2016 - Algorithmic Approach To Neuroendocrine Tumors - Duan - 2016

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Review Article

Algorithmic Approach to Neuroendocrine Tumors in Targeted


Biopsies: Practical Applications of Immunohistochemical Markers
Kai Duan, MD1,2 and Ozgur Mete, MD, FRCPC1,2,3

Neuroendocrine tumors (NETs) constitute a heterogeneous group of neoplasms with distinct biological behaviors, depending on the site
of origin and the degree of tumor proliferation. Although advances in biochemical and radiological modalities have enhanced the ability to
detect NETs, tissue diagnosis remains the gold standard to assess tumor characteristics for treatment decision making. In an era with
growing demands for precision diagnostics based on smaller tissue samples, immunohistochemistry has become an indispensable tool in
the pathologist’s repertoire. In conjunction with clinical findings and cytomorphology, complementary use of 1) markers of neuroendocrine
differentiation, 2) markers confirming epithelial nature, 3) markers of cellular proliferation, 4) transcription factors and hormonal markers,
as well as 5) predictive and prognostic markers may be necessary to guide patient management in NETs. The current review summarizes
common applications of these immunohistochemical markers when confronted with a potential neuroendocrine neoplasm, and proposes a
stepwise algorithmic approach to avoid diagnostic errors in targeted biopsies. Cancer Cytopathol 2016;124:871-84. V
C 2016 American Can-

cer Society.

KEY WORDS: biomarkers; hormones; immunohistochemistry; neuroendocrine tumor; paraganglioma; poorly differentiated neuroendocrine
carcinoma; transcription factors.

INTRODUCTION
Neuroendocrine tumors (NETs) encompass a wide spectrum of neoplasms arising from various anatomic locations, with diverse
biological behaviors.1–6 Despite advances in diagnostic imaging and biochemical techniques, some patients still present with
unknown primary site prior to the pathological examination.1–3,7–9 Accurate detection and subtyping are critical for patient man-
agement and can be challenging on small biopsies.5,10,11 In particular, assessment of tumor proliferation and confirmation of the
primary site are frequently requested, given their treatment implications.1–3,7,8 In an era with increasing demands for precise
diagnosis based on smaller tissue samples, immunohistochemistry has become an indispensable adjunct tool to accurately classify
these tumors.2,4,7–9 The current review summarizes common applications of biomarkers through an algorithmic approach to
diagnose NETs in targeted biopsies (Table 1).

Confirmation of Neuroendocrine Differentiation


The diagnosis of a suspected neuroendocrine neoplasm typically begins by confirming neuroendocrine differentiation, using a
panel of general neuroendocrine immunohistochemical markers.1,7,8 In the setting of a metastatic tumor of unknown primary,
this may be part of a wider immunohistochemical panel to identify the site of origin.2,12,13 Although several biomarkers (includ-
ing CD56, CD57, protein gene product 9.5 [PGP9.5], and neuron-specific enolase) have been described to date, the most
widely used and reliable neuroendocrine markers are chromogranin A and synaptophysin (Figs. 1 and 2).1,2,14–16 While chromo-
granin A is the most specific marker, synaptophysin is often regarded as the most sensitive marker in the diagnosis of
Corresponding author: Ozgur Mete, MD, FRCPC, Department of Pathology, University Health Network, 200 Elizabeth St, 11th Fl, Toronto, ON M5G 2C4, Canada; Fax:
(416) 340-5517; ozgur.mete2@uhn.ca

We thank Dr. Gilda Santos (Department of Laboratory Medicine Program, University Health Network) for providing some of the photomicrographs used in Figures 1
and 2.
1
Department of Pathology, University Health Network, Toronto, Ontario, Canada; 2Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto,
Ontario, Canada; 3Endocrine Oncology Site Group, Princess Margaret Cancer Centre, Toronto, Ontario, Canada

Received: June 19, 2016; Accepted: June 27, 2016

Published online August 16, 2016 in Wiley Online Library (wileyonlinelibrary.com)

DOI: 10.1002/cncy.21765, wileyonlinelibrary.com

Cancer Cytopathology December 2016 871


Review Article

TABLE 1. Overview of Immunohistochemical Markers issue is of clinical significance as most laboratories do not have
for NETs
access to chromogranin B immunohistochemistry. In light of
1) Markers for neuroendocrine differentiation these findings, the concurrent use of chromogranin A and syn-
Chromogranin A (most specific; recommended)
Synaptophysin (most sensitive; recommended) aptophysin is recommended to accurately confirm or exclude
CD56, CD57, PGP9.5, and neuron-specific enolase (less specific markers, neuroendocrine differentiation in a suspected well-
not recommended for well-differentiated NETs)
2) Markers for epithelial differentiation differentiated NET. In addition, it should be noted that some
Cytokeratins (AE1/AE3 and CAM5.2) NETs can be seen in the context of mixed tumors (ie, mixed
3) Marker for tumor proliferation
Ki-67/MIB-1 adeno-NEC), whereas other tumors may demonstrate neuro-
4) Markers for site of origin
Pituitary: Pit-1, T-pit, and SF-1
endocrine differentiation (ie, adenocarcinoma with focal diver-
Parathyroid: PTH, GCM-2, and GATA-3 gent neuroendocrine differentiation); the distinction of these
Medullary thyroid carcinoma: TTF-1, monoclonal CEA (diffuse), and calcito-
nin/calcitonin-gene related peptide
entities can be difficult in small biopsy samples.1,22–24 How-
Pulmonary: TTF-1, calcitonin/calcitonin-gene related peptide, bombesin, and ever, the extent of neuroendocrine marker staining in adeno-
serotonin
Midgut: CDX-2 (strong and diffuse) and serotonin carcinoma with divergent neuroendocrine differentiation is
Pancreatic: ISL-1, PDX-1, and CDX-2 (6)a generally <30% of the tumor volume.25,26 Therefore, quanti-
Duodenal: ISL-1, PDX-1, xenin, and CDX-2 (6)a
Hindgut/rectal: PAP, pancreatic polypeptide, glucagon-like peptide 1, peptide fication of neuroendocrine marker staining is pertinent,
YY, ISL-1 (6), CDX-2 (6),a and serotonin (6) although it may not reflect the characteristics of the overall
Paraganglioma: tyrosine hydroxylase and GATA-3
5) Markers with potential prognostic or therapeutic implications tumor in a cytology specimen. Accurate recognition of these
SSTRs 1-5
Hormones
diagnostic pitfalls, along with strict correlation of cytomor-
E-cadherin and b-catenin phology and immunohistochemical findings, is crucial to pre-
Pancreatic NETs: DAXX, ATRX, menin, CEACAM1, CK19, CD117, PTEN, PR,
cyclooxygenase-2, p21, p18, and Rb
vent misdiagnosis.
SDHB immunohistochemistry
Mismatch repair proteins (MLH1, MSH2, MSH6, and PMS2)
Confirmation of Epithelial Nature
Abbreviations: 6 , variable immunohistochemical expression; ATRX, ATP-
dependent helicase ATRX, X-linked helicase II; CDX-2, caudal type homeobox After confirmation of neuroendocrine differentiation, a panel
2; CEA, carcinoembryonic antigen; CEACAM1, carcinoembryonic antigen-
related cell adhesion molecule 1; CK19, cytokeratin 19; DAXX, death domain-
of general epithelial biomarkers (cytokeratins AE1/AE3,
associated protein; GATA-3, GATA-binding protein 3; GCM-2, glial cell missing 2 CAM5.2, and/or CK18) is recommended to exclude a poten-
protein; ISL-1, insulin gene enhancer protein; MLH1, MutL homolog 1; MSH2,
MutS protein homolog 2; MSH6, MutS protein homolog 6; NET, neuroendocrine tial paraganglioma (Figs. 1 and 3).1,8,16,27 This distinction is
tumor; PAP, prostatic acid phosphatase; PDX-1, pancreatic and duodenal crucial as the management of paraganglioma can differ from
homeobox 1; PGP.9.5, protein gene product 9.5; Pit-1, pituitary-specific
transcription factor 1; PMS2, PMS1 homolog 2; PR, progesterone receptor; that of other NETs.1,10,16,27,28 Although a small percentage of
PTEN, phosphatase and tensin homolog; PTH, parathyroid hormone; SDHB,
succinate dehydrogenase B; SF-1, Steroidogenic factor 1; SSTRs, somatostatin
NETs may stain negative for epithelial markers, a paragan-
receptors; TTF-1, thyroid transcription factor-1. glioma should always be excluded in a cytokeratin-negative
a
Although well-differentiated neuroendocrine tumors (WDNETs) from the pan-
creas, foregut, and hindgut can sometimes express CDX-2 positivity, the stain- NET.1,16 Paragangliomas (including pheochromocytomas,
ing pattern is usually weak and patchy compared with the strong and diffuse which are intra-adrenal sympathetic paragangliomas) are
staining noted in midgut enterochromaffin cell WDNETs.
“specialized” neuroendocrine neoplasms arising from neural
crest-driven endocrine organs (known as paraganglia) that pro-
well-differentiated NETs.17,18 It should be noted that the level duce catecholamines (epinephrine, norepinephrine, and dopa-
of chromogranin A expression may vary according to the mine).1,27,28 Recent evidence suggests that up to 40% of these
degree of tumor differentiation (Fig. 2).2 Thus, focal, weak, or neoplasms are associated with genetic susceptibility, with clini-
even absent chromogranin A staining may be noted in poorly cal implications for genetic testing.16,27,29–34 Although lack of
differentiated neuroendocrine carcinomas (NECs) (Fig. 2); immunohistochemical expression of transcription factors (ie,
however, synaptophysin should still be present in these cases. thyroid transcription factor 1 [TTF-1], caudal type homeobox
Nonetheless, it is important to note that positivity for synapto- 2 [CDX-2], paired box 8 [PAX-8], islet-1, pancreatic and
physin alone does not warrant a diagnosis of NET in all cases. duodenal homeobox 1 [PDX-1]) supports a diagnosis of a
For example, solid pseudopapillary tumor of the pancreas and paraganglioma in a cytokeratin-negative NET, this finding
adrenal cortical carcinoma can also express synaptophy- should be corroborated with positivity for tyrosine hydroxy-
sin.1,19,20 On another note, some NETs, including the major- lase, which is the rate-limiting enzyme in catecholamine syn-
ity of L-cell NETs of hindgut origin, may preferentially thesis (Fig. 3).1,27,29,30 Demonstration of S100-positive
express chromogranin B instead of chromogranin A.1,21 This sustentacular network also favors a diagnosis of paraganglioma

872 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

Figure 1. Well-differentiated neuroendocrine tumor (A: Giemsa stain) with (B) chromogranin A, (C) synaptophysin, and (D) AE1/AE3
positivity.

in surgical specimens; however, S100-positive sustentacular protein [GCM-2], etc), hormones (ie, parathyroid hormone
cells can be identified in other NETs, including pituitary [PTH] and monoclonal calcitonin) and monoclonal carcino-
adenomas, gastroenteropancreatic and pulmonary embryonic antigen (CEA) may also be required to render a
1,16,35,36
NETs. Therefore, the identification of sustentacular diagnosis of GATA-3-positive and tyrosine hydroxylase-
cells is not helpful in this distinction. The real challenge arises negative paraganglioma in the head and neck region.1,16
when confronted with a cytokeratin-negative neuroendocrine Exceptional examples of paragangliomas, namely cauda equina
neoplasm that is also negative for tyrosine hydroxylase. This paragangliomas and gangliocytic paragangliomas (comprised
can occur in a subset of nonfunctioning paragangliomas which of a mixture of epithelioid neuroendocrine cells, Schwann-like
are mostly segregated in the head and neck region.1,16,30 cells, and scattered ganglion-like cells) may also demonstrate
Although some head and neck paragangliomas can still dem- positivity for cytokeratin, a phenomenon believed to be related
onstrate positivity for tyrosine hydroxylase, the majority will to divergent epithelial differentiation.1,42
have focal-to-weak or absent staining for tyrosine hydroxy-
Assessment of Tumor Proliferation
lase.1,16,30 In this specific context (keratin-negative and
tyrosine hydroxylase-negative NET), positivity for trans-acting After confirmation of neuroendocrine and epithelial differen-
T-cell-specific transcription factor (GATA-binding protein 3 tiation, the next step pertains to differentiating a well-
[GATA-3]) may help to support a diagnosis of paraganglioma differentiated NET (WDNET) from a poorly differentiated
(Fig. 3); however, it should be noted that GATA-3 is also NEC.1,7,8,10 While tumor cytomorphology, including
expressed in nonendocrine tumors as well as parathyroid neo- mitotic activity, may be sufficient in making this distinction,
plasms (which are positive for neuroendocrine the concurrent use of a proliferative biomarker (Ki-67/MIB-
markers).1,16,30,37–41 Ultimately, absent staining for relevant 1) is generally advised.1,5,7,8,43–46 The classification of NET
transcription factors (ie, TTF-1, CDX-2, glial cell missing 2 is site- and grade-dependent, requiring careful evaluation of

Cancer Cytopathology December 2016 873


Review Article

Figure 2. Poorly differentiated neuroendocrine carcinoma (NEC) of small cell type. In this composite figure, pitfalls associated with
poorly differentiated NECs are illustrated. The fine-needle aspiration biopsy identified a small cell NEC (A: Papanicolaou stain). (B)
Unlike well-differentiated neuroendocrine tumors, positivity for chromogranin A can be weak or even absent. Dot-like paranuclear
chromogranin-A staining can be identified in some cases. Positivity for synaptophysin and (C) CD56 often remain in poorly differenti-
ated NECs. (D) High proliferative activity is often reflected in high Ki-67 labeling.

cytomorphology along with proliferative features of the Determination of the Site of Origin
tumor.1,5,7,8,43–46 Although the most recent World Health Given the treatment implications, a diagnosis of well-
Organization classification still does not recognize the impact differentiated NET may require further immunohistochemical
of the Ki-67 index in the grading of mediastinal NETs, the workup to establish the site of origin, particularly if this infor-
distinction between a poorly differentiated NEC (ie, small mation is ambiguous to the treating clinician.1,7,8,53 In con-
cell NEC, large cell NEC) and a WDNET is usually not a trast, determination of the primary site in poorly
major challenge in cytology specimens.1,5,7,8,43–46 Nonethe- differentiated NEC is often not requested, given the uniform-
less, the use of Ki-67 may still be helpful (Fig. 2).45 The real ity in treatment modalities and disease behavior.1,5,7,10,54 In
challenge arises when distinguishing between low-grade ver- combination with clinical and cytomorphological findings, a
sus intermediate-grade NETs. The proliferation index in panel of markers for transcription factors and hormones offers
fine-needle aspiration cell blocks should be interpreted with the highest sensitivity and specificity to identify the site of ori-
caution.43,44,47–52 Because neuroendocrine neoplasms can be gin.1,2,7,8,17 A detailed description of each marker is beyond
quite heterogeneous with respect to their proliferative activ- the scope of this paper, and has been reported elsewhere.1,8
ity, small biopsy specimens often fail to meet the minimum For brevity, we will review common applications of immuno-
requirements for formal grading.43,44,47–52 The reported histochemistry, pertaining to frequent sites where NETs can
concordance between endoscopic ultrasound-guided fine- arise (head and neck, thorax, pancreas, and various parts of the
needle aspiration cytology versus surgical pathology in NET gastrointestinal tract). In the setting of a metastatic WDNET
grading (using the Ki-67 index) varies from 50% to of unknown primary, we recommend a panel approach using
89.5%.43,44,47–52 multiple immunohistochemical markers simultaneously.1,8,55

874 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

Figure 3. Keratin-negative neuroendocrine tumor should alert the diagnostician to the possibility of a paraganglioma. A young patient
presented with an enlarged neck mass. The fine-needle aspiration and synchronous core needle biopsy specimens demonstrated a
neuroendocrine neoplasm (A: core needle biopsy; H & E). Immunohistochemically, the tumor cells were positive for (B) chromogranin
A and (C) synaptophysin, but were negative for (D) AE1/AE3 and CAM5.2 (not shown). The initial panel raised the possibility of a par-
aganglioma; (E) positivity for tyrosine hydroxylase confirmed the diagnosis of paraganglioma. (F) GATA-binding protein 3 (GATA-3)
was also expressed in the tumor cells. There was no loss of succinate dehydrogenase B (SDHB) expression to suggest succinate dehy-
drogenase (SDHx)-related pathogenesis (not shown).

Extra-abdominal locations calcitonin gene-related peptide), as well as other hormones


As a general rule, positivity for TTF-1 in a WDNET favors a (including bombesin and serotonin).1,8,58 Therefore, concur-
primary site from either the head and neck (specifically medul- rent staining for monoclonal CEA is recommended. While
lary thyroid carcinoma [MTC]) or the thorax (specifically pul- rare pulmonary NETs have been reported with variable CEA
monary NET) (Figs. 4 and 5).1,8,56,57 Although positivity for positivity, diffuse CEA positivity is consistent with the diagno-
calcitonin is often used to support a diagnosis of MTC, pul- sis of MTC in a TTF-1-positive and calcitonin-positive
monary NETs can also produce calcitonin (more precisely WDNET.1 Alternatively, positivity for bombesin supports a

Cancer Cytopathology December 2016 875


Review Article

Figure 4. Medullary thyroid carcinoma. The fine-needle aspiration biopsy from a lateral neck mass identified neuroendocrine tumor
cells with eccentric nuclei and a plasmacytoid appearance (A: Giemsa stain and B: Papanicolaou stain). The cell block was subjected
to a panel of markers. The tumor cells were diffusely positive for (C) chromogranin A, (D) thyroid transcription factor 1 (TTF-1), (E)
calcitonin, and (F) carcinoembryonic antigen. The cytomorphology and immunohistochemical findings were diagnostic of medullary
thyroid carcinoma.

diagnosis of pulmonary NET in the appropriate setting, particular, TTF-1 is commonly expressed in poorly differenti-
although most laboratories do not have access to this test.1,58 ated NECs regardless of the site of origin.1,8,60 However, in
In cases where there is a need to distinguish between a pulmo- some cases, the pathologist may be asked to exclude a Merkel
nary versus thymic NET, positivity for TTF-1 favors a pulmo- cell carcinoma from a metastatic NEC in the skin, given the
nary origin.59 different treatment implications.1,7,8,61,62 In such cases, para-
At this point, it is important to emphasize that transcrip- nuclear dot-like positivity for CK20, Merkel cell polyoma
tion factors often lack lineage specificity in poorly differenti- virus, and negativity for TTF-1 suggest the possibility of a
ated NECs, which can be a source of diagnostic error.1,8 In Merkel cell carcinoma.1,7,8,61,62

876 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

Figure 5. Pulmonary neuroendocrine tumor (NET). The fine-needle aspiration biopsy from the lung nodule identified a spindle cell
NET (A: Giemsa stain and B: cell block; H & E). The tumor cells were positive for (C) chromogranin A, (D) thyroid transcription factor 1
(TTF-1), and (E) CD56, and were negative for (F) carcinoembryonic antigen. The overall features were those of a pulmonary NET.

Various other NETs can arise in the head and neck if a pituitary adenoma is suspected, positivity for adenohypo-
region, including pituitary and parathyroid neoplasms.1,63,64 physeal cell lineage transcription factors (pituitary-specific
As mentioned previously, paragangliomas can be found in this transcription factor 1 [Pit-1], T-pit, and steroidogenic factor 1
location (ie, carotid body paragangliomas, thyroid paragan- [SF-1]) is helpful in confirming the adenohypophyseal cell ori-
gliomas, laryngeal paragangliomas) and should be excluded gin.68,69 Parathyroid neoplasms can sometimes present as a
using epithelial markers and/or tyrosine hydroxylase. While mass in the neck or mediastinum, or even as an intrathyroidal
pituitary adenomas tend to arise in the sella turcica region, mass.40,41,70,71 Parathyroid tumors express neuroendocrine
they can invade into adjacent structures or present at ectopic and epithelial markers as well as PTH, although the latter can
locations, including the nasopharynx, sphenoid wing, sphe- occasionally be expressed in some other NETs.40,41,72–75 Con-
noid sinus, nasal cavity, and temporal bone.1,65–67 Therefore, sequently, positivity for GCM-2 and GATA-3 is helpful in

Cancer Cytopathology December 2016 877


Review Article

Figure 6. Metastatic enterochromaffin cell neuroendocrine tumor (NET) of midgut origin. The fine-needle aspiration and core needle
biopsy specimens from the liver lesions identified a well-differentiated NET. Diffuse positivity for (A) caudal type homeobox 2 (CDX-
2) and (B) serotonin suggested an enterochromaffin cell NET of midgut origin. Subsequent clinical and imaging workup confirmed the
presence of a primary NET of the small bowel.

the confirmation of parathyroid origin in a PTH-positive serotonin and CDX-2 is a characteristic feature of an entero-
NET.1,39–41,76 chromaffin cell NET of midgut origin (Fig. 6).97 Although
WDNETs from the pancreas, foregut, and hindgut can also
Intra-abdominal locations express CDX-2 positivity, the staining pattern is usually weak
The intra-abdominal cavity is a common source of NETs, and patchy compared with the strong and diffuse staining
which can arise from the pancreas and various gastrointestinal observed in midgut WDNETs.1,8,17,89–96 Therefore, assess-
sites.1,7,17,77–79 In particular, primary pancreatic NETs are ment for both extent and intensity of CDX-2 staining, along
important to recognize because they can be treated with newer with other hormones, increases the likelihood of a primary
therapy regimens (including sunitinib and everolimus) and are midgut origin.1,8,17,89–96 Furthermore, it should be noted that
more sensitive to chemotherapy than NETs arising from other recent evidence suggests that the midgut (in particular the
sites.7,80–82 Furthermore, midgut NETs may be amenable to ileum) is the most common primary site for metastatic
octreotide therapy, with promising outcomes reported in a WDNETs of unknown primary.1,7,9,79
subset of patients with advanced disease.7,83,84 In addition, In contrast, an immunohistochemical panel demonstrat-
identification of primary small-bowel NETs (even in the pres- ing TTF-1 negativity, negative or patchy or weak staining for
ence of metastatic disease) may result in surgical intervention CDX-2, ISL-1 positivity, and PDX-1 positivity suggests a
to prevent further complications, including bowel obstruction, WDNET originating from the pancreas or duodenum.1,7,8
ischemia, and perforation.1,7,79,85,86 Although the liver is a ISL-1 or Islet-1 is a transcription factor implicated in the dif-
common site of metastasis for NETs, it is important to note ferentiation of neuroendocrine pancreatic cells, although it can
that primary paraganglioma of the liver does exist.87,88 There- be found in chromaffin cells, calcitonin-producing parafollicu-
fore, the identification of a NET in the liver is not always a lar C cells, as well as the anterior/intermediate pituitary lobes
sign of metastatic disease. Furthermore, the presence of a and some neurons involved in autonomic and endocrine con-
TTF-1-positive WDNET in the liver warrants further immu- trol.1,8,98–102 In addition to pancreatic NETs, ISL-1 may be
nohistochemistry, with calcitonin and monoclonal CEA, to expressed in NETs of duodenal and colorectal origin.1,8,102,103
distinguish between a metastatic pulmonary NET versus a When distinguishing between a pancreatic/duodenal
metastatic MTC. WDNET versus a colorectal WDNET, concurrent positivity
An immunohistochemical panel showing TTF-1 nega- for PDX-1 is helpful to support a pancreatic/duodenal origin.
tivity, strong and diffuse staining for CDX-2, insulin gene As its name implies, PDX-1 is a transcription factor impli-
enhancer protein (ISL-1) negativity, and PDX-1-negativity in cated in the development of the pancreas and duode-
a WDNET favors a midgut origin (usually jejunoileal or num.1,8,104–107 Alternatively, demonstration of positivity for
appendiceal).1,8,17,89–96 CDX-2 is a nuclear transcription fac- prostatic acid phosphatase suggests a hindgut origin (usually
tor found in gastrointestinal epithelial cells distal to the stom- rectal NET; 82% positivity), with infrequent reports at non-
ach.1,8,17,89–96 In particular, diffuse positivity for both rectal sites (small bowel NETs; 20% positivity).1,8,77,108,109

878 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

Positivity for ISL-1 and CDX-2 (usually patchy/weak positiv- potential circulating hormones to monitor for disease progres-
ity) is also described in NETs of the hindgut.1,8,77,102 In a sion biochemically.1,75,121,123,133
ISL-1-positive and PDX-1-positive WDNET, the distinction In patients with pulmonary and gastrointestinal NETs,
between a pancreatic versus a duodenal WDNET can be chal- loss of E-cadherin and b-catenin expression appears to be asso-
lenging. Immunohistochemical positivity for xenin, a hor- ciated with higher tumor grade.134,135 In those with pancreatic
mone peptide found in endocrine cells of the duodenum, may NETs, immunohistochemical positivity for CEA-related cell
be helpful to support a duodenal origin; however, this marker adhesion molecule 1 (CEACAM1), cytokeratin 19 (CK19),
is not available in the majority of laboratories.1,78,110 Although and CD117 (C-kit) may be related to more aggressive behav-
PAX-8 is a nuclear transcription factor implicated in the devel- ior and worse clinical outcomes.93,120,136–138 A recent study
opment of the optic, thyroid, urinary, and reproductive sys- also revealed that pancreatic NETs commonly harbor muta-
tems, it has also been shown to have strong tions in multiple endocrine neoplasia type 1 [MEN1] (44%),
immunohistochemical expression in normal human islets and DAXX/ATRX (death domain-associated proteins; 43%), and
pancreatic NETs.1,8,111–119 However, it should be noted that mechanistic target of rapamycin (mTOR) pathway genes
these findings were primarily based on a commercially avail- (14%).139 Subsequent studies reported that loss of DAXX,
able polyclonal PAX-8 antibody that also identifies other ATRX, and phosphatase and tensin homolog (PTEN) expres-
members of the PAX family (including PAX-3, PAX-5, and sion are associated with worse survival in patients with pancre-
PAX-6).1,8,111–119 In other words, it is likely that polyclonal atic NETs.17,120,140,141 Negative progesterone receptor
PAX-8 is cross-reacting with PAX-6 (which is highly expressed expression may be associated with a worse prognosis.17,141
in pancreatic islet cells), because PAX-8 messenger RNA Expression of cyclooxygenase-2, p21, p18, and Rb have been
(mRNA) expression is generally low in islet cells.1,8,111–119, correlated with poorer survival in patients with gastroentero-
Similar to xenin and prostatic acid phosphatase, other pancreatic NETs.17,132,142 In patients with hindgut NETs, the
hormonal markers may also help locate WDNETs within the distinction of L-cell phenotype from enterochromaffin cell ori-
gastrointestinal tract.1,8,17 WDNETs of the foregut and gin using hormonal markers (glucagon-like peptide and pep-
midgut often produce serotonin and substance P, whereas tide YY) also appears to have prognostic implications.143,144
WDNETs of hindgut origin tend to produce the peptide Various immunohistochemical markers associated with
spectrum of L cells, including glucagon-like peptide 1, pancre- genetic susceptibility have been reported in neuroendocrine
atic polypeptide, and peptide YY.1,17,77,97 neoplasms. For instance, loss of p27 may suggest an underly-
ing germline CDKN1B mutation associated with multiple
endocrine neoplasia type 4 (MEN4) syndrome.40,145–149
Role of Predictive and Prognostic Biomarkers
However, one should also be aware of the relative frequency of
In addition to the primary site and proliferation index of a CDKN1B/p27 aberrancies in small bowel NETs, which can
NET, several biomarkers with potential predictive and/or lead to multifocal disease independent of MEN4 syn-
prognostic usefulness have been described.1,8,75,120–127 The drome.150 Although loss of menin (the gene product of
majority of these assessments are performed on surgical speci- MEN1) in pancreatic NETs may provide prognostic informa-
mens. However, select markers may be considered on biopsy tion, it can help to select some patients for genetic testing of
specimens upon clinical request in specific presentations. MEN1 syndrome.139,145,148,151,152 However, it should be
Although controversial, the detection and subtyping of noted that a significant number of NETs harbor somatic and
somatostatin receptors (SSTRs 1-5) by immunohistochemistry epigenetic alterations leading to reduced menin expression.139
is advocated to select patients for somatostatin analog therapy In addition, extracolonic manifestations of Lynch syndrome
and targeted peptide receptor radiotherapy.122,124–131 In addi- have also been expanded with the inclusion of various
tion, loss of expression for SSTR-2 (one of the common targets NETs.153–155 Therefore, testing for mismatch repair proteins
of octreotide) correlates with poorer survival in patients with is informative in the appropriate clinical context.
gastroenteropancreatic NETs.17,124–128,132 In patients with In patients with paragangliomas/pheochromocytomas,
symptomatic NETs, tumor positivity for hormonal markers loss of succinate dehydrogenase B (SDHB) expression is con-
may be of clinical use because those who develop secondary sidered a harbinger of familial disease associated with any
endocrine syndromes tend to have increased morbidity and SDH mutations.16,27,29–34,156,157 Furthermore, loss of SDHB
mortality; furthermore, this approach may help identify also provides prognostic data given the high frequency of

Cancer Cytopathology December 2016 879


Review Article

Figure 7. Proposed algorithmic approach to neuroendocrine tumors (NETs) in targeted biopsies. -, negative immunohistochemical
expression; 1 , positive immunohistochemical expression; CDX-2, caudal type homeobox 2; CEA, carcinoembryonic antigen; ISL-1, insu-
lin gene enhancer protein (Islet-1); MTC, medullary thyroid carcinoma; PDNEC, poorly differentiated neuroendocrine carcinoma; PDX-1,
pancreatic and duodenal homeobox 1; PAP, prostatic acid phosphatase; SDHB, succinate dehydrogenase B; TTF-1, thyroid transcription
factor 1;WDNET, well-differentiated neuroendocrine tumor. *Evaluation of tumor proliferative activity depends on the anatomic site.

malignant disease associated with SDHB-driven paraganglio- applications of immunohistochemical markers) is advised to
mas.16,27,29–34,156–158 Recent advances have also expanded the avoid diagnostic errors (Fig. 7). In patients with a suspected
spectrum of SDH-deficient neoplasms with the inclusion of NET, we recommend to: 1) start by confirming neuroendo-
NETs.159,160 Similar to paragangliomas and pheochromocyto- crine differentiation, using chromogranin A and synaptophy-
mas, the use of SDHB immunohistochemistry may be consid- sin concurrently; 2) consider the diagnostic category of
ered to predict SDH-related pathogenesis in other paraganglioma in a keratin-negative and transcription factor-
neuroendocrine neoplasms.160 negative neuroendocrine neoplasm (positivity for tyrosine
hydroxylase confirms a diagnosis of paraganglioma); and 3)
Conclusions
distinguish a WDNET from a poorly differentiated NEC by
Over the past decade, our understanding of the clinical spec- assessing cytomorphology and/or degree of proliferation, using
trum and pathogenesis of NETs has grown substantially with the Ki-67 labeling index. In a WDNET of unknown primary,
advances in molecular and cellular pathology. As a result, the we suggest an initial panel that combines transcriptional fac-
management of these tumors has become increasingly com- tors (TTF-1, CDX-2, ISL-1, and PDX-1) along with hor-
plex, with greater emphasis on the diagnostician’s role in guid- mone and structural biomarkers as deemed necessary to
ing therapy.161 For instance, the treatment of patients with determine the site of origin; depending on the primary site
advanced WDNETs is increasingly dependent on the site of and clinical indications, additional biomarkers may be per-
origin of the tumor.7,10 However, there are still clinical presen- formed to provide prognostic information to guide therapy
tations in which the primary site remains unknown prior to
and/or screen patients for genetic testing.
the pathological examination (ie, in a biopsy of a liver or
lymph node metastasis).7,8 In conjunction with clinical and
morphological findings, immunohistochemistry has become
an indispensable ancillary tool to help solve this clinical conun- FUNDING SUPPORT
drum. As a general rule, a stepwise algorithmic approach (with No specific funding was disclosed.

880 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

CONFLICT OF INTEREST DISCLOSURES neuroendocrine tumours of the gastrointestinal tract. Virchows Arch.
1995;426:361-367.
The authors made no disclosures. 22. Shinji S, Naito Z, Ishiwata T, et al. Neuroendocrine cell differentiation
of poorly differentiated colorectal adenocarcinoma correlates with liver
metastasis. Int J Oncol. 2006;29:357-364.
REFERENCES
23. Visscher DW, Zarbo RJ, Trojanowski JQ, Sakr W, Crissman JD. Neu-
1. Hafezi-Bakthiari S, Mete O. The approach to metastatic endocrine roendocrine differentiation in poorly differentiated lung carcinomas: a
tumors of unknown primary site. In: Mete O, Asa SL, eds. Endocrine light microscopic and immunohistologic study. Mod Pathol. 1990;3:
Pathology. New York: Cambridge University Press; 2016. 508-512.
2. Kandalaft PL, Gown AM. Practical applications in immunohistochem- 24. Hamilton K, Chiappori A, Olson S, Sawyers J, Johnson D, Washington
istry: carcinomas of unknown primary site. Arch Pathol Lab Med. K. Prevalence and prognostic significance of neuroendocrine cells in
2016;140:508-523. esophageal adenocarcinoma. Mod Pathol. 2000;13:475-481.
3. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: 25. Bosman F, Carneiro F, Hruban R, Theise N. WHO Classification of
epidemiology of and prognostic factors for neuroendocrine tumors in Tumours of the Digestive System. Lyon, France: IARC Press; 2010.
35,825 cases in the United States. J Clin Oncol. 2008;26:3063-3072. 26. Gurzu S, Kadar Z, Bara T, et al. Mixed adenoneuroendocrine carci-
4. Modlin IM, Sandor A. An analysis of 8305 cases of carcinoid tumors. noma of gastrointestinal tract: report of 2 cases. World J Gastroenterol.
Cancer. 1997;79:813-829. 2015;21:1329-1333.
5. Kunz PL, Reidy-Lagunes D, Anthony LB, et al; North American Neu- 27. Mete O, Tischler AS, de Krijger R, et al. Protocol for the examination
roendocrine Tumor Society. Consensus guidelines for the management of specimens from patients with pheochromocytomas and extra-adrenal
and treatment of neuroendocrine tumors. Pancreas. 2013;42:557-577. paragangliomas. Arch Pathol Lab Med. 2014;138:182-188.
6. Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treat- 28. Joynt KE, Moslehi JJ, Baughman KL. Paragangliomas: etiology, presen-
ment of neuroendocrine tumors: consensus report of the National Can- tation, and management. Cardiol Rev. 2009;17:159-164.
cer Institute Neuroendocrine Tumor clinical trials planning meeting. 29. Tischler AS. Pheochromocytoma and extra-adrenal paraganglioma:
J Clin Oncol. 2011;29:934-943. updates. Arch Pathol Lab Med. 2008;132:1272-1284.
7. Bergsland EK, Nakakura EK. Neuroendocrine tumors of unknown pri- 30. Tischler AS, Pacak K, Eisenhofer G. The adrenal medulla and extra-
mary: is the primary site really not known? JAMA Surg. 2014;149:889- adrenal paraganglia: then and now. Endocr Pathol. 2014;25:49-58.
890. 31. King KS, Pacak K. Familial pheochromocytomas and paragangliomas.
8. Bellizzi AM. Assigning site of origin in metastatic neuroendocrine neo- Mol Cell Endocrinol. 2014;386:92-100.
plasms: a clinically significant application of diagnostic immunohisto- 32. Welander J, Soderkvist P, Gimm O. Genetics and clinical characteris-
chemistry. Adv Anat Pathol. 2013;20:285-314. tics of hereditary pheochromocytomas and paragangliomas. Endocr
9. Kirshbom PM, Kherani AR, Onaitis MW, Feldman JM, Tyler DS. Relat Cancer. 2011;18:R253-R276.
Carcinoids of unknown origin: comparative analysis with foregut, 33. Vicha A, Musil Z, Pacak K. Genetics of pheochromocytoma and para-
midgut, and hindgut carcinoids. Surgery. 1998;124:1063-1070. ganglioma syndromes: new advances and future treatment options.
10. Kulke MH, Shah MH, Benson AB, et al; National Comprehensive Curr Opin Endocrinol Diabetes Obes. 2013;20:186-191.
Cancer Network. Neuroendocrine tumors, version 1.2015. J Natl 34. Peczkowska M, Kowalska A, Sygut J, et al. Testing new susceptibility
Compr Canc Netw. 2015;13:78-108. genes in the cohort of apparently sporadic phaeochromocytoma/para-
11. Klimstra DS, Beltran H, Lilenbaum R, Bergsland E. The spectrum of ganglioma patients with clinical characteristics of hereditary syndromes.
neuroendocrine tumors: histologic classification, unique features and Clin Endocrinol (Oxf). 2013;79:817-823.
areas of overlap. Am Soc Clin Oncol Educ Book. 2015:92-103. 35. Gosney JR, Denley H, Resl M. Sustentacular cells in pulmonary neuro-
12. Hillen HF. Unknown primary tumours. Postgrad Med J. 2000;76:690- endocrine tumours. Histopathology. 1999;34:211-215.
693. 36. Hirabayashi K, Zamboni G, Nishi T, Tanaka A, Kajiwara H,
13. Conner JR, Hornick JL. Metastatic carcinoma of unknown primary: Nakamura N. Histopathology of gastrointestinal neuroendocrine neo-
diagnostic approach using immunohistochemistry. Adv Anat Pathol. plasms. Front Oncol. 2013;3:2.
2015;22:149-167. 37. So JS, Epstein JI. GATA3 expression in paragangliomas: a pitfall
14. Wilson BS, Lloyd RV. Detection of chromogranin in neuroendocrine potentially leading to misdiagnosis of urothelial carcinoma. Mod Pathol.
cells with a monoclonal antibody. Am J Pathol. 1984;115:458-468. 2013;26:1365-1370.
15. Wiedenmann B, Franke WW, Kuhn C, Moll R, Gould VE. Synapto- 38. Miettinen M, McCue PA, Sarlomo-Rikala M, et al. GATA3: a multi-
physin: a marker protein for neuroendocrine cells and neoplasms. Proc specific but potentially useful marker in surgical pathology: a systematic
Natl Acad Sci U S A. 1986;83:3500-3504. analysis of 2500 epithelial and nonepithelial tumors. Am J Surg Pathol.
16. Hayashi T, Mete O. Head and neck paragangliomas: what does the 2014;38:13-22.
pathologist need to know? Diagn Histopathol. 2014;20:316-325. 39. Ordonez NG. Value of GATA3 immunostaining in the diagnosis of
17. Kim JY, Hong SM. Recent updates on neuroendocrine tumors from parathyroid tumors. Appl Immunohistochem Mol Morphol. 2014;22:756-
the gastrointestinal and pancreatobiliary tracts. Arch Pathol Lab Med. 761.
2016;140:437-448. 40. Duan K, Gomez Hernandez K, Mete O. Clinicopathological correlates
18. Klimstra DS, Modlin IR, Adsay NV, et al. Pathology reporting of neu- of hyperparathyroidism. J Clin Pathol. 2015;68:771-787.
roendocrine tumors: application of the Delphic consensus process to 41. Duan K, Mete O. Parathyroid carcinoma: diagnosis and clinical impli-
the development of a minimum pathology data set. Am J Surg Pathol. cations. Turk Patoloji Derg. 2015;31(suppl 1):80-97.
2010;34:300-313. 42. Gucer H, Mete O. Endobronchial gangliocytic paraganglioma: not all
19. Notohara K, Hamazaki S, Tsukayama C, et al. Solid-pseudopapillary keratin-positive endobronchial neuroendocrine neoplasms are pulmo-
tumor of the pancreas: immunohistochemical localization of neuroen- nary carcinoids. Endocr Pathol. 2014;25:356-358.
docrine markers and CD10. Am J Surg Pathol. 2000;24:1361-1371. 43. Farrell JM, Pang JC, Kim GE, Tabatabai ZL. Pancreatic neuroendo-
20. Weissferdt A, Phan A, Suster S, Moran CA. Adrenocortical carcinoma: crine tumors: accurate grading with Ki-67 index on fine-needle aspira-
a comprehensive immunohistochemical study of 40 cases. Appl Immu- tion specimens using the WHO 2010/ENETS criteria. Cancer (Cancer
nohistochem Mol Morphol. 2014;22:24-30. Cytopathol). 2014;122:770-778.
21. Fahrenkamp AG, Wibbeke C, Winde G, et al. Immunohistochemical 44. Jin M, Roth R, Gayetsky V, Niederberger N, Lehman A, Wakely PE Jr.
distribution of chromogranins A and B and secretogranin II in Grading pancreatic neuroendocrine neoplasms by Ki-67 staining on

Cancer Cytopathology December 2016 881


Review Article

cytology cell blocks: manual count and digital image analysis of 58 cases. clinicopathologic and immunophenotypic study of 32 cases with a com-
J Am Soc Cytopathol. doi:10.1016/j.jasc.2016.03.002. http://www.science- prehensive review of the English literature. Head Neck Pathol. 2012;6:75-
direct.com/science/article/pii/S2213294516300011. [Epub ahead of print]. 100.
45. Pelosi G, Rindi G, Travis WD, Papotti M. Ki-67 antigen in lung neu- 64. Levy MT, Braun JT, Pennant M, Thompson LD. Primary paragan-
roendocrine tumors: unraveling a role in clinical practice. J Thorac glioma of the parathyroid: a case report and clinicopathologic review.
Oncol. 2014;9:273-284. Head Neck Pathol. 2009;4:37-43.
46. van Velthuysen ML, Groen EJ, van der Noort V, van de Pol A, Tesselaar 65. Rasmussen P, Lindholm J. Ectopic pituitary adenomas. Clin Endocrinol
ME, Korse CM. Grading of neuroendocrine neoplasms: mitoses and (Oxf). 1979;11:69-74.
Ki-67 are both essential. Neuroendocrinology. 2014;100:221-227. 66. Wu X, Wen M. CT finding of ectopic pituitary adenoma: case report
47. Chatzipantelis P, Konstantinou P, Kaklamanos M, Apostolou G, Salla and review of literature. Head Neck. 2015;37:E120-E124.
C. The role of cytomorphology and proliferative activity in predicting 67. Asa SL. Tumors of the Pituitary Gland. The Atlas of Tumor Pathol-
biologic behavior of pancreatic neuroendocrine tumors: a study by ogy. Fascicle 15. Washington DC: Armed Forces Institute of Pathol-
endoscopic ultrasound-guided fine-needle aspiration cytology. Cancer ogy; 2011.
(Cancer Cytopathol). 2009;117:211-216. 68. Mete O, Asa SL. Clinicopathological correlations in pituitary adeno-
48. Kaklamanos M, Karoumpalis I, Salla C, et al. Diagnostic accuracy and mas. Brain Pathol. 2012;22:443-453.
clinical significance of the fine needle aspiration Ki-67 labelling index 69. Mete O, Asa SL. Therapeutic implications of accurate classification of
in pancreatic endocrine tumours. Endocr Relat Cancer. 2011;18:L1-L3. pituitary adenomas. Semin Diagn Pathol. 2013;30:158-164.
49. Weynand B, Borbath I, Bernard V, et al. Pancreatic neuroendocrine 70. Erovic BM, Goldstein DP, Kim D, et al. Parathyroid cancer: outcome
tumour grading on endoscopic ultrasound-guided fine needle aspira- analysis of 16 patients treated at the Princess Margaret Hospital. Head
tion: high reproducibility and inter-observer agreement of the Ki-67 Neck. 2013;35:35-39.
labelling index. Cytopathology. 2014;25:389-395. 71. Schulte KM, Talat N. Diagnosis and management of parathyroid can-
50. Piani C, Franchi GM, Cappelletti C, et al. Cytological Ki-67 in pan- cer. Nat Rev Endocrinol. 2012;8:612-622.
creatic endocrine tumours: an opportunity for pre-operative grading. 72. DeLellis RA. Parathyroid tumors and related disorders. Mod Pathol.
Endocr Relat Cancer. 2008;15:175-181. 2011;24(suppl 2):S78-S93.
51. Hasegawa T, Yamao K, Hijioka S, et al. Evaluation of Ki-67 index in 73. DeLellis RA. Tumors of the parathyroid gland. In: Atlas of Tumor
EUS-FNA specimens for the assessment of malignancy risk in pancre- Pathology. Vol 6. Washington, DC: Armed Forces Institute of Pathol-
atic neuroendocrine tumors. Endoscopy. 2014;46:32-38. ogy; 1993.
52. Moyana TN, Kendal WS, Chatterjee A, et al. Role of fine-needle aspi- 74. Kamp K, Feelders RA, van Adrichem RC, et al. Parathyroid hormone-
ration in the surgical management of pancreatic neuroendocrine related peptide (PTHrP) secretion by gastroenteropancreatic neuroen-
tumors: utility and limitations in light of the new World Health Orga- docrine tumors (GEP-NETs): clinical features, diagnosis, management,
nization classification. Arch Pathol Lab Med. 2014;138:896-902. and follow-up. J Clin Endocrinol Metab. 2014;99:3060-3069.
53. Maxwell JE, Sherman SK, Stashek KM, O’Dorisio TM, Bellizzi AM, 75. Duan K, Ezzat S, Asa SL, Mete O. Pancreatic neuroendocrine tumors
Howe JR. A practical method to determine the site of unknown primary producing GHRH, GH, ghrelin, PTH, or PTHrP. In: Rosa SL, Sessa
in metastatic neuroendocrine tumors. Surgery. 2014;156:1359-1366. F, eds. Pancreatic Neuroendocrine Neoplasms. New York: Springer
54. Strosberg JR, Coppola D, Klimstra DS, et al; North American Neuroen- International Publishing; 2015:125-139.
docrine Tumor Society (NANETS). The NANETS consensus guidelines 76. Nonaka D. Study of parathyroid transcription factor Gcm2 expression
for the diagnosis and management of poorly differentiated (high-grade) in parathyroid lesions. Am J Surg Pathol. 2011;35:145-151.
extrapulmonary neuroendocrine carcinomas. Pancreas. 2010;39:799-800. 77. Wong HH, Chu P. Immunohistochemical features of the gastrointesti-
55. Srivastava A, Hornick JL. Immunohistochemical staining for CDX-2, nal tract tumors. J Gastrointest Oncol. 2012;3:262-284.
PDX-1, NESP-55, and TTF-1 can help distinguish gastrointestinal car- 78. Grin A, Streutker CJ. Neuroendocrine tumors of the luminal gastroin-
cinoid tumors from pancreatic endocrine and pulmonary carcinoid testinal tract. Arch Pathol Lab Med. 2015;139:750-756.
tumors. Am J Surg Pathol. 2009;33:626-632. 79. Wang SC, Parekh JR, Zuraek MB, et al. Identification of unknown
56. Lau SK, Luthringer DJ, Eisen RN. Thyroid transcription factor-1: a primary tumors in patients with neuroendocrine liver metastases. Arch
review. Appl Immunohistochem Mol Morphol. 2002;10:97-102. Surg. 2010;145:276-280.
57. Ordonez NG. Thyroid transcription factor-1 is a marker of lung and 80. Raymond E, Hammel P, Dreyer C, et al. Sunitinib in pancreatic neu-
thyroid carcinomas. http://journals.lww.com/anatomicpathology/Full- roendocrine tumors. Target Oncol. 2012;7:117-125.
text/2000/07020/Thyroid_Transcription_Factor_1_is_a_Marker_of_ 81. Delbaldo C, Faivre S, Dreyer C, Raymond E. Sunitinib in advanced
Lung.7.aspx. Accessed May 20, 2016. pancreatic neuroendocrine tumors: latest evidence and clinical potential.
58. Lommel AV. Pulmonary neuroendocrine cells (PNEC) and neuroepi- Ther Adv Med Oncol. 2012;4:9-18.
thelial bodies (NEB): chemoreceptors and regulators of lung develop- 82. Strosberg J. Advances in the treatment of pancreatic neuroendocrine
ment. Paediatr Respir Rev. 2001;2:171-176. tumors (pNETs). Gastrointest Cancer Res. 2013;6(4 suppl 1):S10-S12.
59. Weissferdt A, Tang X, Wistuba II, Moran CA. Comparative immuno- 83. Oberg KE. Is it time to widen the use of somatostatin analogs in neu-
histochemical analysis of pulmonary and thymic neuroendocrine carci- roendocrine tumors? J Clin Oncol. 2009;27:4635-4636.
nomas using PAX8 and TTF-1. Mod Pathol. 2013;26:1554-1560. 84. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled,
60. Mete O, Asa SL. Composite medullary and papillary thyroid carcinoma double-blind, prospective, randomized study on the effect of octreotide
in a patient with MEN 2B: case report and review of C-cell lesions of LAR in the control of tumor growth in patients with metastatic neuro-
the thyroid. Pathol Case Rev. 2009;14:208-213. endocrine midgut tumors: a report from the PROMID Study Group.
61. Byrd-Gloster AL, Khoor A, Glass LF, et al. Differential expression of J Clin Oncol. 2009;27:4656-4663.
thyroid transcription factor 1 in small cell lung carcinoma and Merkel 85. Boudreaux JP, Putty B, Frey DJ, et al. Surgical treatment of advanced-
cell tumor. Hum Pathol. 2000;31:58-62. stage carcinoid tumors: lessons learned. Ann Surg. 2005;241:839-845;
62. Cheuk W, Kwan MY, Suster S, Chan JK. Immunostaining for thyroid discussion 845-846.
transcription factor 1 and cytokeratin 20 aids the distinction of small cell 86. Wang SC, Fidelman N, Nakakura EK. Management of well-
carcinoma from Merkel cell carcinoma, but not pulmonary from extrapul- differentiated gastrointestinal neuroendocrine tumors metastatic to the
monary small cell carcinomas. Arch Pathol Lab Med. 2001;125:228-231. liver. Semin Oncol. 2013;40:69-74.
63. Thompson LD, Seethala RR, Muller S. Ectopic sphenoid sinus pituitary 87. Chang H, Xu L, Mu Q. Primary functioning hepatic paraganglioma: a
adenoma (ESSPA) with normal anterior pituitary gland: a case report. Adv Ther. 2006;23:817-820.

882 Cancer Cytopathology December 2016


Algorithmic Approach to NETs in Targeted Biopsies/Duan and Mete

88. Corti B, D’Errico A, Pierangeli F, Fiorentino M, Altimari A, Grigioni differ in their transcription factor signatures: expression of ISL1,
W. Primary paraganglioma strictly confined to the liver and mimicking PDX1, NGN3, and CDX2. Virchows Arch. 2011;459:147-154.
hepatocellular carcinoma: an immunohistochemical and in situ hybrid- 107. Chan ES, Alexander J, Swanson PE, Jain D, Yeh MM. PDX-1,
ization study. http://journals.lww.com/ajsp/Fulltext/2002/07000/Pri- CDX-2, TTF-1, and CK7: a reliable immunohistochemical panel for
mary_Paraganglioma_Strictly_Confined_to_the.15.aspx. Accessed May pancreatic neuroendocrine neoplasms. Am J Surg Pathol. 2012;36:737-
21, 2016. 743.
89. Silberg DG, Swain GP, Suh ER, Traber PG. Cdx1 and cdx2 expression 108. Federspiel BH, Burke AP, Sobin LH, Shekitka KM. Rectal and colo-
during intestinal development. Gastroenterology. 2000;119:961-971. nic carcinoids. A clinicopathologic study of 84 cases. Cancer. 1990;65:
90. Moskaluk CA, Zhang H, Powell SM, Cerilli LA, Hampton GM, 135-140.
Frierson HF. Cdx2 protein expression in normal and malignant human 109. Burke AP, Thomas RM, Elsayed AM, Sobin LH. Carcinoids of the
tissues: an immunohistochemical survey using tissue microarrays. Mod jejunum and ileum: an immunohistochemical and clinicopathologic
Pathol. 2003;16:913-919. study of 167 cases. Cancer. 1997;79:1086-1093.
91. Barbareschi M, Roldo C, Zamboni G, et al. CDX-2 homeobox gene 110. Feurle GE, Anlauf M, Hamscher G, Arnold R, Kloppel G, Weihe E.
product expression in neuroendocrine tumors: its role as a marker of Xenin-immunoreactive cells and extractable xenin in neuroendocrine
intestinal neuroendocrine tumors. Am J Surg Pathol. 2004;28:1169- tumors of duodenal origin. Gastroenterology. 2002;123:1616-1626.
1176. 111. Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid
92. Erickson LA, Papouchado B, Dimashkieh H, Zhang S, Nakamura N, gland require Pax8 gene function. Nat Genet. 1998;19:87-90.
Lloyd RV. Cdx2 as a marker for neuroendocrine tumors of unknown 112. Plachov D, Chowdhury K, Walther C, Simon D, Guenet JL, Gruss
primary sites. Endocr Pathol. 2004;15:247-252. P. Pax8, a murine paired box gene expressed in the developing excre-
93. La Rosa S, Rigoli E, Uccella S, Novario R, Capella C. Prognostic and tory system and thyroid gland. Development. 1990;110:643-651.
biological significance of cytokeratin 19 in pancreatic endocrine 113. Bouchard M, Souabni A, Mandler M, Neubuser A, Busslinger M.
tumours. Histopathology. 2007;50:597-606. Nephric lineage specification by Pax2 and Pax8. Genes Dev. 2002;16:
94. Jaffee IM, Rahmani M, Singhal MG, Younes M. Expression of the 2958-2970.
intestinal transcription factor CDX2 in carcinoid tumors is a marker of 114. Long KB, Srivastava A, Hirsch MS, Hornick JL. PAX8 expression in
midgut origin. Arch Pathol Lab Med. 2006;130:1522-1526. well-differentiated pancreatic endocrine tumors: correlation with clini-
95. Lin X, Saad RS, Luckasevic TM, Silverman JF, Liu Y. Diagnostic value copathologic features and comparison with gastrointestinal and pul-
of CDX-2 and TTF-1 expressions in separating metastatic neuroendo- monary carcinoid tumors. Am J Surg Pathol. 2010;34:723-729.
crine neoplasms of unknown origin. Appl Immunohistochem Mol Mor- 115. Sangoi AR, Ohgami RS, Pai RK, Beck AH, McKenney JK, Pai RK.
phol. 2007;15:407-414. PAX8 expression reliably distinguishes pancreatic well-differentiated
96. Saad RS, Ghorab Z, Khalifa MA, Xu M. CDX2 as a marker for intes- neuroendocrine tumors from ileal and pulmonary well-differentiated
tinal differentiation: its utility and limitations. World J Gastrointest neuroendocrine tumors and pancreatic acinar cell carcinoma. Mod
Surg. 2011;3:159-166. Pathol. 2011;24:412-424.
97. La Rosa S, Franzi F, Albarello L, et al. Serotonin-producing entero- 116. Haynes CM, Sangoi AR, Pai RK. PAX8 is expressed in pancreatic
chromaffin cell tumors of the pancreas: clinicopathologic study of 15 well-differentiated neuroendocrine tumors and in extrapancreatic
cases and comparison with intestinal enterochromaffin cell tumors. poorly differentiated neuroendocrine carcinomas in fine-needle aspira-
Pancreas. 2011;40:883-895. tion biopsy specimens. Cancer (Cancer Cytopathol). 2011;119:193-
98. Agaimy A, Erlenbach-Wunsch K, Konukiewitz B, et al. ISL1 expression 201.
is not restricted to pancreatic well-differentiated neuroendocrine neo- 117. Lorenzo PI, Jimenez Moreno CM, Delgado I, et al. Immunohisto-
plasms, but is also commonly found in well and poorly differentiated chemical assessment of Pax8 expression during pancreatic islet devel-
neuroendocrine neoplasms of extrapancreatic origin. Mod Pathol. 2013; opment and in human neuroendocrine tumors. Histochem Cell Biol.
26:995-1003. 2011;136:595-607.
99. Thor S, Ericson J, Brannstrom T, Edlund T. The homeodomain LIM 118. Moreno CM, Lorenzo PI, Delgado I, et al. Pax8 detection in well-
protein Isl-1 is expressed in subsets of neurons and endocrine cells in differentiated pancreatic endocrine tumors: how reliable is it? Am J
the adult rat. Neuron. 1991;7:881-889. Surg Pathol. 2011;35:1906-1908.
100. Ahlgren U, Pfaff SL, Jessell TM, Edlund T, Edlund H. Independent 119. Ordonez NG. Value of PAX 8 immunostaining in tumor diagnosis: a
requirement for ISL1 in formation of pancreatic mesenchyme and islet review and update. Adv Anat Pathol. 2012;19:140-151.
cells. Nature. 1997;385:257-260. 120. Lloyd RV, Rosenbaum JN, Erickson LA. Immunohistochemical
101. Schmitt AM, Riniker F, Anlauf M, et al. Islet 1 (Isl1) expression is a approach to the diagnosis and prognostic evaluation of pancreatic
reliable marker for pancreatic endocrine tumors and their metastases. neuroendocrine neoplasms. In: Rosa SL, Sessa F, eds. Pancreatic Neu-
Am J Surg Pathol. 2008;32:420-425. roendocrine Neoplasms. New York: Springer International Publishing;
102. Graham RP, Shrestha B, Caron BL, et al. Islet-1 is a sensitive but not 2015:63-74.
entirely specific marker for pancreatic neuroendocrine neoplasms and 121. Modlin IM, Bodei L, Kidd M. Neuroendocrine tumor biomarkers:
their metastases. Am J Surg Pathol. 2013;37:399-405. from monoanalytes to transcripts and algorithms. Best Pract Res Clin
103. Koo J, Zhou X, Moschiano E, De Peralta-Venturina M, Mertens RB, Endocrinol Metab. 2016;30:59-77.
Dhall D. The immunohistochemical expression of islet 1 and PAX8 122. Kaemmerer D, Specht E, Sanger J, et al. Somatostatin receptors in
by rectal neuroendocrine tumors should be taken into account in the bronchopulmonary neuroendocrine neoplasms: new diagnostic, prog-
differential diagnosis of metastatic neuroendocrine tumors of nostic, and therapeutic markers. J Clin Endocrinol Metab. 2014;100:
unknown primary origin. Endocr Pathol. 2013;24:184-190. 831-840.
104. Park JY, Hong SM, Klimstra DS, Goggins MG, Maitra A, Hruban 123. Eriksson B, Oberg K, Stridsberg M. Tumor markers in neuroendo-
RH. Pdx1 expression in pancreatic precursor lesions and neoplasms. crine tumors. Digestion. 2000;62(suppl 1):33-38.
Appl Immunohistochem Mol Morphol. 2011;19:444-449. 124. Righi L, Volante M, Tavaglione V, et al. Somatostatin receptor tissue
105. Fendrich V, Ramerth R, Waldmann J, et al. Sonic hedgehog and distribution in lung neuroendocrine tumours: a clinicopathologic and
pancreatic-duodenal homeobox 1 expression distinguish between duode- immunohistochemical study of 218 “clinically aggressive” cases. Ann
nal and pancreatic gastrinomas. Endocr Relat Cancer. 2009;16:613-622. Oncol. 2010;21:548-555.
106. Hermann G, Konukiewitz B, Schmitt A, Perren A, Kloppel G. Hor- 125. Volante M, Brizzi MP, Faggiano A, et al. Somatostatin receptor type
monally defined pancreatic and duodenal neuroendocrine tumors 2A immunohistochemistry in neuroendocrine tumors: a proposal of

Cancer Cytopathology December 2016 883


Review Article

scoring system correlated with somatostatin receptor scintigraphy. 143. Kim JY, Kim KS, Kim KJ, et al. Non-L-cell immunophenotype and
Mod Pathol. 2007;20:1172-1182. large tumor size in rectal neuroendocrine tumors are associated with
126. Okuwaki K, Kida M, Mikami T, et al. Clinicopathologic characteris- aggressive clinical behavior and worse prognosis. Am J Surg Pathol.
tics of pancreatic neuroendocrine tumors and relation of somatostatin 2015;39:632-643.
receptor type 2A to outcomes. Cancer. 2013;119:4094-4102. 144. Gastrointestinal Pathology Study Group of Korean Society of Patholo-
127. Corleto VD, Falconi M, Panzuto F, et al. Somatostatin receptor sub- gists, Sohn JH, Cho MY, et al. Prognostic significance of defining L-
types 2 and 5 are associated with better survival in well-differentiated cell type on the biologic behavior of rectal neuroendocrine tumors in
endocrine carcinomas. Neuroendocrinology. 2009;89:223-230. relation with pathological parameters. Cancer Res Treat. 2015;47:813-
128. Kulaksiz H, Eissele R, Rossler D, et al. Identification of somatostatin 822.
receptor subtypes 1, 2A, 3, and 5 in neuroendocrine tumours with 145. Duan K, Mete O. Familial hyperparathyroidism syndromes. Diagn
subtype specific antibodies. Gut. 2002;50:52-60. Histopathol. 2016;22:92-100.
129. Cives M, Strosberg J. The expanding role of somatostatin analogs in 146. Pellegata NS. MENX and MEN4. Clinics (Sao Paulo). 2012;67(suppl
gastroenteropancreatic and lung neuroendocrine tumors. Drugs. 2015; 1):13-18.
75:847-858. 147. Molatore S, Pellegata NS. The MENX syndrome and p27: relation-
130. Lapa C, Hanscheid H, Wild V, et al. Somatostatin receptor expres- ships with multiple endocrine neoplasia. Prog Brain Res. 2010;182:
sion in small cell lung cancer as a prognostic marker and a target for 295-320.
peptide receptor radionuclide therapy [published online ahead of print 148. Thakker RV. Multiple endocrine neoplasia type 1 (MEN1) and type
February 25, 2016]. Oncotarget. doi:10.18632/oncotarget.7706. 4 (MEN4). Mol Cell Endocrinol. 2014;386:2-15.
131. Papotti M, Croce S, Macri L, et al. Correlative immunohistochemical and 149. Pellegata NS, Quintanilla-Martinez L, Siggelkow H, et al. Germ-line
reverse transcriptase polymerase chain reaction analysis of somatostatin mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome
receptor type 2 in neuroendocrine tumors of the lung. http://journals.lww. in rats and humans. Proc Natl Acad Sci U S A. 2006;103:15558-
com/molecularpathology/Fulltext/2000/03000/Correlative_Immunohisto- 15563.
chemical_and_Reverse.8.aspx. Accessed May 21, 2016. 150. Francis JM, Kiezun A, Ramos AH, et al. Somatic mutation of
132. Kim HS, Lee HS, Kim WH. Clinical significance of protein expres- CDKN1B in small intestine neuroendocrine tumors. Nat Genet.
sion of cyclooxygenase-2 and somatostatin receptors in gastroentero-
2013;45:1483-1486.
pancreatic neuroendocrine tumors. Cancer Res Treat. 2011;43:181-
151. Thakker RV. Multiple endocrine neoplasia type 1. Indian J Endocrinol
188.
Metab. 2012;16(suppl 2):S272-S274.
133. Crona J, Norlen O, Antonodimitrakis P, Welin S, Stalberg P,
152. Brandi ML, Gagel RF, Angeli A, et al. Guidelines for diagnosis and
Eriksson B. Multiple and secondary hormone secretion in patients
therapy of MEN type 1 and type 2. J Clin Endocrinol Metab. 2001;
with metastatic pancreatic neuroendocrine tumours. J Clin Endocrinol
86:5658-5671.
Metab. 2015;101:445-452.
153. Yousef I, Siyam F, Layfield L, Freter C, Sowers JR. Cervical neuroen-
134. Salon C, Moro D, Lantuejoul S, et al. E-cadherin-beta-catenin adhe-
docrine tumor in a young female with Lynch Syndrome. Neuro Endo-
sion complex in neuroendocrine tumors of the lung: a suggested role
crinol Lett. 2014;35:89-94.
upon local invasion and metastasis. Hum Pathol. 2004;35:1148-1155.
154. Miquel C, Sabourin JC, Elias D, et al. An appendix carcinoid tumor
135. Li CC, Xu B, Hirokawa M, et al. Alterations of E-cadherin, alpha-cat-
in a patient with hereditary nonpolyposis colorectal cancer. Hum
enin and beta-catenin expression in neuroendocrine tumors of the gas-
Pathol. 2004;35:1564-1567.
trointestinal tract. Virchows Arch. 2002;440:145-154.
155. Karamurzin Y, Zeng Z, Stadler ZK, et al. Unusual DNA mismatch
136. Zhang L, Smyrk TC, Oliveira AM, et al. KIT is an independent
prognostic marker for pancreatic endocrine tumors: a finding derived repair-deficient tumors in Lynch syndrome: a report of new cases and
from analysis of islet cell differentiation markers. Am J Surg Pathol. review of the literature. Hum Pathol. 2012;43:1677-1687.
2009;33:1562-1569. 156. Eisenhofer G, Tischler AS, de Krijger RR. Diagnostic tests and bio-
137. Deshpande V, Fernandez-del Castillo C, Muzikansky A, et al. Cyto- markers for pheochromocytoma and extra-adrenal paraganglioma:
keratin 19 is a powerful predictor of survival in pancreatic endocrine from routine laboratory methods to disease stratification. Endocr
tumors. Am J Surg Pathol. 2004;28:1145-1153. Pathol. 2012;23:4-14.
138. Serra S, Asa SL, Bamberger AM, Wagener C, Chetty R. CEACAM1 157. Castelblanco E, Santacana M, Valls J, et al. Usefulness of negative
expression in pancreatic endocrine tumors. Appl Immunohistochem Mol and weak-diffuse pattern of SDHB immunostaining in assessment of
Morphol. 2009;17:286-293. SDH mutations in paragangliomas and pheochromocytomas. Endocr
139. Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR Pathol. 2013;24:199-205.
pathway genes are frequently altered in pancreatic neuroendocrine 158. Gimenez-Roqueplo AP, Favier J, Rustin P, et al. Mutations in the
tumors. Science. 2011;331:1199-1203. SDHB gene are associated with extra-adrenal and/or malignant phaeo-
140. Marinoni I, Kurrer AS, Vassella E, et al. Loss of DAXX and ATRX chromocytomas. Cancer Res. 2003;63:5615-5621.
are associated with chromosome instability and reduced survival of 159. Niemeijer ND, Papathomas TG, Korpershoek E, et al. Succinate
patients with pancreatic neuroendocrine tumors. Gastroenterology. dehydrogenase (SDH)-deficient pancreatic neuroendocrine tumor
2014;146:453-460.e5. expands the SDH-related tumor spectrum. J Clin Endocrinol Metab.
141. Estrella JS, Broaddus RR, Mathews A, et al. Progesterone receptor 2015;100:E1386-E1393.
and PTEN expression predict survival in patients with low- and 160. Gill AJ, Toon CW, Clarkson A, et al. Succinate dehydrogenase defi-
intermediate-grade pancreatic neuroendocrine tumors. Arch Pathol Lab ciency is rare in pituitary adenomas. Am J Surg Pathol. 2014;38:560-
Med. 2014;138:1027-1036. 566.
142. Lee HS, Chen M, Kim JH, et al. Analysis of 320 gastroenteropancre- 161. Singh S, Asa SL, Dey C, et al. Diagnosis and management of gastro-
atic neuroendocrine tumors identifies TS expression as independent intestinal neuroendocrine tumors: an evidence-based Canadian consen-
biomarker for survival. Int J Cancer. 2014;135:128-137. sus. Cancer Treat Rev. 2016;47:32-45.

884 Cancer Cytopathology December 2016

You might also like