You are on page 1of 20

All Together Now

Standardization of Nomenclature for


Neuroendocrine Neoplasms across Multiple
Organs
Pari Jafari, MD*, Aliya N. Husain, MBBS, Namrata Setia, MD

KEYWORDS
 Neuroendocrine neoplasm (NEN)  Neuroendocrine tumor (NET)  Small-cell/
large-cell neuroendocrine carcinoma (SC/LC NEC)  Typical/atypical carcinoid
 Small-cell lung carcinoma (SCLC)

Key points
 Neuroendocrine neoplasms (NENs) arise across virtually all organ systems but are most common in the
gastroenteropancreatic (GEP) tract and lungs.
 NENs across anatomic sites can be grouped into well- and poorly differentiated tiers (neuroendocrine
tumors [NETs] and neuroendocrine carcinomas [NECs], respectively) based on morphology and prolif-
erative activity. Underpinning this paradigm are distinct, and often mutually exclusive, molecular and
immunohistochemical (IHC) profiles.
 NECs are by definition high-grade and are typically associated with dismal prognoses. In contrast,
NETs exhibit a complex spectrum of behavior, necessitating a two- or three-tiered grading rubric,
with proliferative activity serving as a crucial parameter.
 Nomenclature and grading of NENs have historically varied across anatomic sites, though efforts to-
ward conceptual unification are underway, most notably with the adoption of a standardized rubric
for GEP-NENs.

ABSTRACT NEN classification, with a focus on NENs of the

N
pancreas and lungs.
euroendocrine neoplasms (NENs) span
virtually all organ systems and exhibit a
broad spectrum of behavior, from indolent OVERVIEW
to highly aggressive. Historically, nomenclature
and grading practices have varied widely across, In 1907, physicians abreast of the literature might
and even within, organ systems. However, certain have noted a brief but striking report by the
core features are recapitulated across anatomic German pathologist Siegfried Oberndorfer in the
sites, including characteristic morphology and Frankfurt Journal of Pathology, describing a new
the crucial role of proliferative activity in prognos- and intriguing class of small bowel neoplasms.1
tication. A recent emphasis on unifying themes These lesions appeared relatively indolent–
has driven an increasingly standardized approach though Oberndorfer later came to recognize that
to NEN classification, as delineated in the World a subset harbored significant metastatic
Health Organization’s Classification of Tumours potential–and were well-circumscribed, submuco-
surgpath.theclinics.com

series. Here, we review recent developments in sal, and composed of relatively bland cells forming

Department of Pathology, The University of Chicago Medicine, 5841 South Maryland Avenue, MC 6101, Room
S-638, Chicago, IL 60637, USA
* Corresponding author.
E-mail address: Pari.Jafari@uchicagomedicine.org

Surgical Pathology 16 (2023) 131–150


https://doi.org/10.1016/j.path.2022.09.012
1875-9181/23/Ó 2022 Elsevier Inc. All rights reserved.
132 Jafari et al

nests or rosettes. Today, Oberndorfer’s “benign index (LI), in NEN classification and
carcinomas” or carcinoid tumors (Karzinoid prognostication.8,15–18
Tumoren) are known to arise from the neuroendo-
crine system, a unique population of cells exhibit- Despite these common denominators, ap-
ing neurosecretory properties and disseminated proaches to NEN classification and nomenclature
across multiple anatomic sites, from the pituitary have long differed across organ systems. This het-
to the pancreas.1–6 Corresponding neoplasms erogeneity reflects subspecialty preferences and
have been known by a variety of site- and era- historical associations, and has given rise to persis-
specific names over the years (Box 1); at present, tent concerns about miscommunication between
the more neutral and inclusive term neuroendo- specialties and the challenge of remaining up-to-
crine neoplasm is generally favored.2–8 date with each organ system’s evolving classifica-
Neuroendocrine neoplasms (NENs) represent tion criteria.5,7,9 Those concerns are heightened
an unusual class of tumors: at once capacious, by the rising incidence of NENs, from 1.09 to 6.98
yet bound together by many of the distinctive cases per 100,000 between 1973 and 2012 in the
morphologic features that Oberndorfer described United States, likely attributable in large measure
(Fig. 1, Table 1). Indeed, the study of NENs is to enhanced detection modalities.19 In response,
characterized by several overarching themes, an expert committee convened by the World Health
including: Organization (WHO)/International Agency for
Research on Cancer (IARC) issued a proposal in
1. Bifurcation of neoplasms into relatively well- 2018 for streamlining NEN classification across or-
differentiated (“neuroendocrine tumor,” NET) gan systems, using a 2017 WHO scheme for
and poorly differentiated (“neuroendocrine car- pancreatic NENs (panNENs) as a conceptual tem-
cinoma,” NEC) tiers, each with a distinct clinico- plate.9 As detailed below, this rubric has since
pathologic profile.7,8 NETs exhibit a complex been extended, with site-specific adjustments, to
spectrum of behavior, from indolent to relatively include all gastroenteropancreatic (GEP)-NENs as
aggressive, necessitating a superimposed two- well as NENs of the upper aerodigestive tract and
or three-tiered grading scheme (Figs. 2–4).7–11 salivary glands.7,8,16,17,20–22 A similar approach
NECs are invariably aggressive and are high- has been discussed in the context of other organ
grade by definition; they are further classified systems, particularly the lungs.10,15 This unfolding
based on small-cell (SC; Fig. 5) versus large- paradigm shift is captured in the WHO’s Classifica-
cell (LC; Fig. 6) morphology.7,8 tion of Tumours series (colloquially known as the
2. The increasingly crucial role of ancillary testing WHO “Blue Books”), most notably in Endocrine
in classification and grading, including molecu- and Neuroendocrine Tumors (2022), the first Blue
lar and immunohistochemical (IHC) Book to discuss both endocrine and neuroendo-
studies.7–10,12–14 crine neoplasms across endocrine and non-
3. The importance of proliferative activity, as endocrine sites.7,8 Current WHO classification
gauged by mitotic count or Ki67 labeling criteria are summarized in Table 2.

Box 1
Selected historic nomenclature associated with NENs
Nomenclature reflecting morphology
Carcinoid/atypical carcinoid (still in use at certain sites, particularly in the thorax)
Bronchial adenoma
Oat cell carcinoma/sarcoma
Nomenclature reflecting tumor cell function/hormonal products
Amine precursor uptake and decarboxylation (APUD)oma
Glucagonoma, gastrinoma, insulinoma, vasoactive intestinal peptide (VIP)oma, somatostatinoma
Nomenclature reflecting presumptive cell of origin
Islet cell tumor
Kulchitsky cell tumor
Argentaffin/(Entero)chromaffin(-type)/Argyrophil cell tumor
Standardized Nomenclature for Neuroendocrine Neoplasms 133

Fig. 1. Neuroendocrine tumors (NETs) classically exhibit organoid growth, as seen here on hematoxylin and eosin
(H&E) staining. Common patterns include nests (A), rosettes (B), and trabeculae (C). However, diffuse growth is
also observed, as in this atypical carcinoid of the lung (D).

In this article, we explore the shifting landscape carcinoid is retained at certain anatomic sites,
of NEN classification with an emphasis on the most notably in the thorax.1,2,4,7,8,15 Under the cur-
themes described above. Although these princi- rent schema, NETs are juxtaposed with SC and LC
ples are perhaps best delineated in NENs of the NECs, which are intrinsically high-grade and are
pancreas and lungs, two organs at the center of associated with poor prognoses. NETs and
NEN clinicopathologic investigation over the NECs each exhibit characteristic morphology
years, they are readily applicable to NENs across across anatomic sites (see Table 1).7,8
anatomic sites.7,8
Of note, non-epithelial NENs, such as pheochro-
mocytomas/paragangliomas; non-neoplastic en- WELL-DIFFERENTIATED NEUROENDOCRINE
tities, such as diffuse idiopathic pulmonary NEOPLASMS: NEUROENDOCRINE TUMORS
neuroendocrine hyperplasia (DIPNECH); mixed
neuroendocrine-non-neuroendocrine neoplasms The “classic” NET is a well-circumscribed sub-
(MiNENs); and special considerations associated mucosal lesion composed of bland-appearing
with functional tumors are beyond the scope of cells with granular, eosinophilic cytoplasm and
this article and will not be discussed here. round or polygonal contours. The nuclei are typi-
cally round with distinctive “salt and pepper”
clumping of chromatin and inconspicuous
NEUROENDOCRINE NEOPLASMS: A TWO- nucleoli.7,8,23 Atypia and degenerative changes
TIERED PARADIGM may be present, particularly in higher-grade
NETs, but are not in and of themselves worrisome
Oberndorfer’s original article on “carcinoids” features.23,24 Crucially, mitoses are relatively
described a class of relatively well-differentiated infrequent.7,8,15–17 As described by Soga and
NENs known today as NETs, though the term Tazawa in their seminal 1971 analysis,
134 Jafari et al

Table 1
Key histologic features of neuroendocrine tumors and neuroendocrine carcinomas (NECs)

NET NEC
Morphologic Microarchitecture Organoid (nests, rosettes, Sheetlike/diffuse
features trabeculae, and
pseudoglands) or solid;
mixed phenotypes
common
Necrosis Absent or punctate Marked/geographic
Mitoses Rare to brisk Numerous; often atypical
Cytologic features Medium-sized, SC NEC: Small cells (<3x size of
round cells; low lymphocyte) with minimal
N:C ratio; “salt and basophilic cytoplasm; granular
pepper” chromatin; chromatin and inconspicuous
moderate to abundant nucleoli; molding and crush
eosinophilic cytoplasm; artifact often present
inconspicuous nucleoli LC NEC: Large, pleomorphic cells
(w3–5x size of lymphocyte) with
moderate/abundant amphophilic
cytoplasm; prominent nucleoli
IHC profile Core Cytokeratin 8/181, CK 8 and 181/ , CAM5.21/
neuroendocrine CAM5.21 [keratins, particularly CAM5.2,
panel Chromogranin1 may exhibit dot-like staining in SC
Synaptophysin1 NECs]
Less routinely: Chromogranin 
CD561 Synaptophysin1
INSM11 Less routinely:
SSTR2A/51 CD561
INSM11
SSTR2A/5 /1
Peptide hormones Positive staining in Almost uniformly negative
functional tumors (exception: hormone-producing
SCLC)
Ki67 Low (<20% for G1-G2; High (>20%; frequently >55% and
typically <55% for often >70%)
G3-type tumors)
Rb Rb retention Rb loss
p53 Wild-type pattern p53 mutant pattern (compete loss or
diffuse positivity)
Selected  Pancreatic origin: ATRX or DAXX loss (panNETs); 1ISL1;  CDX2
site-specific  Tubular gastrointestinal tract (particularly mid-gut): 1CDX2
findings  Jejunoileal NET: clusterin
 Lung, thyroid: 1TTF-1
Molecular Selected  11q13 deletions most  RB and/or TP53 mutations across
profile site-specific common (contains anatomic sites
findings MEN1 locus)  MYC amplifications (particularly
 MEN1, ATRX, or DAXX panNECs)
loss in panNETs  ASCL1, NEUROD1, POU2F3, YAP1
 MEN1 mutations in alterations in SCLC
lung carcinoids; other  NSCLC-type mutations (eg, KRAS)
alterations (eg, PSIP, in NSCLC-like lung LC NEC
EIF1AX, KMT2, ARID1A)
also described

Abbreviations: LC NEC, large-cell neuroendocrine carcinoma; N:C ratio, nuclear:cytoplasmic ratio; NSCLC, non-small-cell
lung carcinoma; SC NEC, small-cell neuroendocrine carcinoma; SCLC, small-cell lung carcinoma.

microarchitecture classically assumes one of four pseudoglandular (eg, tubules, acini, and ro-
patterns, or a combination thereof: type A, settes); and type D, diffuse or otherwise
nodular/nested; type B, trabecular; type C, atypical.25
Standardized Nomenclature for Neuroendocrine Neoplasms 135

Fig. 2. Gastroentero-pancreatic (GEP)-NETs are graded based on mitotic activity and/or Ki67 labeling index (LI).
G1 GEP-NET with a diffuse pattern of growth (H&E, A) and <3% Ki67 LI (B). Note that Ki67 may also highlight
lymphocytes and endothelial cells, serving as a useful positive internal control; however, these non-lesional cells
should not be included in the LI assessment. G2 GEP-NET (H&E, C) showing trabecular architecture and Ki67 LI of
18% (D). G3 GEP-NET exhibiting multiple mitoses (H&E, E) and Ki67 LI of 25% (F).

Neuroendocrine Tumor Grading: A Three- reflected in a series of proposed grading parame-


Tiered Approach ters dating from the 1990s.16,26–28 Though these
schemes were originally highly site-specific, the
Despite their relatively bland appearance as a
WHO Blue Books have embraced an increasingly
whole, NETs exhibit a striking spectrum of
unified approach in recent years (see Table 2),
behavior, ranging from indolent to aggressive.
beginning with the 2019 publication of Digestive
Prognosis-driven, clinically meaningful stratifica-
System Tumours, which extended the existing
tion of these tumors has proven challenging, as
136 Jafari et al

Fig. 3. Typical carcinoid of the lung, demonstrating (H&E, A) bland cells with moderate amounts of amphophilic
cytoplasm and prominent round nuclei with characteristic “salt and pepper” chromatin. Necrosis is absent, and
mitoses are exceedingly rare (<2 mitoses/2 mm2). The cells exhibit diffuse staining for CD56 (B; membranous
pattern), chromogranin (C; cytoplasmic), and synaptophysin (D; cytoplasmic).

panNEN rubric to include all GEP-NENs. In this since been extended to the upper aerodigestive
paradigm, NENs are strictly partitioned into tract/salivary glands while retaining certain site-
NETs and NECs, with NECs considered exclu- specific nuances, such as the inclusion of necro-
sively high-grade and NETs stratified (G1–G3) sis as a NET grading parameter and the caveat
based on proliferative activity (see that G3 NETs have yet to be fully characterized
Fig. 2).7,9,16,20,21 This bifurcated approach has in the head and neck.7,8,17,22

Fig. 4. H&E-stained sections of an atypical carcinoid of the lung, demonstrating (A) increased atypia and mitoses
as well as (B) focal necrosis.
Standardized Nomenclature for Neuroendocrine Neoplasms 137

Fig. 5. Small-cell lung carcinoma demonstrating (A, H&E) characteristic scant amphophilic cytoplasm, high nucle-
ar:cytoplasmic ratio, and nuclear molding. Staining with (B) synaptophysin and (C) chromogranin confirms neuro-
endocrine differentiation. Ki67 LI is 90% (D).

The roots of this approach can be traced to the >20%.26 However, the NEC category was subse-
WHO’s 2010 panNEN grading rubric, which split quently shown to include both “bona fide
tumors into G1/G2 (NET) or G3 (NEC) categories NECs”––poorly differentiated neoplasms with
solely based on proliferative activity: G3 tumors either SC or LC morphology–and a vexing subcat-
were defined by > 20 mitoses/2 mm2 or Ki67 LI egory of relatively well-differentiated tumors with

Fig. 6. Large-cell neuroendocrine carcinoma of the lung, showing (A) abundant amphophilic cytoplasm and
irregularly shaped nuclei with predominantly vesicular or clumped chromatin, arranged here in a sheetlike
pattern with focal necrosis. Synaptophysin IHC (B) shows patchy positivity.
138 Jafari et al

Table 2
Classification of neuroendocrine neoplasms across selected anatomic sites: World Health Organization
criteria (5th edition)8,15–17

Neuroendocrine Tumor Neuroendocrine Carcinoma


Key histologic Bland cells with round nuclei, salt- Small-cell (SC) NEC: Diameter <3
features across and-pepper chromatin, times that of lymphocyte; scant
anatomic sites inconspicuous nucleoli; classically cytoplasm with high N:C ratio;
arranged in rosettes, nests, or granular chromatin;
trabeculae; focal necrosis and inconspicuous nucleoli; sheetlike
increased atypia seen with arrangement with extensive
increasing grade necrosis
Large-cell (LC) NEC: Diameter w3–5
times that of lymphocyte;
amphophilic cytoplasm; relatively
low N:C ratio; organoid
architecture with extensive
necrosis

WHO 5th Edition NET Grading WHO 5th Edition NEC Subclassification
Criteria (G1-G3) Criteria (SC vs LC NEC)
Gastroentero- G1 NET <2 mitoses/2 mm2 Small-cell NEC Characteristic small-cell
pancreatic and Ki67 < 3% morphology and
tract G2 NET 2–20 mitoses/2 mm2 > 20 mitoses/2 mm2
and/or Ki67 3–20% and/or Ki67 > 20%
(typically >70%)
G3 NET >20 mitoses/2 mm2 Large-cell NEC Characteristic large-cell
and/or Ki67 > 20% morphology and
(typically <55%) > 20 mitoses/2 mm2
and/or Ki67 > 20%
(typically > 70%)
Lung and thymus Typical <2 mitoses/2 mm2 Small-cell Characteristic small-cell
carcinoid and no necrosis (lung) morphology and
Atypical 2–10 mitoses/2 mm2 carcinoma >10 mitoses/2 mm2;
carcinoid and/or necrosis (SCLC) Ki67 not part of
formal criteria but
often >30%
(typically >75%)
Emerging Atypical carcinoid Large-cell NEC Characteristic large-cell
categorya: morphology morphology and
carcinoids and >10 mitoses/ >10 mitoses/2 mm2;
with high 2 mm2; Ki67 Ki67 not part of
proliferative typically >30% formal criteria but
activity often >30%
(typically >75%)
Upper G1 NET <2 mitoses/2 mm2 Small-cell NEC Characteristic small-cell
aerodigestive and no necrosis; morphology and
tract and Ki67 typically >10 mitoses/2 mm2
salivary glands <20% and/or Ki67 > 20%
(typically >70%)
G2 NET 2–10 mitoses/2 mm2 Large-cell NEC Characteristic large-cell
and/or necrosis; morphology and
Ki67 typically <20% >10 mitoses/2 mm2
Emerging Relatively and/or Ki67 > 20%
categorya: well-differentiated (typically >50%)
G3 NET histology and
>10 mitoses/2 mm2;
Ki67 > 20%

Abbreviations: N:C ratio, nuclear:cytoplasmic ratio.


a
Lung/thymic carcinoids with high proliferative activity (analogous to G3 GEP-NETs) are discussed, but not formally
categorized, in the fifth edition of the WHO Classification of Tumours series. Similarly, G3 NETs of the upper aerodigestive
tract/salivary glands are mentioned as a potential category, but have yet to be fully delineated.
Standardized Nomenclature for Neuroendocrine Neoplasms 139

an elevated Ki67 LI (typically 30–40%; generally components supports a diagnosis of G3 NET,


<55%), prompting a default diagnosis of whereas the presence of interspersed non-
NEC.11,29–41 Interestingly, several studies noted neuroendocrine elements (eg, adenocarcinoma,
comparatively low (<20 mitoses/2 mm2) mean squamous cell carcinoma) favors NEC.24,33 Table 3
mitotic counts for this subgroup, despite the summarizes these and other “rules of thumb” for
elevated Ki67 LI.11,31,32,36,37 Prognosis fell some- differentiating between G3 NETs and NECs.
where between that of G2 NETs and the bona
fide (poorly -differentiated) NECs, with reduced POORLY DIFFERENTIATED NEUROENDOCRINE
amenability to cisplatin-based therapy relative to NEOPLASMS: NEUROENDOCRINE
NECs.11,29–41 Crucially, ancillary testing revealed CARCINOMAS
a molecular and IHC profile relatively similar to
that of G1/G2 NETs.31,32,34,42 In response to these Unlike NETs, all NECs are high-grade by default:
findings, the WHO’s 2017 Classification of Tu- as such, there is no role for a superimposed
mours of Endocrine Organs refashioned panNETs grading scheme. Instead, NECs are subdivided
into three tiers (G1, G2, and G3), with G3 serving to based on SC versus LC morphology.7,8 Of note,
house the former “well-differentiated NEC” sub- pulmonary SC NECs are traditionally known as
category. This change broadened the definition small-cell lung carcinomas (SCLCs), as not all ex-
of panNET to include all relatively well- press core neuroendocrine markers such as chro-
differentiated tumors, regardless of proliferative mogranin and synaptophysin.10,15 Both SC and LC
activity, while narrowing the panNEC category to NECs are characterized by extensive necrosis and
include only those “true” carcinomas that united marked proliferative activity (see Figs. 5–6).7,8 SC
poorly differentiated histology with high prolifera- NEC cells measure up to three times the diameter
tive activity.43,44 This rubric was subsequently of a lymphocyte and exhibit round, ovoid, or angu-
expanded to encompass all GEP-NENs.16,20,21 lated contours; granular chromatin; and scant
A similar reappraisal is currently underway cytoplasm, resulting in a high nuclear:cytoplasmic
across other organ systems, most notably in ratio. Nuclear molding is common, as is extensive
the lungs, where NETs (known in the thorax as necrosis, and crush artifact and/or encrustation of
carcinoids) have traditionally been bifurcated vessel walls with extruded chromatin may be
into typical and atypical carcinoids (TCs and observed. Growth is classically sheetlike, though
ACs, respectively) based on mitotic count and organoid microarchitecture has also been
presence of necrosis (see Table 2).10,15 Howev- described.7,8,10,53–55 In contrast, large-cell NECs
er, this paradigm has been challenged by the exhibit variable quantities of amphophilic cyto-
recent identification of “highly proliferative carci- plasm and are approximately three to five times
noids,” which combine the morphology of ACs the diameter of a lymphocyte in size. The chro-
with the marked proliferative activity character- matin may be granular or vesicular; nucleoli are
istic of NECs (>10 mitoses/2 mm2; Ki67 LI typi- often prominent. The cells typically palisade or
cally 20–40%). Though available data are form nests, trabeculae, or rosettes, among other
limited, outcomes appear to be intermediate be- patterns.7,8,10,54
tween those of lung ACs and NECs, whereas the As noted above, there may be considerable
associated molecular and IHC profile is similar to morphologic overlap among high-grade NENs
that of carcinoids.7,8,10,45–52 These findings sug- (ie, G3 NETs and SC and LC NECs): indeed, in
gest that lung carcinoids may be amenable to Tang and colleagues’32 2016 evaluation of pan-
G1-G3 stratification, analogous to GEP- NECs and G3 panNETs, diagnostic consensus
NETs.7,10,15 Such a change would mark another was reached in just a third of cases.8,24,50 Both
key step toward the unification of NEN nomen- SC and LC NECs also raise the differential of
clature and categorization. poorly differentiated non-neuroendocrine carci-
Of note, as the diagnostic criteria for G3 NETs/ nomas, whereas SC NECs may warrant consider-
highly proliferative carcinoids include a threshold ation of other “small round blue cell tumors,” such
for mitotic activity, but no ceiling, histology plays as lymphoma, neuroblastoma, and Ewing
a pivotal role in diagnosis. By definition, G3 NET sarcoma.8,10,15,53,55
histology falls somewhere between that of G1/G2
NETs and (LC) NECs; in practice, this distinction CLINICAL IMPLICATIONS OF
may be quite challenging, as G3 NETs may exhibit NEUROENDOCRINE NEOPLASM
high-grade, NEC-like features, such as nuclear aty- CLASSIFICATION
pia and necrosis (punctate or even
geographic).8,24,32,50 It has been suggested that Each NEN category–NET versus NEC as well as in-
identification of lower-grade (G1-G2) NET dividual NET tiers–carries substantial clinical
140 Jafari et al

Table 3
Features favoring a diagnosis of G3 neuroendocrine tumor versus neuroendocrine carcinoma

G3 NET NEC
Proliferative  Ki67 LI typically 20%-55%  Ki67 LI >55% strongly favors NEC
activity  Mitotic count may be  Mitotic count typically concordant
discordant (within G2 NET with Ki67 LI
range)
IHC profile  Rb1 retained, p53 wild-type  Rb1 loss and/or p53-mutated pattern
expression (global loss or diffuse positivity)
 Loss of menin  ATRX/DAXX  Menin, ATRX, and DAXX retained
 Retained SSTR2A  Loss of SSTR2A
Molecular profile  MEN1  ATRX or DAXX  Mutated RB and/or TP53
mutations (panNET)  Absence of MEN1 or ATRX/DAXX
 Mutations in genes associated mutations
with chromatin remodeling;
MEN1 mutations most common
 RB and TP53 wild-type
Other findings Presence of areas with G1/G2 Presence of areas with non-neuroendocrine
histology differentiation (eg, adenocarcinoma or
squamous cell carcinoma)

ramifications. Recently reported median overall NETs and analogous lesions at other sites re-
survival rates for panNETs, for instance, improve mains to be more fully defined, as their clinical
in stepwise fashion from G3 (19–99 months) to profile is intermediate between that of G2
G1 (over 100 months), versus approximately 10- NETs and NECs.10,11,29–41,45–52
16 months for panNECs.11,29–39,41,56 Each tier  NECs: Treatment of SC NECs across
also dictates a distinct approach to treatment, as anatomic sites has traditionally followed the
outlined below. template established for SCLC, which is char-
acterized by striking initial sensitivity to
 NETs: Resection is generally curative for platinum-based chemotherapy and radia-
localized/locoregional NETs (stages I-II).57–61 tion.30,59,73–76 Very early stage SCLCs may
Non-resectable/advanced disease is primarily undergo resection, though the evidence
managed with somatostatin analogs (SSAs), base is mixed and it should be emphasized
which exploit NET overexpression of somato- that SCLC is typically relatively advanced at
statin receptors (SSTRs). Examples include presentation, precluding resection.73,77 Of
octreotide long-acting release (LAR) and lan- note, immune checkpoint inhibitors, such as
reotide, which preferentially bind to SSTR2 pembrolizumab and nivolumab, are often
and SSTR5, exerting an antiproliferative effect combined with conventional chemotherapy
while also providing symptomatic relief for in extensive-stage SCLC; their role in extrap-
functional tumors.58–65 In patients with a high ulmonary SC NECs remains to be
burden of hepatic metastases, debulking or explored.73–75
liver-directed therapies, such as transarterial
chemoembolization (TACE), may be appro- Data regarding the management of LC NECs are
priate.59,60,66,67 In the event of disease pro- limited and are largely derived from studies of pul-
gression on SSAs, options include peptide monary LC NECs. In contrast to SCLCs, pulmo-
receptor radionuclide therapy (PRRT), which nary LC NECs presenting through at least stage
consists of an SSA chelated to a radioisotope, II appear relatively amenable to resection and
such as 111In, 90Y, or 177Lu, enabling adjuvant chemotherapy.78–81 More advanced dis-
enhanced visualization and targeted treat- ease is generally treated with platinum-based reg-
ment of lesional tissue.58–61,68 Although the imens, following the SCLC model.30,59,78,79,82 Of
role of conventional chemotherapy remains note, a subset of pulmonary LC NECs exhibits a
under debate, the biologics sunitinib and molecular profile more similar to that of non-
everolimus have recently shown promise in SCLCs (NSCLCs) than to that of SCLCs, suggest-
managing advanced-stage NETs.59,61,69–72 ing that such cases may be more effectively
At this time, optimal therapy for G3 GEP- managed by NSCLC protocols.10,47,78,79,82–88
Standardized Nomenclature for Neuroendocrine Neoplasms 141

ANCILLARY TESTING IN THE CLASSIFICATION delineation of G3 GEP-NETs and their conceptual


OF NEUROENDOCRINE NEOPLASMS counterparts in other organ systems. G3 panNETs,
for instance, share frequent alterations in MEN1
The molecular and IHC profiles of NETs and NECs (21%) and ATRX or DAXX (approximately 30%)
are essentially mutually exclusive, reinforcing the with G1/G2 panNETs, and generally lack the RB/
dichotomous NEN paradigm. These ancillary TP53 mutations associated with pan-
studies can be leveraged in diagnostically chal- NECs.31,32,34,89 Similarly, emerging evidence sug-
lenging cases and, in some instances, may offer gests that highly proliferative carcinoids of the
prognostic insights or clarify therapeutic op- lung lack the RB/TP53 alterations or broader
tions.7–10,12–14,89 Here, we briefly review key facets genomic instability seen in SCLC, supporting their
of ancillary testing (summarized in Table 1), with classification as carcinoids (NETs), rather than
the pancreas and lungs as prototypes. NECs.10,45,47,50–52

MOLECULAR TESTING Molecular Profile of Neuroendocrine


Molecular Profile of Neuroendocrine Tumors Carcinomas
Broadly speaking, NETs exhibit relative genomic The genomic landscape of NECs, particularly
stability, with significantly lower tumor mutational those with SC morphology, is classically domi-
burden and copy number alterations in compari- nated by alterations in TP53 and
son to NECs.10,90–93 Both lung carcinoids and pan- RB.31,32,34,47,83–92,108,113,114 Among panNECs, for
NETs are classically associated with loss of instance, molecular and IHC surrogate studies
chromosome 11q (which harbors the MEN1 locus), have identified TP53 mutations and RB alterations
though other cytogenetic abnormalities, including (encompassing both mutations and copy number
gains of 17q, 7q, and 20q and losses of 6q and alterations) in up to 95% and 74% of cases,
11p in panNETs, have also been described.12,93–96 respectively; in contrast, such alterations are rarely
At the gene-specific level, sporadic panNETs are encountered in panNETs.31,32,34,89 Mutations in a
closely associated with alterations in MEN1, which variety of other driver genes, including APC,
codes for a chromatin remodeling protein (menin): KRAS, and BRAF, are also described, and MYC
mutations have been reported in up to nearly two- and KDM5A gains/amplifications have been re-
thirds of cases.96–99 In addition, molecular and IHC ported in approximately half of all panNECs.31,34
studies have demonstrated mutually exclusive SCLCs are likewise notable for high rates of con-
mutations in either DAXX or ATRX, two genes current RB1 and TP53 loss, with reported fre-
that govern histone interactions, in up to 43% of quencies ranging from 24% to >90% and 64% to
panNETs; loss of DAXX/ATRX expression leads nearly 100%, respectively.92,108,113,114 As in lung
to alternative lengthening of telomeres (ALT), a carcinoids, alterations in chromatin-remodeling
senescence escape mechanism.12,96,97,100–104 genes occur in approximately half of all SCLCs,
Other processes/pathways implicated in panNET though it is postulated that their tumorigenic impact
tumorigenesis include mTOR signaling (eg, may be overshadowed by that of RB1 and TP53
TSC1/2, PTEN), histone modification/chromatin losses.91 SCLCs also exhibit MYC amplifications
remodeling (SETD2, MLL3), and DNA damage and frequent mutations in NOTCH and KMT2D;
repair (MUTYH, CHEK2, BRCA2).96,97 Of note, the latter is associated with improved prog-
several studies have linked DAXX and ATRX muta- nosis.91,92,113,114 Interestingly, recent studies have
tions to more aggressive tumor behavior and delineated multiple SCLC subgroups based on
poorer prognosis.12,96,101–106 dominant transcription factor expression profiles,
In contrast to panNETs, lung carcinoids largely including ASCL1 (accounting for some 70% of
lack consistent, “hallmark” mutations, though SCLCs), NEUROD1, POU2F3, and YAP1.115–118
nearly half harbor alterations in genes governing Emerging evidence suggests that these subtypes
chromatin remodeling pathways.91,107–109 Of these, exhibit unique clinical profiles, with YAP1- and
MEN1 alterations are most frequent, with rates of ASCL1-dominant cases associated with the most
up to w13% across several recent studies; muta- and least favorable outcomes, respectively.118
tions in PSIP, EIF1AX, KMT2, and ARID1A are Similarly, molecular profiling of pulmonary LC
also described.91,93,107–112 Though the evidence is NECs has delineated two key subtypes, first re-
somewhat mixed, MEN1 mutations appear to occur ported in a key study by Rekhtman and col-
more commonly in ACs, where they are associated leagues.47 The first of these, SCLC-like,
with poorer prognosis.108,109,111,112 constitutes approximately 36%-40% of cases
Molecular profiling and associated IHC-based and is characterized by dual mutations/losses in
studies have likewise been instrumental in the TP53 and RB1, often in conjunction with MYCL
142 Jafari et al

amplification and/or other SCLC-type alterations. peptides), staining for peptide hormones, such
In contrast, the NSCLC-like subtype, comprising as insulin and glucagon, may occasionally be indi-
approximately 56% of pulmonary LC NECs, lacks cated in the appropriate clinical context.13
concurrent TP53/RB1 loss and often exhibits mu-
tations in genes classically associated with b. Determining the origin of NEN metastases of
NSCLCs (particularly adenocarcinoma), such as uncertain primary. In approximately one-tenth
STK11, KRAS, and KEAP1.47,83,84,86,87 Compara- of metastatic NEN cases, the site of the primary
ble rates of TP53/RB co-alterations have been re- malignancy cannot be determined; IHC studies
ported among extrapulmonary LC NECs as well, may offer some clarity, as highlighted below,
though available data are quite limited.87 These though the specificity and/or sensitivity of
distinctions may have important clinical ramifica- many markers is often greater in NETs than in
tions, with RB-wild-type/NSCLC-like tumors NECs.13,119,123–126
potentially benefitting from NSCLC-type i. Thyroid tissue transcription factor-1 (TTF-1):
management.10,47,78,79,82–88 Highly specific for lung carcinoids (both AC
and TC) and thyroid NETs (namely, medullary
IMMUNOHISTOCHEMICAL STUDIES thyroid carcinoma [MTC]), with positivity for
calcitonin or CEA favoring a diagnosis of
IHC plays several valuable roles in the work-up of MTC.13,14,119,123 In contrast, among NECs,
NENs, as outlined below. TTF-1 is expressed not only by tumors of pul-
monary origin, but also by approximately
a. Establishing neuroendocrine differentiation. one-third of extrapulmonary NECs.13,123
Epithelial NENs are characterized by a dual ii. CDX2: Classically associated with
neural and epithelial phenotype. The neural intestinal-type differentiation, and particu-
aspect encompasses the production of peptide larly sensitive for tumors originating in the
hormones (eg, insulin and glucagon) or mid-gut.13,14,123 Notably, some 13%-16%
biogenic amines (eg, serotonin), as well as of panNENs stain with CDX2, whereas
expression of “classic” neuroendocrine expression in pulmonary NENs is exceed-
markers, such as synaptophysin (more sensi- ingly rare.13,14,123,127,128
tive) and chromogranin A (more specific). These iii. Additional stains: CK7 is expressed in
markers, particularly chromogranin, may be approximately 40% of lung carcinoids and
lost in NECs; in such cases, a second-line is typically negative in GEP-NENs, whereas
marker, eg, CD56, may be considered, though CK20 positivity is seen in approximately
these are often quite nonspecific.7,13,14,119 Of 25% of GEP-NETs.123 Emerging markers
note, relatively recent additions to the NEN include ISL-1, highly sensitive for panNETs,
IHC toolkit include insulinoma-associated pro- though not particularly specific, and clus-
tein 1 (INSM1), which is highly specific for terin, which exhibits strong expression in
NENs across a variety of anatomic sites, and NETs outside of the jejunum and
SSTR2A/5, which is significantly more likely to ileum.13,14,119,129,130
be expressed in GEP-NETs (particularly in c. Assessing proliferative activity. Ki67 IHC plays a
low-grade tumors and/or low-stage disease) valuable role in NEN grading, particularly in the
than in GEP-NECs.7,13,62,120–122 GEP tract; see discussion below.7,8,15–18
In addition to these neural markers, epithelial d. Distinguishing between NET and NEC. As dis-
NENs classically express low-molecular weight cussed above, high-grade NETs and SC/LC
keratins (LMWKs), eg, CK8 and CK18, and as NECs may overlap morphologically, creating a
such stain with a variety of broad-spectrum/pan- diagnostic challenge. In tumors of pancreatic
keratin markers, including CAM5.2 and AE1/AE3; origin, loss of MEN1 and/or DAXX/ATRX sup-
NECs, particularly SC NECs, may exhibit dot-like ports a diagnosis of panNET; across anatomic
staining.7,8,13,14 CK7 and CK20 are typically nega- sites, Rb1 loss or aberrant p53 expression favors
tive, though site-specific exceptions do occur.13 In a diagnosis of NEC.7,10,13,31,89,97,98 Among GEP-
the rare event of a keratin-negative NET, negativity NENs, SSTR2A/5 expression favors a diagnosis
for tyrosine hydroxylase (highly sensitive for pheo- of NET over NEC.7,13,62
chromocytoma and, to a lesser extent, paragan-
glioma) or S100 (to demonstrate the absence of PROLIFERATIVE ACTIVITY: THE CRUX OF
sustentacular cells) may help to exclude a non- NEUROENDOCRINE NEOPLASM GRADING
epithelial NEN.13,14
Though the vast majority of NENs are nonfunc- Across organ systems, proliferative activity has
tional (ie, do not produce biologically active emerged as a powerful common denominator for
Standardized Nomenclature for Neuroendocrine Neoplasms 143

Box 2
Approaches to evaluation of Ki-67 LI within proliferation “hotspots”
"Eyeballing"(gestalt impression; no counting/assessment of discrete cells):
 Benefits: highly efficient (w1 min/slide); no additional resources required
 Drawbacks: poor interobserver agreement/accuracy
Manual counting at microscope:
 Benefits: highly accurate; no additional resources required
 Drawbacks: time-intensive (w6 min/slide); poor interobserver agreement
Manual counting using printed micrograph:
 Benefits: most accurate method, with highest interobserver agreement
 Drawbacks: time-intensive (w8 min/slide); requires microscope fitted with camera and access to high-
quality printer
Digital image analysis (DIA):
 Benefits: increasingly accurate with continued advances in software; relatively efficient (w3–5 min/
slide)
 Drawbacks: requires investment in slide scanning technology and image analysis software; persistent
concerns regarding under-/over-counting and risk of software failure; current iterations typically
require pathologist to screen slides and identify hotspots, raising concerns for inter-observer vari-
ability

NEN classification: it is the sole parameter for that evaluating metastatic panNETs via prolifera-
GEP-NET grading and one of just two parameters tive “hotspots” rather than a whole-slide average
(in conjunction with necrosis) for thoracic and head generated grades that more accurately correlated
and neck NET/carcinoid grading. Likewise, NECs with prognosis. At present, the WHO endorses the
are defined based on proliferative activity and hotspot approach, with grading typically based on
histology.7,8,15–17 the number of mitoses per 2 mm2 (approximately
Proliferative activity can be assessed via either 10 HPFs) or percent Ki67 positivity across at least
mitotic count or Ki67 LI. Ki67 IHC was initially 500 hotspot cells.15,16
reserved for biopsies too small for a meaningful
mitotic count (ie, less than 2 mm2 of tissue, or MITOTIC COUNT
the equivalent of 10 standard high powered fields
[HPFs]); however, in a 2010 survey, nearly half of Given the prognostic significance of specific NET
queried pathologists supported incorporation of grades, an accurate, reproducible count is of
Ki67 into routine NEN assessment.131 Today, paramount importance. To this end, the WHO
both Ki67 and mitotic count are included in the criteria specify that NEN mitoses be counted
WHO grading parameters for GEP-NENs, and per 2 mm2, which equates to approximately 10
Ki67 IHC is recognized as a valuable adjunct HPFs using most standard contemporary micro-
(though not a formal parameter) in the work-up scopes. This approach sidesteps the potential
and classification of lung NENs.8,15,16 Among for inter-microscope variability, a concern when
NETs, mitotic count and Ki67 LI exhibit grade- count cutoffs are defined on a per-HPF ba-
discordance in approximately a quarter to a third sis.8,138–140 Mitotic counts near a grade cutoff
of cases; in such instances, the higher grade (usu- should prompt evaluation of at least three hot-
ally associated with the Ki67 LI rather than mitotic spots, with grade based on an average count.15
count) is assigned, as this appears to more accu- Of note, mitotic figures may be difficult to distin-
rately reflect tumor behavior.16,132,133 guish from lymphocytes or from pyknotic or
Among NENs, as in other tumor types, there hyperchromatic nuclei; as such, the WHO Blue
may be substantial variability in proliferative activ- Books offer guidelines for identification of
ity within a single neoplasm and, indeed, on a sin- mitoses, such as the presence of basophilic cyto-
gle slide. This heterogeneity raises the possibility plasm and hairy chromosomal projec-
of sampling error and, by extension, inaccurate tions.15,141,142 In addition, emerging evidence
grading.134–137 As such, multiple approaches to suggests that the mitosis-specific IHC marker
the evaluation of mitoses and Ki67 have been pro- phosphohistone H3 may be a valuable adjunct
posed; Yang and colleagues136 reported in 2011 in evaluating NEN mitotic counts.141,142
144 Jafari et al

KI67 LABELING INDEX shifting classification parameters, and the rise of


ancillary testing–and seem likely to do so for de-
Optimal assessment of Ki67 within a given hotspot cades to come.
remains an area of active discussion. Approaches
include “eyeballing” (gestalt impression of Ki67 LI,
without counting individual cells) and manual CLINICS CARE POINTS
counting, either at the microscope or using a
printed photomicrograph. A study by Reid and col-
leagues143 in 2015 identified manual counting of
photomicrographs as the most accurate, albeit  Neuroendocrine neoplasms (NENs) can be
relatively time-intensive (approximately 8 min per divided into neuroendocrine tumors (NETs)
slide), method. Recently, digital image analysis and neuroendocrine carcinomas (NECs),
each with a distinct molecular, immunohisto-
(DIA) has emerged as a promising alternative,
chemical, and clinical profile. NETs are rela-
though further validation and discussion are tively indolent, though prognosis worsens
necessary.144–147 Benefits and drawbacks of in tandem with increasing grade. In contrast,
each approach are outlined in Box 2. NECs are high-grade by default and typically
It should be emphasized that although Ki67 carry a dismal prognosis.
plays a key role in the WHO’s grading criteria for
 Resection is the cornerstone of NET treat-
GEP-NENs, this is not the case across anatomic
ment; approaches to disseminated disease
sites.15,17 In the lung, for instance, Ki67 is not include somatostatin analogues, peptide re-
included in the WHO’s formal grading parameters; ceptor radionuclide therapy, and biologic
this reflects longstanding concerns about overlap- therapies (eg, sunitinib and everolimus). He-
ping Ki67 ranges between successive diagnostic patic metastases may be managed via de-
categories and potential discordance when diag- bulking and liver-directed therapy, where
noses based on Ki67 versus mitotic count are appropriate.
compared.15,18,148 However, in light of growing ev-  Platinum-based chemotherapy and radio-
idence that Ki67 LI in lung NENs correlates signif- therapy is the bedrock of small-cell lung car-
icantly with prognosis, the use of Ki67 as an cinoma (SCLC) management, with the
alternative to mitotic count is now under discus- addition of immune checkpoint inhibitors in
sion.18,148–151 In addition, there is evidence to sup- extensive-stage disease; resection is generally
port the use of Ki67 as a complement to mitotic considered only in very early stage disease. In
count in technically challenging cases, such as contrast, early-stage pulmonary large-cell
carcinoid tumor biopsies that exhibit considerable (LC) NECs appear relatively amenable to
crush artifact, which may not only obscure mitoses resection and adjuvant therapy, whereas
more advanced disease is typically treated
but also raise the morphologic differential of
with platinum-based chemotherapy. Of
SCLC.10,18,152 note, there is a paucity of data regarding
treatment of extrapulmonary SC and LC
SUMMARY NECs, which have typically been managed
similarly to their counterparts in the lung.
In the years since Siegfried Oberndorfer first  Molecular profiling has enabled a more
described the seemingly indolent, histologically nuanced characterization of NENs, revealing
unprepossessing Karzinoid Tumoren of the small at least two subtypes of lung LC NECs (SCLC-
bowel, NENs have emerged as an ever more like and NSCLC-like) as well as at least four
nuanced clinicopathologic domain, encompassing subtypes of SCLC. These subtypes appear to
a disparate array of morphologies and behaviors. be clinically distinct, raising the possibility
In this article, we have explored several key unify- of more tailored management: for instance,
NSCLC-like LC NEC may benefit from an
ing threads, including the presence of two histo-
NSCLC-type approach.
logic tiers and their distinct clinicopathologic
profiles; the role of ancillary testing in corrobo-  An emerging group of NENs is characterized
rating and clarifying current grading schemes; by relatively well-differentiated histology
and the primacy of proliferative activity in NET and high proliferative activity (typically be-
grading. These themes take on heightened signif- tween 20% and 55%). Tumor behavior ap-
pears to be intermediate between that of
icance in light of ongoing efforts to standardize
G2 NETs and NECs, with less robust response
and harmonize NEN classification across organ to platinum-based chemotherapy than
systems. Just as importantly, they have brought NECs. Optimal clinical management for these
stability and coherence to the century-old NEN “G3 NETs” remains under investigation.
narrative, weathering nomenclature changes,
Standardized Nomenclature for Neuroendocrine Neoplasms 145

CONFLICT OF INTEREST what can brown do for you? Hum Pathol 2020;96:
8–33.
The authors have no funding or conflicts of inter- 14. Duan K, Mete O. Algorithmic approach to neuroen-
est to declare. docrine tumors in targeted biopsies: practical ap-
plications of immunohistochemical markers.
REFERENCES Cancer Cytopathol 2016;124(12):871–84.
15. WHO Classification of Tumours Editorial Board.
1. Oberndorfer S. Karzinoide tumoren des dunn- Thoracic tumours. 5th ed. Lyon: IARC Press; 2022.
darms. Frankf Z Pathol 1907;1:426–32. 16. WHO Classification of Tumours Editorial Board.
2. Modlin IM, Shapiro MD, Kidd M. Siegfried Obern- Digestive system tumours. 5th ed. Lyon: IARC
dorfer: origins and perspectives of carcinoid tu- Press; 2019.
mors. Hum Pathol 2004;35(12):1440–51. 17. WHO Classification of Tumours Editorial Board.
3. Modlin IM, Champaneria MC, Bornschein J, et al. Head and neck tumours. 5th ed. Lyon: IARC Press;
Evolution of the diffuse neuroendocrine system: 2022.
clear cells and cloudy origins. Neuroendocrinology 18. Pelosi G, Rindi G, Travis WD, et al. Ki-67 antigen in
2006;84(2):69–82. lung neuroendocrine tumors: unraveling a role in
4. Klöppel G. Oberndorfer and his successors: from clinical practice. J Thorac Oncol 2014;9(3):273–84.
carcinoid to neuroendocrine carcinoma. Endocr 19. Dasari A, Shen C, Halperin D, et al. Trends in the
Pathol 2007;18(3):141–4. incidence, prevalence, and survival outcomes in
5. Asa SL, Lloyd RV, Tischler AS. Neuroendocrine patients with neuroendocrine tumors in the United
neoplasms: historical background and terminol- States. JAMA Oncol 2017;3(10):1335–42.
ogies. In: Asa SL, La Rosa S, Mete O, editors. 20. Nagtegaal ID, Odze RD, Klimstra D, et al. The 2019
The spectrum of neuroendocrine neoplasia: a prac- WHO classification of tumours of the digestive sys-
tical approach to diagnosis, classification and ther- tem. Histopathology 2020;76(2):182–8.
apy. Cham, Switzerland: Springer International 21. Assarzadegan N, Montgomery E. What is new in
Publishing; 2021. p. 1–14. the 2019 World Health Organization (WHO) Classi-
6. Gosney J. Identification, morphology, and secretory fication of Tumors of the Digestive System: review
products of the pulmonary endocrine system. In: Pul- of selected updates on neuroendocrine neo-
monary endocrine pathology: endocrine cells and plasms, appendiceal tumors, and molecular
endocrine tumours of the lung. Oxford (United testing. Arch Pathol Lab Med 2020;145(6):664–77.
Kingdom): Butterworth-Heinemann Ltd; 1992. p. 6–24. 22. Mete O, Wenig BM. Update from the 5th edition of
7. Rindi G, Mete O, Uccella S, et al. Overview of the the World Health Organization Classification of
2022 WHO classification of neuroendocrine neo- Head and Neck Tumors: overview of the 2022
plasms. Endocr Pathol 2022;33(1):115–54. WHO Classification of Head and Neck Neuroendo-
8. WHO Classification of Tumours Editorial Board. crine Neoplasms. Head Neck Pathol 2022;16(1):
Endocrine and neuroendocrine tumours. 5th edi- 123–42.
tion. Lyon: IARC Press; 2022. 23. Xue Y, Reid MD, Pehlivanoglu B, et al. Morphologic
9. Rindi G, Klimstra DS, Abedi-Ardekani B, et al. variants of pancreatic neuroendocrine tumors: clin-
A common classification framework for neuroendo- icopathologic analysis and prognostic stratifica-
crine neoplasms: an International Agency for tion. Endocr Pathol 2020;31(3):239–53.
Research on Cancer (IARC) and World Health Or- 24. Kim H, An S, Lee K, et al. Pancreatic high-grade
ganization (WHO) expert consensus proposal. neuroendocrine neoplasms in the Korean popula-
Mod Pathol 2018;31(12):1770–86. tion: a multicenter study. Cancer Res Treat 2020;
10. Rekhtman N. Lung neuroendocrine neoplasms: 52(1):263–76.
recent progress and persistent challenges. Mod 25. Soga J, Tazawa K. Pathologic analysis of carci-
Pathol 2022;35(1):36–50. noids: histologic re-evaluation of 62 cases. Cancer
11. Basturk O, Yang Z, Tang LH, et al. The high-grade 1971;28(4):990–8.
(WHO G3) pancreatic neuroendocrine tumor cate- 26. WHO Classification of Tumours Editorial Board.
gory is morphologically and biologically heteroge- WHO classification of tumours of the digestive sys-
nous and includes both well differentiated and tem. 4th ed. Lyon: IARC Press; 2010.
poorly differentiated neoplasms. Am J Surg Pathol 27. Capella C, Heitz PU, Höfler H, et al. Revised clas-
2015;39(5):683–90. sification of neuroendocrine tumours of the lung,
12. Tirosh A, Kebebew E. Genetic and epigenetic alter- pancreas and gut. Virchows Archiv A Pathol Anat
ations in pancreatic neuroendocrine tumors. 1995;425(6).
J Gastrointest Oncol 2020;11(3):567–77. 28. Hochwald S, Sui Z, Conlon K, et al. Prognostic fac-
13. Bellizzi AM. Immunohistochemistry in the diagnosis tors in pancreatic endocrine neoplasms: an anal-
and classification of neuroendocrine neoplasms: ysis of 136 cases with a proposal for low-grade
146 Jafari et al

and intermediate-grade groups. J Clin Oncol 2002; proliferation and back. Endocr Pathol 2014;25(2):
20(11):2633–42. 193–8.
29. Sorbye H, Welin S, Langer SW, et al. Predictive and 41. Lithgow K, Venkataraman H, Hughes S, et al. Well-
prognostic factors for treatment and survival in 305 differentiated gastroenteropancreatic G3 NET: find-
patients with advanced gastrointestinal neuroendo- ings from a large single centre cohort. Sci Rep
crine carcinoma (WHO G3): the NORDIC NEC 2021;11(1):17947.
study. Ann Oncol 2013;24(1):152–60. 42. Konukiewitz B, Jesinghaus M, Steiger K, et al.
30. Sorbye H, Strosberg J, Baudin E, et al. Gastroenter- Pancreatic neuroendocrine carcinomas reveal a
opancreatic high-grade neuroendocrine carci- closer relationship to ductal adenocarcinomas
noma. Cancer 2014;120(18):2814–23. than to neuroendocrine tumors G3. Hum Pathol
31. Hijioka S, Hosoda W, Matsuo K, et al. RB loss and 2018;77:70–9.
KRAS mutation are predictors of the response to 43. WHO Classification of Tumours Editorial Board.
platinum-based chemotherapy in pancreatic WHO classification of tumours of endocrine organs.
neuroendocrine neoplasm with grade 3: a Japa- 4th ed. Lyon: IARC Press; 2017.
nese multicenter pancreatic NEN-G3 study. Clin 44. Rindi G, Klersy C, Albarello L, et al. Competitive
Cancer Res 2017;23(16):4625–32. testing of the WHO 2010 versus the WHO 2017
32. Tang LH, Basturk O, Sue JJ, et al. A practical grading of pancreatic neuroendocrine neoplasms:
approach to the classification of WHO grade 3 data from a large international cohort study. Neuro-
(G3) well-differentiated neuroendocrine tumor endocrinology 2018;107(4):375–86.
(WD-NET) and poorly differentiated neuroendo- 45. Vivero M, Sholl L. Borderline” neuroendocrine car-
crine carcinoma (PD-NEC) of the pancreas. Am J cinomas of the lung are clinically and genomically
Surg Pathol 2016;40(9):1192–202. distinct from large-cell neuroendocrine carcinoma
33. Tang LH, Untch BR, Reidy DL, et al. Well-differenti- [abstract only]. Mod Pathol 2016;29:467–88.
ated neuroendocrine tumors with a morphologically 46. Megyesi M, Berta M, Khoor A. Endobronchial
apparent high-grade component: a pathway large-cell neuroendocrine carcinoma. Pathol Oncol
distinct from poorly differentiated neuroendocrine Res 2003;9(3):198–200.
carcinomas. Clin Cancer Res 2016;22(4):1011–7. 47. Rekhtman N, Pietanza MC, Hellmann MD, et al.
34. Venizelos A, Elvebakken H, Perren A, et al. The mo- Next-generation sequencing of pulmonary large-
lecular characteristics of high-grade gastroentero- cell neuroendocrine carcinoma reveals small-cell
pancreatic neuroendocrine neoplasms. Endocr carcinoma-like and non-small-cell carcinoma-like
Relat Cancer 2021;29(1):1–14. subsets. Clin Cancer Res 2016;22(14):3618–29.
35. Heetfeld M, Chougnet CN, Olsen IH, et al. Charac- 48. Inafuku K, Yokose T, Ito H, et al. Two cases of lung
teristics and treatment of patients with G3 gastro- neuroendocrine carcinoma with carcinoid
enteropancreatic neuroendocrine neoplasms. morphology. Diagn Pathol 2019;14(1):104.
Endocr Rel Cancer 2015;22(4):657–64. 49. Hermans BCM, Derks JL, Moonen L, et al. Pulmo-
36. Shi H, Chen L, Zhang Q, et al. Concordance be- nary neuroendocrine neoplasms with well differen-
tween the Ki-67 index cutoff value of 55% and dif- tiated morphology and high proliferative activity:
ferentiation in neuroendocrine tumor and illustrated by a case series and review of the litera-
neuroendocrine carcinoma in grade 3 pancreatic ture. Lung Cancer 2020;150:152–8.
neuroendocrine neoplasms. Pancreas 2020; 50. Zhang Y, Wang W, Hu Q, et al. Clinic and genetic
49(10):1378–82. similarity assessments of atypical carcinoid, neuro-
37. Vélayoudom-Céphise FL, Duvillard P, Foucan L, endocrine neoplasm with atypical carcinoid
et al. Are G3 ENETS neuroendocrine neoplasms morphology and elevated mitotic count and large-
heterogeneous? Endocr Relat Cancer 2013;20(5): cell neuroendocrine carcinoma. BMC Cancer
649–57. 2022;22(1):321.
38. Jann H, Kayser A, Wiedenmann B, et al. Treatment 51. Sazonova O, Manem V, Orain M, et al. Transcrip-
outcomes of patients with G3 neuroendocrine neo- tomic data helps refining classification of pulmo-
plasms [abstract only]. J Clin Oncol 2020; nary carcinoid tumors with increased mitotic
38(4_suppl):622. counts. Mod Pathol 2020;33(9):1712–21.
39. Raj N, Valentino E, Capanu M, et al. Treatment 52. Quinn AM, Chaturvedi A, Nonaka D. High-grade
response and outcomes of grade 3 pancreatic neuroendocrine carcinoma of the lung with carci-
neuroendocrine neoplasms based on morphology: noid morphology: a study of 12 cases. Am J Surg
well differentiated versus poorly differentiated. Pathol 2017;41(2):263–70.
Pancreas 2017;46(3):296–301. 53. Nicholson SA, Beasley MB, Brambilla E, et al.
40. La Rosa S, Sessa F. High-grade poorly differenti- Small-cell lung carcinoma (SCLC): a clinicopatho-
ated neuroendocrine carcinomas of the gastroen- logic study of 100 cases with surgical specimens.
teropancreatic system: from morphology to Am J Surg Pathol 2002;26(9):1184–97.
Standardized Nomenclature for Neuroendocrine Neoplasms 147

54. Travis WD. Advances in neuroendocrine lung tu- neuroendocrine tumors: an updated review. J Clin
mors. Ann Oncol 2010;21:vii65–71. Med 2021;10(6):1267.
55. Travis WD. Update on small-cell carcinoma and its 69. Wong MH, Chan DL, Lee A, et al. Systematic review
differentiation from squamous cell carcinoma and and meta-analysis on the role of chemotherapy in
other non-small-cell carcinomas. Mod Pathol advanced and metastatic neuroendocrine tumor
2012;25(1):S18–30. (NET). PLoS ONE 2016;11(6):e0158140.
56. Fahmy JN, Varsanik MA, Hubbs D, et al. Pancreatic 70. Liu IH, Kunz PL. Biologics in gastrointestinal and
neuroendocrine tumors: surgical outcomes and pancreatic neuroendocrine tumors. J Gastrointest
survival analysis. Am J Surg 2021;221(3):529–33. Oncol 2017;8(3):457–65.
57. Okereke IC, Taber AM, Griffith RC, et al. Outcomes 71. Yao JC, Shah MH, Ito T, et al. Everolimus for
after surgical resection of pulmonary carcinoid tu- advanced pancreatic neuroendocrine tumors.
mors. J Cardiothorac Surg 2016;11:35. N Engl J Med 2011;364(10).
58. Hu Y, Ye Z, Wang F, et al. Role of somatostatin re- 72. Faivre S, Niccoli P, Castellano D, et al. Sunitinib in
ceptor in pancreatic neuroendocrine tumor devel- pancreatic neuroendocrine tumors: updated
opment, diagnosis, and therapy. Front Endocrinol progression-free survival and final overall survival
(Lausanne) 2021;12:679000. from a phase III randomized study. Ann Oncol
59. Pavel M, Öberg K, Falconi M, et al. Gastroentero- 2017;28(2):339–43.
pancreatic neuroendocrine neoplasms: ESMO 73. AKP Ganti, Loo BW, Bassetti M, et al. Small-cell
Clinical Practice Guidelines for diagnosis, treat- lung cancer, version 2.2022, NCCN Clinical Prac-
ment and follow-up. Ann Oncol 2020;31(7):844–60. tice Guidelines in Oncology. J Natl Compr Canc
60. Starr JS, Sonbol MB, Hobday TJ, et al. Peptide re- Netw 2021;19(12):1441–64.
ceptor radionuclide therapy for the treatment of 74. Saltos A, Shafique M, Chiappori A. Update on the
pancreatic neuroendocrine tumors: recent insights. biology, management, and treatment of small-cell
Onco Targets Ther 2020;13:3545–55. lung cancer (SCLC). Front Oncol 2020;10:1074.
61. Baudin E, Caplin M, Garcia-Carbonero R, et al. 75. Stelwagen J, de Vries EGE, Walenkamp AME. Cur-
Lung and thymic carcinoids: ESMO Clinical Prac- rent treatment strategies and future directions for
tice Guidelines for diagnosis, treatment and extrapulmonary neuroendocrine carcinomas: a re-
follow-up. Ann Oncol 2021;32(4):439–51. view. JAMA Oncol 2021;7(5):759–70.
62. Popa O, Taban SM, Pantea S, et al. The new WHO 76. Walenkamp AME, Sonke GS, Sleijfer DT. Clinical
classification of gastrointestinal neuroendocrine tu- and therapeutic aspects of extrapulmonary small-
mors and immunohistochemical expression of so- cell carcinoma. Cancer Treat Rev 2009;35(3):
matostatin receptor 2 and 5. Exp Ther Med 2021; 228–36.
22(4):1–9. 77. Barnes H, See K, Barnett S, et al. Surgery for
63. Cakir M, Dworakowska D, Grossman A. Somato- limited-stage small-cell lung cancer. Cochrane
statin receptor biology in neuroendocrine and pitu- Database Syst Rev 2017;2017(4):CD011917.
itary tumours: part 2––clinical implications. J Cell 78. Ferrara MG, Stefani A, Simbolo M, et al. Large-cell
Mol Med 2010;14(11):2585–91. neuro-endocrine carcinoma of the lung: current
64. Rinke A, Müller HH, Schade-Brittinger C, et al. Pla- treatment options and potential future opportu-
cebo-controlled, double-blind, prospective, ran- nities. Front Oncol 2021;11:650293.
domized study on the effect of octreotide LAR in 79. Corbett V, Arnold S, Anthony L, et al. Management
the control of tumor growth in patients with meta- of large-cell neuroendocrine carcinoma. Front On-
static neuroendocrine midgut tumors: a report col 2021;11:653162.
from the PROMID Study Group. J Clin Oncol 80. Sakurai H, Asamura H. Large-cell neuroendocrine
2009;27(28):4656–63. carcinoma of the lung: surgical management.

65. Caplin ME, Pavel M, Cwik1a JB, et al. Lanreotide in Thorac Surg Clin 2014;24(3):305–11.
metastatic enteropancreatic neuroendocrine tu- 81. Raman V, Jawitz OK, Yang CFJ, et al. Outcomes for
mors. N Engl J Med 2014;371(3):224–33. surgery in large-cell lung neuroendocrine cancer.
66. Gupta S. Intra-arterial liver-directed therapies for J Thorac Oncol 2019;14(12):2143–51.
neuroendocrine hepatic metastases. Semin Inter- 82. Atieh T, Huang CH. Treatment of advanced-stage
vent Radiol 2013;30(1):28–38. large-cell neuroendocrine cancer (LCNEC) of the
67. Baere T de, Deschamps F, Tselikas L, et al. GEP- lung: a tale of two diseases. Front Oncol 2021;11.
NETs update: Interventional radiology: role in the 83. Derks JL, Leblay N, Thunnissen E, et al. Molecular
treatment of liver metastases from GEP-NETs. Eur subtypes of pulmonary large-cell neuroendocrine
J Endocrinol 2015;172(4):R151–66. carcinoma predict chemotherapy treatment
68. Camus B, Cottereau AS, Palmieri LJ, et al. Indica- outcome. Clin Cancer Res 2018;24(1):33–42.
tions of peptide receptor radionuclide therapy 84. George J, Walter V, Peifer M, et al. Integrative
(PRRT) in gastroenteropancreatic and pulmonary genomic profiling of large-cell neuroendocrine
148 Jafari et al

carcinomas reveals distinct subtypes of high-grade in pancreatic neuroendocrine tumors. Science


neuroendocrine lung tumors. Nat Commun 2018; 2011;331(6021):1199–203.
9(1):1048. 98. Corbo V, Dalai I, Scardoni M, et al. MEN1 in pancre-
85. Miyoshi T, Umemura S, Matsumura Y, et al. Genomic atic endocrine tumors: analysis of gene and protein
profiling of large-cell neuroendocrine carcinoma of status in 169 sporadic neoplasms reveals alter-
the lung. Clin Cancer Res 2017;23(3):757–65. ations in the vast majority of cases. Endocr Relat
86. Baine MK, Rekhtman N. Multiple faces of pulmo- Cancer 2010;17(3):771–83.
nary large-cell neuroendocrine carcinoma: update 99. Raj NP, Soumerai T, Valentino E, et al. Next-gener-
with a focus on practical approach to diagnosis. ation sequencing (NGS) in advanced well differen-
Transl Lung Cancer Res 2020;9(3). tiated pancreatic neuroendocrine tumors (WD
87. Saghaeiannejad Esfahani H, Vela CM, Chauhan A. pNETs): a study using MSK-IMPACT. J Clin Oncol
Prevalence of TP-53/RB-1 co-mutation in large-cell 2016;34(15_suppl):e15661.
neuroendocrine carcinoma. Front Oncol 2021;11: 100. Heaphy CM, de Wilde RF, Jiao Y, et al. Altered telo-
653153. meres in tumors with ATRX and DAXX mutations.
88. Zhuo M, Guan Y, Yang X, et al. The prognostic and Science 2011;333(6041):425.
therapeutic role of genomic subtyping by 101. Marinoni I, Kurrer AS, Vassella E, et al. Loss of
sequencing tumor or cell-free DNA in pulmonary DAXX and ATRX are associated with chromosome
large-cell neuroendocrine carcinoma. Clin Cancer instability and reduced survival of patients with
Res 2020;26(4):892–901. pancreatic neuroendocrine tumors. Gastroenter-
89. Yachida S, Vakiani E, White CM, et al. Small-cell ology 2014;146(2):453–60.e5.
and large-cell neuroendocrine carcinomas of the 102. Kim JY, Brosnan-Cashman JA, An S, et al. Alterna-
pancreas are genetically similar and distinct from tive lengthening of telomeres in primary pancreatic
well-differentiated pancreatic neuroendocrine tu- neuroendocrine tumors is associated with aggres-
mors. Am J Surg Pathol 2012;36(2):173–84. sive clinical behavior and poor survival. Clin Can-
90. van Riet J, van de Werken HJG, Cuppen E, et al. cer Res 2017;23(6):1598–606.
The genomic landscape of 85 advanced neuroen- 103. Singhi AD, Liu TC, Roncaioli JL, et al. Alternative
docrine neoplasms reveals subtype-heterogeneity lengthening of telomeres and loss of DAXX/ATRX
and potential therapeutic targets. Nat Commun expression predicts metastatic disease and poor
2021;12(1):4612. survival in patients with pancreatic neuroendocrine
91. Simbolo M, Mafficini A, Sikora K, et al. Lung neuro- tumors. Clin Cancer Res 2017;23(2):600–9.
endocrine tumours: deep sequencing of the four 104. Park JK, Paik WH, Lee K, et al. DAXX/ATRX and
World Health Organization histotypes reveals MEN1 genes are strong prognostic markers in
chromatin-remodelling genes as major players pancreatic neuroendocrine tumors. Oncotarget
and a prognostic role for TERT, RB1, MEN1 and 2017;8(30):49796–806.
KMT2D. J Pathol 2017;241(4):488–500. 105. Chan CS, Laddha SV, Lewis PW, et al. ATRX, DAXX
92. Clinical Lung Cancer Genome Project (CLCGP), or MEN1 mutant pancreatic neuroendocrine tu-
Network Genomic Medicine (NGM). A genomics- mors are a distinct alpha-cell signature subgroup.
based classification of human lung tumors. Sci Nat Commun 2018;9(1):4158.
Transl Med 2013;5(209):209ra153. 106. Heaphy CM, VandenBussche CJ. Prognostic bio-
93. Walch AK, Zitzelsberger HF, Aubele MM, et al. markers in pancreatic neuroendocrine tumors.
Typical and atypical carcinoid tumors of the lung Cancer Cytopathol 2021;129(11):841–3.
are characterized by 11q deletions as detected 107. Derks JL, Leblay N, Lantuejoul S, et al. New in-
by comparative genomic hybridization. Am J sights into the molecular characteristics of pulmo-
Pathol 1998;153(4):1089–98. nary carcinoids and large-cell neuroendocrine
94. Capurso G, Festa S, Valente R, et al. Molecular pa- carcinomas, and the impact on their clinical man-
thology and genetics of pancreatic endocrine tu- agement. J Thorac Oncol 2018;13(6):752–66.
mours. J Mol Endocrinol 2012;49(1):R37–50. 108. Simbolo M, Di Noia V, D’Argento E, et al. Exploring
95. Floridia G, Grilli G, Salvatore M, et al. Chromosomal the molecular and biological background of lung
alterations detected by comparative genomic hy- neuroendocrine tumours. J Thorac Dis 2019;
bridization in nonfunctioning endocrine pancreatic 11(Suppl 9):S1194–8.
tumors. Cancer Genet Cytogenet 2005;156(1): 109. Fernandez-Cuesta L, Peifer M, Lu X, et al. Frequent
23–30. mutations in chromatin-remodelling genes in pul-
96. Scarpa A, Chang DK, Nones K, et al. Whole- monary carcinoids. Nat Commun 2014;27(5):3518.
genome landscape of pancreatic neuroendocrine 110. Debelenko LV, Brambilla E, Agarwal SK, et al. Iden-
tumours. Nature 2017;543(7643):65–71. tification of MEN1 gene mutations in sporadic
97. Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, carcinoid tumors of the lung. Hum Mol Genet
and mTOR pathway genes are frequently altered 1997;6(13):2285–90.
Standardized Nomenclature for Neuroendocrine Neoplasms 149

111. Swarts DRA, Scarpa A, Corbo V, et al. MEN1 gene 125. Catena L, Bichisao E, Milione M, et al. Neuroendo-
mutation and reduced expression are associated crine tumors of unknown primary site: gold dust or
with poor prognosis in pulmonary carcinoids. misdiagnosed neoplasms? Tumori 2011;97(5):
J Clin Endocrinol Metab 2014;99(2):E374–8. 564–7.
112. Rossi G, Bertero L, Marchiò C, et al. Molecular al- 126. Scoazec JY, Couvelard A, Monges G, et al. Profes-
terations of neuroendocrine tumours of the lung. sional practices and diagnostic issues in neuroen-
Histopathology 2018;72(1):142–52. docrine tumour pathology: results of a prospective
113. George J, Lim J, Lang S, et al. Comprehensive one-year survey among French pathologists (the
genomic profiles of small-cell lung cancer. Nature PRONET study). Neuroendocrinology 2017;105(1):
2015;524(7563):47–53. 67–76.
114. Peifer M, Fernández-Cuesta L, Sos ML, et al. Inte- 127. Chan ES, Alexander J, Swanson PE, et al. PDX-1,
grative genome analyses identify key somatic CDX-2, TTF-1, and CK7: a reliable immunohisto-
driver mutations of small-cell lung cancer. Nat chemical panel for pancreatic neuroendocrine neo-
Genet 2012;44(10):1104–10. plasms. Am J Surg Pathol 2012;36(5):737–43.
115. Baine MK, Hsieh MS, Lai WV, et al. Small-cell lung 128. Erickson LA, Papouchado B, Dimashkieh H, et al.
carcinoma subtypes defined by ASCL1, NEUROD1, CDX2 as a marker for neuroendocrine tumors of
POU2F3, and YAP1: comprehensive immunohisto- unknown primary sites. Endocr Pathol 2004;15(3):
chemical and histopathologic characterization. 247–52.
J Thorac Oncol 2020;15(12):1823–35. 129. Agaimy A, Erlenbach-Wünsch K, Konukiewitz B,
116. Rudin CM, Poirier JT, Byers LA, et al. Molecular et al. ISL1 expression is not restricted to pancreatic
subtypes of small-cell lung cancer: a synthesis of well-differentiated neuroendocrine neoplasms, but
human and mouse model data. Nat Rev Cancer is also commonly found in well and poorly differen-
2019;19(5):289–97. tiated neuroendocrine neoplasms of extrapancre-
117. Borromeo MD, Savage TK, Kollipara RK, et al. ASCL1 atic origin. Mod Pathol 2013;26(7):995–1003.
and NEUROD1 reveal heterogeneity in pulmonary 130. Czeczok TW, Stashek KM, Maxwell JE, et al. Clus-
neuroendocrine tumors and regulate distinct genetic terin in neuroendocrine epithelial neoplasms:
programs. Cell Rep 2016;16(5):1259–72. absence of expression in a well-differentiated tu-
118. Qi J, Zhang J, Liu N, et al. Prognostic implications mor suggests a jejunoileal origin. Appl Immunohis-
of molecular subtypes in primary small-cell lung tochem Mol Morphol 2018;26(2):94–100.
cancer and their correlation with cancer immunity. 131. Klimstra DS, Modlin IR, Adsay NV, et al. Pathology
Front Oncol 2022;12:779276. reporting of neuroendocrine tumors: application of
119. Bellizzi AM. An algorithmic immunohistochemical the Delphic consensus process to the develop-
approach to define tumor type and assign site of ment of a minimum pathology data set. Am J
origin. Adv Anat Pathol 2020;27(3):114–63. Surg Pathol 2010;34(3):300–13.
120. Rooper L, Sharma R, LI Q, et al. INSM1 demon- 132. McCall CM, Shi C, Cornish TC, et al. Grading of
strates superior performance to the individual and well-differentiated pancreatic neuroendocrine tu-
combined use of synaptophysin, chromogranin mors is improved by the inclusion of both Ki67 pro-
and CD56 for diagnosing neuroendocrine tumors liferative index and mitotic rate. Am J Surg Pathol
of the thoracic cavity. Am J Surg Pathol 2017; 2013;37(11):1671–7.
41(11):1561–9. 133. van Velthuysen MLF, Groen EJ, van der Noort V,
121. Rosenbaum JN, Guo Z, Baus RM, et al. INSM1: a et al. Grading of neuroendocrine neoplasms: mito-
novel immunohistochemical and molecular marker ses and Ki-67 are both essential. Neuroendocri-
for neuroendocrine and neuroepithelial neoplasms. nology 2014;100(2–3):221–7.
Am J Clin Pathol 2015;144(4):579–91. 134. Jannink I, Risberg B, van Diest P, et al. Heteroge-
122. Juhlin CC, Zedenius J, Höög A. Clinical routine neity of mitotic activity in breast cancer. Histopa-
application of the second-generation neuroendo- thology 1996;29(5):421–8.
crine markers ISL2, INSM1, and secretagogin in 135. Verhoeven D, Bourgeois N, Derde M, et al. Com-
neuroendocrine neoplasia: staining outcomes and parison of cell growth in different parts of breast
potential clues for determining tumor origin. Endocr cancers. Histopathology 1990;17(6):505–9.
Pathol 2020;31(4):401–10. 136. Yang Z, Tang LH, Klimstra DS. Effect of tumor het-
123. Bellizzi A. Assigning site of origin in metastatic erogeneity on the assessment of Ki67 labeling in-
neuroendocrine neoplasms: a clinically significant dex in well-differentiated neuroendocrine tumors
application of diagnostic immunohistochemistry. metastatic to the liver: implications for prognostic
Adv Anat Pathol 2013;20(5):285–314. stratification. Am J Surg Pathol 2011;35(6):853–60.
124. Bergsland EK, Nakakura EK. Neuroendocrine tu- 137. Couvelard A, Deschamps L, Ravaud P, et al. Het-
mors of unknown primary: is the primary site really erogeneity of tumor prognostic markers: a repro-
not known? JAMA Surg 2014;149(9):889–90. ducibility study applied to liver metastases of
150 Jafari et al

pancreatic endocrine tumors. Mod Pathol 2009; compared with routine manual hot spot evaluation
22(2):273–81. of the Ki67 index. Appl Immunohistochem Mol Mor-
138. Cree IA, Tan PH, Travis WD, et al. Counting mitoses: phol 2021;29(7):499–505.
SI(ze) matters. Mod Pathol 2021;34(9):1651–7. 146. Hacking SM, Sajjan S, Lee L, et al. Potential pitfalls
139. Yigit N, Gunal A, Kucukodaci Z, et al. Are we count- in diagnostic digital image analysis: experience
ing mitoses correctly? Ann Diagn Pathol 2013; with Ki-67 and PHH3 in gastrointestinal neuroendo-
17(6):536–9. crine tumors. Pathol Res Pract 2020;216(3):
140. Facchetti F. A proposal for the adoption of a uni- 152753.
form metrical system for mitosis counting. Int J 147. Volynskaya Z, Mete O, Pakbaz S, et al. Ki67 quan-
Surg Pathol 2005;13(2):157–9. titative interpretation: insights using image anal-
141. Tracht J, Zhang K, Peker D. Grading and prognos- ysis. J Pathol Inform 2019;10:8.
tication of neuroendocrine tumors of the pancreas: 148. Walts AE, Ines D, Marchevsky AM. Limited role of
a comparison study of Ki67 and PHH3. Ki-67 proliferative index in predicting overall
J Histochem Cytochem 2017;65(7):399–405. short-term survival in patients with typical and atyp-
142. Kim MJ, Kwon MJ, Kang HS, et al. Identification of ical pulmonary carcinoid tumors. Mod Pathol 2012;
phosphohistone H3 cutoff values corresponding to 25(9):1258–64.
original WHO grades but distinguishable in well- 149. Zahel T, Krysa S, Herpel E, et al. Phenotyping of
differentiated gastrointestinal neuroendocrine tu- pulmonary carcinoids and a Ki-67-based grading
mors. Biomed Res Int 2018;2018:e1013640. approach. Virchows Arch 2012;460(3):299–308.
143. Reid MD, Bagci P, Ohike N, et al. Calculation of the 150. Grimaldi F, Muser D, Beltrami CA, et al. Partitioning
Ki67 index in pancreatic neuroendocrine tumors: a of bronchopulmonary carcinoids in two different
comparative analysis of four counting methodolo- prognostic categories by Ki-67 score. Front Endo-
gies. Mod Pathol 2015;28(5):686–94. crinol (Lausanne) 2011;2:20.
144. Tang L, Mithat G, Hedvat C, et al. Objective quan- 151. Skov B, Holm B, Erreboe A, et al. ERCC1 and
tification of the Ki67 proliferative index in neuroen- Ki67 in small-cell lung carcinoma and other
docrine tumors of the gastroenteropancreatic neuroendocrine tumors of the lung: distribution
system: a comparison of digital image analysis and impact on survival. J Thorac Oncol 2010;
with manual methods. Am J Surg Pathol 2012; 5(4):453–9.
36(12):1761–70. 152. Aslan D, Gulbahce H, Pambuccian S, et al. Ki-67
145. Lea D, Gudlaugsson EG, Skaland I, et al. Digital immunoreactivity in the differential diagnosis of pul-
image analysis of the proliferation markers Ki67 monary neuroendocrine neoplasms in specimens
and phosphohistone H3 in gastroenteropancreatic with extensive crush artifact. Am J Clin Pathol
neuroendocrine neoplasms: accuracy of grading 2005;123(6):874–8.

You might also like