells in the adult rat. Neuron. 1991;7:881–889. tions for detection of small cell lung carcinoma. Acta Cytol.
142. Ahlgren U, Pfaff SL, Jessell TM, et al. Independent require- 2004;48:635–640.
ment for ISL1 in formation of pancreatic mesenchyme and 161. Zhang H, Liu J, Cagle PT, et al. Distinction of pulmonary
islet cells. Nature. 1997;385:257–260. small cell carcinoma from poorly differentiated squamous cell
143. Di Loreto C, Di Lauro V, Puglisi F, et al. Immunocytochem- carcinoma: an immunohistochemical approach. Mod Pathol.
ical expression of tissue specific transcription factor-1 in lung 2005;18:111–118.
carcinoma. J Clin Pathol. 1997;50:30–32. 162. Nassar H, Albores-Saavedra J, Klimstra DS. High-grade
144. Byrd-Gloster AL, Khoor A, Glass LF, et al. Differential neuroendocrine carcinoma of the ampulla of vater: a
expression of thyroid transcription factor 1 in small cell lung clinicopathologic and immunohistochemical analysis of
carcinoma and Merkel cell tumor. Hum Pathol. 2000;31:58–62. 14 cases. Am J Surg Pathol. 2005;29:588–594.
145. Hanly AJ, Elgart GW, Jorda M, et al. Analysis of thyroid 163. Llombart B, Monteagudo C, Lopez-Guerrero JA, et al.
transcription factor-1 and cytokeratin 20 separates merkel cell Clinicopathological and immunohistochemical analysis of 20
carcinoma from small cell carcinoma of lung. J Cutan Pathol. cases of Merkel cell carcinoma in search of prognostic
2000;27:118–120. markers. Histopathology. 2005;46:622–634.
146. Ordóñez NG. Value of thyroid transcription factor-1 164. Jones TD, Kernek KM, Yang XJ, et al. Thyroid transcription
immunostaining in distinguishing small cell lung carcinomas factor 1 expression in small cell carcinoma of the urinary
from other small cell carcinomas. Am J Surg Pathol. bladder: an immunohistochemical profile of 44 cases. Hum
2000;24:1217–1223. Pathol. 2005;36:718–723.
147. Cheuk W, Kwan MY, Suster S, et al. Immunostaining for 165. Wu M, Szporn AH, Zhang D, et al. Cytology applications of
thyroid transcription factor 1 and cytokeratin 20 aids the p63 and TTF-1 immunostaining in differential diagnosis of
distinction of small cell carcinoma from Merkel cell carci- lung cancers. Diagn Cytopathol. 2005;33:223–227.
noma, but not pulmonary from extrapulmonary small cell 166. Bobos M, Hytiroglou P, Kostopoulos I, et al. Immunohis-
carcinomas. Arch Pathol Lab Med. 2001;125:228–231. tochemical distinction between merkel cell carcinoma and
148. Shin SJ, DeLellis RA, Rosen PP. Small cell carcinoma of the small cell carcinoma of the lung. Am J Dermatopathol.
breast–additional immunohistochemical studies. Am J Surg 2006;28:99–104.
Pathol. 2001;25:831–832. 167. Yao JL, Madeb R, Bourne P, et al. Small cell carcinoma of
149. Leech SN, Kolar AJ, Barrett PD, et al. Merkel cell carcinoma the prostate: an immunohistochemical study. Am J Surg
can be distinguished from metastatic small cell carcinoma Pathol. 2006;30:705–712.
using antibodies to cytokeratin 20 and thyroid transcription 168. Kalhor N, Zander DS, Liu J. TTF-1 and p63 for distinguish-
factor 1. J Clin Pathol. 2001;54:727–729. ing pulmonary small-cell carcinoma from poorly differ-
150. Sturm N, Lantuejoul S, Laverriere MH, et al. Thyroid entiated squamous cell carcinoma in previously pap-stained
transcription factor 1 and cytokeratins 1, 5, 10, 14 cytologic material. Mod Pathol. 2006;19:1117–1123.
(34betaE12) expression in basaloid and large-cell neuro- 169. Yun JP, Zhang MF, Hou JH, et al. Primary small cell
endocrine carcinomas of the lung. Hum Pathol. carcinoma of the esophagus: clinicopathological and immu-
2001;32:918–925. nohistochemical features of 21 cases. BMC Cancer. 2007;7:38.
151. Chhieng DC, Cangiarella JF, Zakowski MF, et al. Use of 170. Carlson JW, Nucci MR, Brodsky J, et al. Biomarker-assisted
thyroid transcription factor 1, PE-10, and cytokeratins 7 and diagnosis of ovarian, cervical and pulmonary small cell
20 in discriminating between primary lung carcinomas and carcinomas: the role of TTF-1, WT-1 and HPV analysis.
metastatic lesions in fine-needle aspiration biopsy specimens. Histopathology. 2007;51:305–312.
Cancer. 2001;93:330–336. 171. El Demellawy D, Khalifa MA, Ismiil N, et al. Primary
152. Wu M, Wang B, Gil J, et al. p63 and TTF-1 immunostaining. colorectal small cell carcinoma: a clinicopathological and
A useful marker panel for distinguishing small cell carcinoma immunohistochemical study of 10 cases. Diagn Pathol.
of lung from poorly differentiated squamous cell carcinoma 2007;2:35.
of lung. Am J Clin Pathol. 2003;119:696–702. 172. Alijo Serrano F, Sanchez-Mora N, Angel Arranz J, et al.
153. Myong NH. Thyroid transcription factor-1 (TTF-1) expres- Large cell and small cell neuroendocrine bladder carcinoma:
sion in human lung carcinomas: its prognostic implication immunohistochemical and outcome study in a single institu-
and relationship with wxpressions of p53 and Ki-67 proteins. tion. Am J Clin Pathol. 2007;128:733–739.
J Korean Med Sci. 2003;18:494–500. 173. Kargi A, Gurel D, Tuna B. The diagnostic value of TTF-1,
154. Yamamoto J, Ohshima K, Ikeda S, et al. Primary esophageal CK 5/6, and p63 immunostaining in classification of lung
small cell carcinoma with concomitant invasive squamous cell carcinomas. Appl Immunohistochem Mol Morphol. 2007;15:
carcinoma or carcinoma in situ. Hum Pathol. 2003;34: 415–420.
1108–1115. 174. Lu J, Xue LY, Lu N, et al. Superficial primary small cell
155. Yang DT, Holden JA, Florell SR. CD117, CK20, TTF-1, and carcinoma of the esophagus: clinicopathological and immu-
DNA topoisomerase II-alpha antigen expression in small cell nohistochemical analysis of 15 cases. Dis Esophagus.
tumors. J Cutan Pathol. 2004;31:254–261. 2010;23:153–159.
156. Liu J, Farhood A. Immunostaining for thyroid transcription 175. Wang W, Epstein JI. Small cell carcinoma of the prostate. A
factor-1 on fine-needle aspiration specimens of lung tumors: a morphologic and immunohistochemical study of 95 cases. Am
comparison of direct smears and cell block preparations. J Surg Pathol. 2008;32:65–71.
Cancer. 2004;102:109–114. 176. Ralston J, Chiriboga L, Nonaka D. MASH1: a useful marker
157. Nagao T, Gaffey TA, Olsen KD, et al. Small cell carcinoma in differentiating pulmonary small cell carcinoma from
of the major salivary glands: clinicopathologic study with Merkel cell carcinoma. Mod Pathol. 2008;21:1357–1362.
emphasis on cytokeratin 20 immunoreactivity and clinical 177. McCluggage WG, Kennedy K, Busam KJ. An immunohisto-
outcome. Am J Surg Pathol. 2004;28:762–770. chemical study of cervical neuroendocrine carcinomas: Neo-
158. Soriano P, Navarro S, Gil M, et al. Small-cell carcinoma of plasms that are commonly TTF1 positive and which may
the urinary bladder. A clinico-pathological study of ten cases. express CK20 and P63. Am J Surg Pathol. 2010;34:525–532.
Virchows Arch. 2004;445:292–297. 178. Li AF, Li AC, Hsu CY, et al. Small cell carcinomas in
159. McCluggage WG, Oliva E, Connolly LE, et al. An gastrointestinal tract: immunohistochemical and clinicopa-
immunohistochemical analysis of ovarian small cell thological features. J Clin Pathol. 2010;63:620–625.

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 311


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

179. Lewis JS Jr, Duncavage E, Klonowski PW. Oral cavity 199. Ishida GM, Kato N, Hayasaka T, et al. Small cell neuro-
neuroendocrine carcinoma: a comparison study with cuta- endocrine carcinomas of the uterine cervix: a histological,
neous Merkel cell carcinoma and other mucosal head and immunohistochemical, and molecular genetic study. Int J
neck neuroendocrine carcinomas. Oral Surg Oral Med Oral Gynecol Pathol. 2004;23:366–372.
Pathol Oral Radiol Endod. 2010;110:209–217. 200. Acebo E, Vidaurrazaga N, Varas C, et al. Merkel cell
180. Sidiropoulos M, Hanna W, Raphael SJ, et al. Expression of carcinoma: a clinicopathological study of 11 cases. J Eur Acad
TdT in Merkel cell carcinoma and small cell lung carcinoma. Dermatol Venereol. 2005;19:546–551.
Am J Clin Pathol. 2011;135:831–838. 201. Saeb-Lima M, Montante-Montes de Oca D, Albores-Saave-
181. Fernández-Aceñero MJ, Cordova S, Manzarbeitia F, et al. dra J. Merkel cell carcinoma with eccrine differentiation: a
Immunohistochemical profile of urothelial and small cell clinicopathologic study of 7 cases. Ann Diagn Pathol.
carcinomas of the bladder. Pathol Oncol Res. 2011;17:519–523. 2008;12:410–414.
182. Thompson S, Cioffi-Lavina M, Chapman-Fredricks J, et al. 202. Chernock RD, Duncavage EJ, Gnepp DR, et al. Absence of
Distinction of high-grade neuroendocrine carcinoma/small Merkel cell polyomavirus in primary parotid high-grade
cell carcinoma from conventional urothelial carcinoma of neuroendocrine carcinomas regardless of cytokeratin 20
urinary bladder: an immunohistochemical approach. Appl immunophenotype. Am J Surg Pathol. 2011;35:1806–1811.
Immunohistochem Mol Morphol. 2011;19:395–399. 203. Moll R, Franke WW. Cytoskeletal differences between human
183. Quinn AM, Blackhall F, Wilson G, et al. Extrapulmonary neuroendocrine tumors: a cytoskeletal protein of molecular
small cell carcinoma: a clinicopathological study with weight 46,000 distinguishes cutaneous from pulmonary neuro-
identification of potential diagnostic mimics. Histopathology. endocrine neoplasms. Differentiation. 1985;30:165–175.
2012;61:454–464. 204. Moll R, Franke WW, Schiller DL, et al. The catalog of
184. Moll R, Lowe A, Laufer J, et al. Cytokeratin 20 in human human cytokeratins: patterns of expression in normal
carcinomas. A new histodiagnostic marker detected by epithelia, tumors and cultured cells. Cell. 1982;31:11–24.
monoclonal antibodies. Am J Pathol. 1992;140:427–447. 205. Moll R, Schiller DL, Franke WW. Identification of protein
185. Brinkschmidt C, Stolze P, Fahrenkamp AG, et al. Immuno- IT of the intestinal cytoskeleton as a novel type I cytokeratin
histochemical demonstration of chromogranin A, chromog- with unusual properties and expression patterns. J Cell Biol.
ranin B, and secretoneurin in Merkel cell carcinoma of the 1990;111:567–580.
skin: an immunohistochemical study on 18 cases suggesting 206. Broers JL, Ramaekers FC, Rot MK, et al. Cytokeratins
two types of Merkel cell carcinoma. Appl Immunohistochem. in different types of human lung cancer as monitored by
1995;3:37–44. chain-specific monoclonal antibodies. Cancer Res. 1988;48:
186. Wang NP, Zee S, Zarbo RJ, et al. Coordinate expression of 3221–3229.
cytokeratins 7 and 20 defined unique subsets of carcinomas. 207. Alsaad KO, Serra S, Schmitt A, et al. Cytokeratins 7 and 20
Appl Immunohistochem. 1995;3:99–107. immunoexpression profile in goblet cell and classical carci-
187. Miettinen M. Keratin 20: immunohistochemical marker for noids of appendix. Endocr Pathol. 2007;18:16–22.
gastrointestinal, urothelial, and Merkel cell carcinomas. Mod 208. Jiang Y, Long H, Wang W, et al. Clinicopathological features
Pathol. 1995;8:384–388. and immunoexpression profiles of goblet cell carcinoid and
188. Chan JK, Suster S, Wenig BM, et al. Cytokeratin 20 typical carcinoid of the appendix. Pathol Oncol Res.
immunoreactivity distinguishes Merkel cell (primary cuta- 2011;17:127–132.
neous neuroendocrine) carcinomas and salivary gland small 209. Ischia R, Lovisetti-Scamihorn P, Hogue-Angeletti R, et al.
cell carcinomas from small cell carcinomas of various sites. Molecular cloning and characterization of NESP55, a novel
Am J Surg Pathol. 1997;21:226–234. chromogranin-like precursor of a peptide with 5-HT1B receptor
189. Schmidt U, Muller U, Metz KA, et al. Cytokeratin and antagonist activity. J Biol Chem. 1997;272:11657–11662.
neurofilament protein staining in Merkel cell carcinoma of the 210. Jakobsen AM, Ahlman H, Kolby L, et al. NESP55, a novel
small cell type and small cell carcinoma of the lung. Am J chromogranin-like peptide, is expressed in endocrine tumours
Dermatopathol. 1998;20:346–351. of the pancreas and adrenal medulla but not in ileal
190. Metz KA, Jacob M, Schmidt U, et al. Merkel cell carcinoma carcinoids. Br J Cancer. 2003;88:1746–1754.
of the eyelid: histological and immunohistochemical features 211. Srivastava A, Padilla O, Fischer-Colbrie R, et al. Neuro-
with special respect to differential diagnosis. Graefes Arch endocrine secretory protein-55 (NESP-55) expression discrim-
Clin Exp Ophthalmol. 1998;236:561–566. inates pancreatic endocrine tumors and pheochromocytomas
191. Scott MP, Helm KF. Cytokeratin 20: a marker for diagnosing from gastrointestinal and pulmonary carcinoids. Am J Surg
Merkel cell carcinoma. Am J Dermatopathol. 1999;21:16–20. Pathol. 2004;28:1371–1378.
192. Nicholson SA, McDermott MB, Swanson PE, et al. CD99 212. Ohlsson H, Karlsson K, Edlund T. IPF1 a homeodomain-
and cytokeratin-20 in small-cell and basaloid tumors containing transactivator of the insulin gene. EMBO J. 1993;
of the skin. Appl Immunohistochem Mol Morphol. 2000;8:37–41. 12:4251–4259.
193. Shin SJ, DeLellis RA, Ying L, et al. Small cell carcinoma of 213. Leonard J, Peers B, Johnson T, et al. Characterization of
the breast: a clinicopathologic and immunohistochemical somatostatin transactivating factor-1, a novel homeobox
study of nine patients. Am J Surg Pathol. 2000;24:1231–1238. factor that stimulates somatostatin expression in pancreatic
194. Chu P, Wu E, Weiss LM. Cytokeratin 7 and cytokeratin 20 islet cells. Mol Endocrinol. 1993;7:1275–1283.
expression in epithelial neoplasms: a survey of 435 cases. Mod 214. Offield MF, Jetton TL, Labosky PA, et al. PDX-1 is required
Pathol. 2000;13:962–972. for pancreatic outgrowth and differentiation of the rostral
195. Jensen K, Kohler S, Rouse RV. Cytokeratin staining in duodenum. Development. 1996;122:983–995.
Merkel cell carcinoma: an immunohistochemical study of 215. Jonsson J, Carlsson L, Edlund T, et al. Insulin-promoter-
cytokeratins 5/6, 7, 17, and 20. Appl Immunohistochem Mol factor 1 is required for pancreas development in mice. Nature.
Morphol. 2000;8:310–315. 1994;371:606–609.
196. Kende AI, Carr NJ, Sobin LH. Expression of cytokeratins 7 216. Wilander E, Grimelius L, Portela-Gomes G, et al. Substance
and 20 in carcinomas of the gastrointestinal tract. Histopa- P and enteroglucagon-like immunoreactivity in argentaffin
thology. 2003;42:137–140. and argyrophil midgut carcinoid tumours. Scand J Gastro-
197. Mott RT, Smoller BR, Morgan MB. Merkel cell carcinoma: a enterol Suppl. 1979;53:19–25.
clinicopathologic study with prognostic implications. J Cutan 217. Alumets J, Alm P, Falkmer S, et al. Immunohistochemical
Pathol. 2004;31:217–223. evidence of peptide hormones in endocrine tumors of the
198. Tanaka Y, Sano T, Qian ZR, et al. Expression of adhesion rectum. Cancer. 1981;48:2409–2415.
molecules and cytokeratin 20 in merkel cell carcinomas. 218. O’Briain DS, Dayal Y, DeLellis RA, et al. Rectal carcinoids
Endocr Pathol. 2004;15:117–129. as tumors of the hindgut endocrine cells: a morphological and

312 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins


Adv Anat Pathol  Volume 20, Number 5, September 2013 Neuroendocrine Neoplasms: Assigning Site of Origin

immunohistochemical analysis. Am J Surg Pathol. 1982;6: 239. Körner M, Waser B, Schonbrunn A, et al. Somatostatin
131–142. receptor subtype 2A immunohistochemistry using a new
219. Yang K, Ulich T, Cheng L, et al. The neuroendocrine monoclonal antibody selects tumors suitable for in vivo
products of intestinal carcinoids. An immunoperoxidase somatostatin receptor targeting. Am J Surg Pathol. 2012;
study of 35 carcinoid tumors stained for serotonin and eight 36:242–252.
polypeptide hormones. Cancer. 1983;51:1918–1926. 240. Erickson JD, Schafer MK, Bonner TI, et al. Distinct
220. Shimoda T, Ishikawa E, Sano T, et al. Histopathological and pharmacological properties and distribution in neurons and
immunohistochemical study of neuroendocrine tumors of the endocrine cells of two isoforms of the human vesicular
rectum. Acta Pathol Jpn. 1984;34:1059–1077. monoamine transporter. Proc Natl Acad Sci U S A. 1996;
221. Warren WH, Memoli VA, Gould VE. Immunohistochemical 93:5166–5171.
and ultrastructural analysis of bronchopulmonary neuroendo- 241. Rindi G, Paolotti D, Fiocca R, et al. Vesicular monoamine
crine neoplasms. I. Carcinoids. Ultrastruct Pathol. 1984;6:15–27. transporter 2 as a marker of gastric enterochromaffin-like cell
222. Federspiel BH, Burke AP, Sobin LH, et al. Rectal and colonic tumors. Virchows Arch. 2000;436:217–223.
carcinoids. A clinicopathologic study of 84 cases. Cancer. 242. Anlauf M, Eissele R, Schafer MK, et al. Expression of the two
1990;65:135–140. isoforms of the vesicular monoamine transporter (VMAT1 and
223. Doglioni C, Gambacorta M, Zamboni G, et al. Immunocy- VMAT2) in the endocrine pancreas and pancreatic endocrine
tochemical localization of progesterone receptors in endocrine tumors. J Histochem Cytochem. 2003;51:1027–1040.
cells of the human pancreas. Am J Pathol. 1990;137:999–1005. 243. Uccella S, Cerutti R, Vigetti D, et al. Histidine decarboxylase,
224. Viale G, Doglioni C, Gambacorta M, et al. Progesterone DOPA decarboxylase, and vesicular monoamine transporter
receptor immunoreactivity in pancreatic endocrine tumors. 2 expression in neuroendocrine tumors: immunohistochemical
An immunocytochemical study of 156 neuroendocrine tumors study and gene expression analysis. J Histochem Cytochem.
of the pancreas, gastrointestinal and respiratory tracts, 2006;54:863–875.
and skin. Cancer. 1992;70:2268–2277. 244. Eissele R, Anlauf M, Schafer MK, et al. Expression of
225. Zhao C, Bratthauer GL, Barner R, et al. Comparative vesicular monoamine transporters in endocrine hyperplasia
analysis of alternative and traditional immunohistochemical and endocrine tumors of the oxyntic stomach. Digestion.
markers for the distinction of ovarian sertoli cell tumor from 1999;60:428–439.
endometrioid tumors and carcinoid tumor: A study of 245. Jakobsen AM, Andersson P, Saglik G, et al. Differential
160 cases. Am J Surg Pathol. 2007;31:255–266. expression of vesicular monoamine transporter (VMAT) 1
226. Sica G, Wagner PL, Altorki N, et al. Immunohistochemical and 2 in gastrointestinal endocrine tumours. J Pathol.
expression of estrogen and progesterone receptors in primary 2001;195:463–472.
pulmonary neuroendocrine tumors. Arch Pathol Lab Med. 246. Feng H, Shuda M, Chang Y, et al. Clonal integration of a
2008;132:1889–1895. polyomavirus in human Merkel cell carcinoma. Science.
227. Sobin LH, Hjermstad BM, Sesterhenn IA, et al. Prostatic acid 2008;319:1096–1100.
phosphatase activity in carcinoid tumors. Cancer. 1986;58: 247. Shuda M, Feng H, Kwun HJ, et al. T antigen mutations are a
136–138. human tumor-specific signature for Merkel cell polyomavirus.
228. Azumi N, Traweek ST, Battifora H. Prostatic acid phospha- Proc Natl Acad Sci USA. 2008;105:16272–16277.
tase in carcinoid tumors. Immunohistochemical and immu- 248. Garneski KM, Warcola AH, Feng Q, et al. Merkel cell
noblot studies. Am J Surg Pathol. 1991;15:785–790. polyomavirus is more frequently present in North American
229. Choe BK, Pontes EJ, Rose NR, et al. Expression of human than Australian Merkel cell carcinoma tumors. J Invest
prostatic acid phosphatase in a pancreatic islet cell carcinoma. Dermatol. 2009;129:246–248.
Invest Urol. 1978;15:312–318. 249. Shuda M, Arora R, Kwun HJ, et al. Human Merkel cell
230. Jobsis AC, De Vries GP, Meijer AE, et al. The immunohis- polyomavirus infection I. MCV T antigen expression in
tochemical detection of prostatic acid phosphatase: its Merkel cell carcinoma, lymphoid tissues and lymphoid
possibilities and limitations in tumour histochemistry. Histo- tumors. Int J Cancer. 2009;125:1243–1249.
chem J. 1981;13:961–973. 250. Busam KJ, Jungbluth AA, Rekthman N, et al. Merkel cell
231. Kimura N, Sasano N, Namiki T. Evidence of hybrid cell of polyomavirus expression in merkel cell carcinomas and its
thyroid follicular cell and carcinoid cell in strumal carcinoid. absence in combined tumors and pulmonary neuroendocrine
Int J Gynecol Pathol. 1986;5:269–277. carcinomas. Am J Surg Pathol. 2009;33:1378–1385.
232. Stagno PA, Petras RE, Hart WR. Strumal carcinoids of the 251. Reisinger DM, Shiffer JD, Cognetta AB Jr, et al. Lack of
ovary. An immunohistologic and ultrastructural study. Arch evidence for basal or squamous cell carcinoma infection with
Pathol Lab Med. 1987;111:440–446. Merkel cell polyomavirus in immunocompetent patients
233. Sidhu J, Sanchez RL. Prostatic acid phosphatase in strumal with Merkel cell carcinoma. J Am Acad Dermatol. 2010;
carcinoids of the ovary. An immunohistochemical study. 63:400–403.
Cancer. 1993;72:1673–1678. 252. Kuwamoto S, Higaki H, Kanai K, et al. Association of Merkel
234. Lundqvist M, Wilander E. Subepithelial neuroendocrine cells cell polyomavirus infection with morphologic differences in
and carcinoid tumours of the human small intestine and Merkel cell carcinoma. Hum Pathol. 2011;42:632–640.
appendix. A comparative immunohistochemical study with 253. Paik JY, Hall G, Clarkson A, et al. Immunohistochemistry
regard to serotonin, neuron-specific enolase and S-100 protein for Merkel cell polyomavirus is highly specific but not
reactivity. J Pathol. 1986;148:141–147. sensitive for the diagnosis of Merkel cell carcinoma in the
235. Moyana TN, Satkunam N. A comparative immunohistochem- Australian population. Hum Pathol. 2011;42:1385–1390.
ical study of jejunoileal and appendiceal carcinoids. Implications 254. Jung HS, Choi YL, Choi JS, et al. Detection of Merkel cell
for histogenesis and pathogenesis. Cancer. 1992;70:1081–1088. polyomavirus in Merkel cell carcinomas and small cell
236. Barbareschi M, Frigo B, Mosca L, et al. Bronchial carcinoids carcinomas by PCR and immunohistochemistry. Histol
with S-100 positive sustentacular cells. A comparative study Histopathol. 2011;26:1231–1241.
with gastrointestinal carcinoids, pheochromocytomas and 255. Ly TY, Walsh NM, Pasternak S. The spectrum of Merkel cell
paragangliomas. Pathol Res Pract. 1990;186:212–222. polyomavirus expression in Merkel cell carcinoma, in a
237. Tsuta K, Raso MG, Kalhor N, et al. Sox10-positive variety of cutaneous neoplasms, and in neuroendocrine
sustentacular cells in neuroendocrine carcinoma of the lung. carcinomas from different anatomical sites. Hum Pathol.
Histopathology. 2011;58:276–285. 2012;43:557–566.
238. Al-Khafaji B, Noffsinger AE, Miller MA. Immunohistologic 256. Rodig SJ, Cheng J, Wardzala J, et al. Improved detection
analysis of gastrointestinal and pulmonary carcinoid tumors. suggests all Merkel cell carcinomas harbor Merkel polyoma-
Hum Pathol. 1998;29:992–999. virus. J Clin Invest. 2012;122:4645–4653.

r 2013 Lippincott Williams & Wilkins www.anatomicpathology.com | 313


Bellizzi Adv Anat Pathol  Volume 20, Number 5, September 2013

257. Johansson L, Tennvall J, Akerman M. Immunohistochemical 261. Shy SW, Lee WH, Chou MC, et al. Small cell lung carcinoma:
examination of 25 cases of Merkel cell carcinoma: a clinicopathological, immunohistochemical, and ultrastructural
comparison with small cell carcinoma of the lung and study. J Surg Oncol. 1990;45:146–161.
oesophagus, and a review of the literature. APMIS. 1990;98: 262. Gaffey MJ, Mills SE, Lack EE. Neuroendocrine carcinoma of
741–752. the colon and rectum. A clinicopathologic, ultrastructural,
258. Shah IA, Netto D, Schlageter MO, et al. Neurofilament and immunohistochemical study of 24 cases. Am J Surg
immunoreactivity in Merkel-cell tumors: a differentiating Pathol. 1990;14:1010–1023.
feature from small-cell carcinoma. Mod Pathol. 1993;6:3–9. 263. Zamboni G, Franzin G, Bonetti F, et al. Small-cell neuro-
259. Lehto VP, Stenman S, Miettinen M, et al. Expression of a endocrine carcinoma of the ampullary region. A clinicopa-
neural type of intermediate filament as a distinguishing thologic, immunohistochemical, and ultrastructural study of
feature between oat cell carcinoma and other lung cancers. three cases. Am J Surg Pathol. 1990;14:703–713.
Am J Pathol. 1983;110:113–118. 264. Basturk O, Yang Z, Tang LH, et al. Increased (> 20%) Ki67
260. van Muijen GN, Ruiter DJ, van Leeuwen C, et al. proliferation index in morphologically well differentiated pan-
Cytokeratin and neurofilament in lung carcinomas. Am J creatic neuroendocrine tumors (PanNETs) correlates with
Pathol. 1984;116:363–369. decreased overall survival. Mod Pathol. 2013;26(suppl 2S):422A.

314 | www.anatomicpathology.com r 2013 Lippincott Williams & Wilkins

You might also like