Sei sulla pagina 1di 14

ORIGINAL ARTICLE

Pathology Reporting of Neuroendocrine Tumors:


Application of the Delphic Consensus Process to the
Development of a Minimum Pathology Data Set
David S. Klimstra, MD,* Irvin R. Modlin, MD, PhD,w N. Volkan Adsay, MD,z
Runjan Chetty, MD,y Vikram Deshpande, MD,J Mithat Gönen, PhD,z Robert T. Jensen, MD,#
Mark Kidd, PhD,w Matthew H. Kulke, MD, ** Ricardo V. Lloyd, MD, PhD, w w
Cesar Moran, MD,zz Steven F. Moss, MD,yy Kjell Oberg, MD,JJ Dermot O’Toole, MD,zz
Guido Rindi, MD,## Marie E. Robert, MD,*** Saul Suster, MD,w w w Laura H. Tang, MD, PhD,*
Chin-Yuan Tzen, MD, PhD,zzz Mary Kay Washington, MD,yyy Betram Wiedenmann, MD,JJJ
and James Yao, MDzzz

multidisciplinary team of physicians interested in NETs was


Abstract: Epithelial neuroendocrine tumors (NETs) have been assembled. At a group meeting, the participants discussed
the subject of much debate regarding their optimal classification. a series of ‘‘yes’’ or ‘‘no’’ questions related to the pathology of
Although multiple systems of nomenclature, grading, and NETs and the minimal data to be included in the reports. After
staging have been proposed, none has achieved universal accep- discussion, anonymous votes were taken, using the Delphic
tance. To help define the underlying common features of these principle that 80% agreement on a vote of either yes or no would
classification systems and to identify the minimal pathology data define a consensus. Questions that failed to achieve a consensus
that should be reported to ensure consistent clinical manage- were rephrased once or twice and discussed, and additional votes
ment and reproducibility of data from therapeutic trials, a were taken. Of 108 questions, 91 were answerable either yes or no
by more than 80% of the participants. There was agreement
From the From the Departments of *Pathology; zEpidemiology and about the importance of proliferation rate for tumor grading, the
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, landmarks to use for staging, the prognostic factors assessable
NY; Departments of wSurgery; ***Pathology, Yale University by routine histology that should be reported, the potential for
School of Medicine, New Haven, CT; zDepartment of Pathology, tumors to progress biologically with metastasis, and the current
Emory University, Atlanta, GA; yDepartment of Pathology,
Toronto General Hospital, Toronto, Ontario, Canada; JDepartment status of advanced immunohistochemical and molecular testing
of Pathology, Massachusetts General Hospital; **Department of for treatment-related biomarkers. The lack of utility of a variety
Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; of immunohistochemical stains and pathologic findings was
#National Institute of Diabetes, Digestive and Kidney Diseases also agreed upon. A consensus could not be reached for the
(RTJ), Bethesda, MD; wwDepartment of Pathology (RVL), Mayo remaining 17 questions, which included both minor points
Clinic, Rochester, MN; Departments of zzPathology; zzzGastro-
intestinal Medical Oncology, M.D. Anderson Cancer Center, related to extent of disease assessment and some major areas
Houston, TX; yyDepartment of Medicine, Rhode Island Hospital/ such as terminology, routine immunohistochemical staining for
Brown University, Providence, RI; JJDepartment of Medical general neuroendocrine markers, use of Ki67 staining to assess
Sciences, University of Upsalla, Upsalla, Sweden; zzDepartment of proliferation, and the relationship of tumor grade to degree of
Medicine, St. James’s Hospital and Trinity College, Dublin, Ireland;
##Department of Pathology and Laboratory Medicine, University of
differentiation. On the basis of the results of the Delphic voting,
Parma, Parma, Italy; wwwDepartment of Pathology, Medical College a minimum pathology data set was developed. Although there
of Wisconsin, Milwaukee, WI; zzzDepartment of Pathology, Cathay remains disagreement among experts about the specific classifi-
General Hospital, Taipei, Taiwan; yyyDepartment of Pathology, cation system that should be used, there is agreement about the
Vanderbilt Medical School, Nashville, TN; and JJJDepartment of fundamental pathology data that should be reported. Examina-
Internal Medicine, Charite Hospital, University of Berlin, Berlin,
Germany. tion of the areas of disagreement reveals significant opportunities
Supported by Novartis Corporation. for collaborative study to resolve unanswered questions.
David S. Klimstra, MD, Irvin R. Modlin, MD, PhD, Shared first
authorship. Key Words: neuroendocrine tumor, consensus, carcinoid tumor,
Correspondence: David S. Klimstra, MD, Surgical Pathology Service, Delphic, classification
Department of Pathology, Memorial Sloan-Kettering Cancer Center,
1275 York Avenue, New York, NY 10065 (e-mail: klimstrd@mskcc. (Am J Surg Pathol 2010;34:300–313)
org).
Supplemental digital content is available for this article. Direct URL
citations appear in the printed text and are provided in the HTML
and PDF versions of this article on the journal’s Web site
(www.ajsp.com).
Copyright r 2010 by Lippincott Williams & Wilkins
E pithelial neuroendocrine tumors (NETs) are a diverse
group of pathologically related neoplasms that can
arise in most epithelial organs of the body but are

300 | www.ajsp.com Am J Surg Pathol  Volume 34, Number 3, March 2010


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

particularly well-described in the lung, tubular gastro- The purpose of this workshop or meeting was to
intestinal tract, and pancreas. As the entity of ‘‘carcinoid bring together recognized experts in the field of neuroen-
tumor’’ was first proposed by Oberndorfer over 100 years docrine tumors, including pathologists and surgeons,
ago,56 the clinical and pathologic features of NETs have oncologists, and gastroenterologists, to (1) review existing
been described by many investigators, with most studies classification systems, (2) evaluate the published literature
focusing on subsets of tumors restricted to 1 organ or related to prognosis-related parameters or treatment-
organ system.30,40,46 For this reason, and also because the related parameters, (3) determine which pathologic para-
larger family of NETs is highly diverse in terms of origin, meters are currently (or likely will become) important to
mechanism of development, functional status, histologic predict prognosis and guide therapy, (4) decide what basic
patterns, and biologic behavior, many different diag- information related to diagnosis, grading, staging, and
nostic terms and classification systems have arisen. For prognosis should be included in pathology reports, and
example, ‘‘neuroendocrine tumor,’’ ‘‘carcinoid tumor,’’ (5) draft a checklist of the minimal pathology data set
‘‘endocrine neoplasm,’’ and ‘‘neuroendocrine carci- for NETs.
noma’’ have all been applied to small intestinal pri-
maries.6,9,47,58,80 In general, carcinoid tumors of the
tubular gastrointestinal tract have been separated from METHODS
pancreatic endocrine neoplasms.8–10,28 Site-specific grad- Physicians with dedicated expertise in the field of
ing, staging, and classification systems have been devel- NETs were identified. An effort was undertaken to recruit
oped by the World Health Organization (WHO) and the individuals from different countries and to include both
European Neuroendocrine Tumor Society (ENETS), and pathologists (12 participants) and clinicians (8 partici-
additional proposals are under development by the pants, including 2 surgeons, 2 gastroenterologists, and
American Joint Committee on Cancer (AJCC), and the 4 medical oncologists). Panel members were recruited
North American Neuroendocrine Tumor Society (NA- from the United States, Canada, Europe (Sweden, Germany,
NETS).9,10,21,28,75,76 Furthermore, multiple different in- Italy, and Ireland), and Asia. Participants in major
dividual classification schema have been published,22,30,45 organizations that have developed (or will develop soon)
and numerous studies have investigated potential prog- classification systems for NETs were selected, including
nostic factors based on morphology, immunophenotype, the WHO, the ENETS, the NANETS, the AJCC, and the
and molecular biology.7,11,17,31,35,44,66,74,99 Although all Cancer Committee of the College of American Patho-
of these different systems have merit and can be used logists. An effort was made to ensure balanced represen-
to stratify the biologic behavior of neuroendocrine tation of individual groups and institutions to provide a
tumors,14,24,29,30,64 the specific criteria differ between the breadth of input from clinical, scientific, and geographic
various anatomic sites, and translation between the classi- perspectives. A biostatistician was also included. A group
fication systems is not always possible.54 As promising meeting was held in Miami Beach, FL from February 6
new therapies arise for this family of tumors,41,61,96 it is to 8, 2009. Each participant was asked to prepare a
of increasing clinical relevance to carry out accurate brief presentation related to specific aspects of the topic,
classification and prognostication to properly tailor to collectively review the existing published data and to
treatment, not only for surgically resected primary tumors ensure that the ensuing discussions would be based on
but also for metastatic disease, from which only biopsy uniform background information.
specimens may be available. The lack of a single uniform The Delphic consensus process,19,25 was used to
system of nomenclature, grading, and staging for NETs achieve agreement on issues related to the analysis of
will hamper efforts to evaluate new therapies and to the pathology of NETs. Before the meeting, a series of
compare the results of published therapeutic trials.54 questions were developed and distributed to the group
A careful examination of the major published for review. Each question was formatted to be answer-
classification systems for NETs reveals considerable able as ‘‘yes’’ or ‘‘no.’’ The questions covered aspects of
overlap in the essential information used to derive the terminology, grading and staging, immunohistochemical
specific subgroups. Tumor size, extent of invasion, and evaluation, assessment of prognostic factors, and other
presence of nodal or distant metastases are well-accepted issues, all related to the information that should be
staging parameters for all primary sites.9,10,21,35,38,75,76 contained in standard pathology reports of NET speci-
The proliferative index has emerged as a fundamental mens. At the meeting, the group discussed each of the
grading characteristic that appears in most major questions to debate controversial issues and achieves
schema.1,5,6,33,62,63,93 Thus, there likely exists a compen- agreement where possible. Consistent with the Delphic
dium of basic information that should be collected for all process, some questions required modification to enable
neuroendocrine tumor specimens that would allow trans- general agreement to be reached. After the discussion
parent and accurate data comparisons. Whereas the stan- period, a formal vote was taken, and all participants were
dardization of the terminology and classification criteria for required to vote on each question. Each question was
all NETs of every anatomic site would be a laudable but read aloud, and anonymous voting was conducted using
likely elusive goal, the development of a ‘‘minimum data computerized electronic voting devices. The results of the
set’’ that could easily be translated into any chosen voting were immediately tabulated. The voting categories
classification system should be more easily attainable. are displayed in Supplementary (Table 1, Supplemental

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 301


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

Digital Content 1, http://links.lww.com/PAS/A49). Votes major recent classification systems (that of the EN-
of ‘‘Agree strongly’’ and ‘‘Agree with minor reservation’’ ETS,75,76 for example), and that to abandon the term
were considered ‘‘yes’’ votes; votes of ‘‘Disagree with at this point would require excessive reeducation (44%
major reservation (disagree moderately)’’ and ‘‘Disagree preferred to retain the term ‘‘tumor’’). The other termino-
strongly’’ were regarded as ‘‘no’’ votes. Consensus was logic issue that could not be resolved involves the use
defined to be achieved if at least 80% of the partici- of the term ‘‘carcinoma,’’ especially when applied to
pants voted ‘‘yes’’ or ‘‘no.’’ In the event of no immediate well-differentiated NETs that have displayed clinically
consensus, the topic was reopened for discussion, and a obvious malignant behavior. The WHO classification
second vote was then taken. If 80% agreement was not for small bowel and pancreatic NETs uses the term
attainable, further debate to allow modification of the ‘‘tumor’’ for organ-confined disease and ‘‘carcinoma’’ for
proposition was then undertaken and a third vote taken. otherwise identical neoplasms with metastases or gross
If a consensus was still not attainable, the question was local invasion.9,28,38 A number (69%) of the participants
deemed to have no agreement. The final phrasing of the considered that this distinction reflects differences in
questions and the result of the final vote on each question tumor stage rather than an inherent difference in tumor
were recorded. biology, as suggested by the use of ‘‘carcinoma.’’
Conversely, others argued for the use of the ‘‘carcinoma’’
RESULTS for all well-differentiated NETs. This discussion and
The complete list of Delphic questions along with the the lack of consensus on this issue amplified the wide
consensus answers are presented in Supplementary (Table 2, diversity of opinions about the implications of diagnostic
Supplemental Digital Content 2, http://links.lww.com/PAS/A50). terminology.
Of 108 questions, consensus was reached based on the
definition of 80% agreement for 91 (84%) of the Diagnostic Immunohistochemistry
questions. Most of the questions upon which there was Demonstration of neuroendocrine differentiation
no agreement (Supplementary Table 3, Supplementary by the use of specific immunohistochemical stains can
Digital Content 3, http://links.lww.com/PAS/A51) showed be helpful in the diagnosis of NETs.50 The vast majority
marked polarization of opinion, participants usually of well-differentiated NETs express 1 or more general
selecting a voting category to register either a ‘‘yes’’ or neuroendocrine markers (such as chromogranin A and
‘‘no’’ vote [rather than choosing ‘‘Agree with major synaptophysin), and it was agreed that the use of
reservation’’ or ‘‘Disagree with minor reservation (dis- immunohistochemistry for diagnostic purposes should
agree mildly),’’ votes that would have suggested no strong be recommended in a majority of cases, including in all
opinion on the question]. For only 1 question with no cases of biopsies of metastatic disease. There was no
agreement would the change of a single vote have allowed agreement that diagnostic immunohistochemistry should
consensus to be reached. A discussion of the general areas be mandated for all NETs, however. Some participants
of consensus and disagreement is presented below, divided (53%) considered that routine immunohistochemical
into broad topic areas. staining is not necessary to diagnose histologically typical
examples of well-differentiated NETs (carcinoid tumors)
Terminology of the ileum, appendix, and stomach (such as the multiple
The group did not attempt to develop a single tumors in patients with hypergastrinemia-associated
standard terminologic classification system for NETs, but neuroendocrine tumors) or certain pancreatic endocrine
did address limited specific issues. The terms ‘‘endocrine’’ tumors such as clinically functional insulinomas. The
and ‘‘neuroendocrine’’ are variously used for this family use of diagnostic immunohistochemistry for cases with
of tumors,4,36 and there is justification for either term. unusual or unclear histologic features was encouraged.
There was agreement that these 2 terms could be used The group agreed that the only 2 stains to be routinely
synonymously for differentiated neoplasms with epithelial recommended are chromogranin A and synaptophysin.
and (neuro)endocrine differentiation in the gastro- There was agreement that staining routinely for other
enteropancreatic system. It was also agreed that ‘‘carci- neuroendocrine markers including chromogranin B,
noid tumor’’ has become archaic and that it should be CD56 (neural cell adhesion molecule, N-CAM), CD57
avoided as the primary diagnostic term. However, there (leu7), and neuron-specific enolase was not indicated, and
was recognition that the term has become entrenched in that the specificity of these markers (especially neuron-
the lexicon, and as a practical matter it may still be specific enolase) was questionable. There was also
reasonable for the foreseeable future to reference this consensus that stains for keratins were not to be routinely
term (for example, parenthetically) in the pathology recommended, although most well-differentiated NETs
report to ensure fluid communication. There was con- do express keratins (in particular, cytokeratins 8 and 18,
siderable debate about the use of ‘‘tumor’’ versus detected by Cam5.250), and that p53 immunostaining
‘‘neoplasm’’ Although some participants pointed out also was not essential. The group further agreed that there
that all NETs are regarded to be neoplastic, justifying are only limited indications to carry out stains for specific
the designation (neuro)endocrine neoplasm (44% favored peptide hormones or bioamines, even when the clinical
this term), others argued that the (neuro)endocrine tumor history suggests the presence of a ‘‘functional’’ NET. As
terminology has become widely disseminated in several functional NETs are not defined by immunohistochemical

302 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

labeling, but rather by clinical symptoms and serology,28 will allow proper tumor staging to be carried out using
a statement about the functional nature of the tumor is any of the well-established systems. For all NETs, the
not needed in the pathology report. Furthermore, there size of the tumor (in 3 dimensions) should be reported. In
are no histologic features that define a functional NET the tubular gastrointestinal tract, the depth of maximal
and there is no difference in biologic behavior between invasion through the wall should be stated, using the
functional and nonfunctional NETs, independent of same landmarks as for the staging of exocrine carcinomas
other prognostic factors such as size, stage, and grade.30 of the corresponding locations.21,75,76 Reporting of the
Irrespective of the clinical syndrome or the bioactive maximal thickness of the tumor was felt to be unnecessary
agent produced, both types are NETs and should be however. For appendiceal NETs, invasion into the
regarded as a single entity. Limited peptide immunohisto- mesoappendix should be reported, and the extent (limited
chemistry could be done (eg, for insulin or gastrin) if there vs. extensive) should be documented. There was no
is a clinical indication to show the production of a specific agreement concerning the measurement of the amount
peptide in a functional tumor, such as in a patient with of mesoappendiceal invasion however. For pancreatic
multiple endocrine neoplasia type 1 (MEN1) with more NETs, the presence of extrapancreatic invasion should be
than 1 pancreatic NET. reported.
For patients presenting with metastatic disease Lymph node metastases should be reported, includ-
from an unknown primary, an attempt should be made ing the number of involved nodes and the total number of
to determine the primary site because of the differing nodes examined. The number of nodes needed to ensure
therapeutic options available for pancreatic versus ‘‘adequate’’ staging has not been defined, but attempts
intestinal NETs.41,61,70,96 In the case of well-differentiated should be made to identify as many lymph nodes in the
NETs, immunohistochemical staining can be helpful to resected specimen as possible. There was no agreement
determine the primary site; stains for CDX2 and TTF1 about the necessity of distinguishing micrometastases
should be carried out, which would point toward an from macrometastases (47% favored making a distinc-
origin in the intestines or pancreas (for CDX2) and lung tion), in part because no universally acceptable definition
(TTF1), respectively.32,78,84 The sensitivity and specificity of micrometastasis has been proposed for NETs. In
of these stains require more study, however, and negative addition, it was not agreed whether the size of the largest
staining does not exclude origin in these sites. It should be lymph node metastasis should be documented (56%
noted that staining for TTF1 is not helpful for high-grade believed the size should be reported). The group agreed
neuroendocrine carcinomas, such as small cell carcinoma, that TNM staging should be carried out using 1 of the
as extrapulmonary examples may express this mar- proposed staging systems,21,75,76 but it was also recog-
ker.13,51,81,92 Peptide immunohistochemistry was felt to nized that several systems exist, none of which is currently
have limited value in the determination of the primary site universally accepted. For this reason, it is critical to
for NETs presenting with metastatic disease and was not indicate in the report which specific TNM staging system
recommended. The group also recommended against the has been used.
use of silver-based histochemical stains such as argentaffin The status of the resection margins should be
or argyrophil reactions, as these stains have largely been reported, both for primary tumor resections and for
replaced by immunohistochemical markers such as resected metastases. For resected tumors with close
chromogranin and synaptophysin. margins (ie, within less than 0.5 cm), it was recommended
The possibility of carrying out immunohistochem- that the distance to the margin be indicated. It was not
ical staining for somatostatin receptors (SSTRs) was agreed to be necessary to document the distance if the
discussed,65,91 given the therapeutic implications of SSTR tumor is more than 0.5 cm from the margin, but 74% still
expression.55,59,72,89 However, it was agreed that these thought it should be reported.
stains are not currently routinely indicated, because well
characterized commercially available SSTR subtype Grading
antibodies do not exist, and the information about SSTR Another fundamental predictor of outcome in
expression to some extent can be inferred based on NETs is the grade of the tumor. The group recognized
somatostatin receptor scintigraphy (SSTR 2 and 5), which 3 grading categories of NETs: low grade, intermediate
is now widely carried out in patients with NETs.60,89 grade, and high grade.38,75,76,87 It was further agreed that
Further evaluation of the clinical relevance of SSTR in general, well-differentiated NETs are regarded to be
subtype evaluation of individual NETs may lead to either low grade or intermediate grade, whereas poorly
increased application of this assessment to determine the differentiated NETs are high grade by definition. The
specific utility of SST analogues with different receptor group recognized the dramatic difference in clinical
profiles.91 behavior between well-differentiated NETs (which can
progress very slowly over years to decades, even in the
Staging presence of metastatic disease) and poorly differentiated
The prognostic importance of tumor stage is widely NETs (which are high-grade carcinomas characterized by
reflected in all of the classification systems for NETs. The rapid dissemination, resistance to treatment, and a
discussions focused on the specific features of the primary quickly fatal course). Thus, the fundamental distinction
tumors that should be included in pathology reports that of well differentiated from poorly differentiated NETs

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 303


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

was recognized to be a critical pathologic determination. should be identified by scanning the slides at intermediate
Grade progression in well-differentiated NETs was magnification, and the formal mitotic count should
discussed, and it was agreed that this phenomenon can include the most active areas to ensure assessment of
occur.43,52,69,100 Thus, it is possible that different regions the worst case scenario for therapeutic strategy.
within a single tumor (or different sites of metastasis) can Significant discussion revolved around the use of
display different grades. However, there has been little immunohistochemical staining for Ki67 as an alternative
histologic documentation of grade progression and it is method to quantify the proliferative fraction.33 The
not clear how often NETs may undergo this change. Ki67 index (expressed as the percentage of neoplastic
There have been rare reports of high-grade transforma- cells showing nuclear labeling) is used in several grading
tion of well-differentiated NETs,86 but this occurrence and classification systems and is incorporated into the
seems to be particularly rare; there are insufficient data recent ENETS proposals.28,75,76 In Europe, Ki67 is widely
to determine the biologic behavior of such tumors. Thus, used and reported as a critical pathologic predictor of
there was no agreement about whether well-differentiated prognosis. In North America, however, the use of Ki67
NETs could ever be biologically high grade (68% felt staining has largely been restricted to selected institutions
that well-differentiated NETs can never be high grade). for specific clinical or pathologic indications, and routine
It was noted that in some circumstances, more than reporting of this marker is not currently the standard of
1 primary NET may arise within a single organ. In the practice. The group was divided regarding the advisability
case of familial multicentric NETs, it was agreed that of incorporating Ki67 labeling indices into reports (53%
the different tumors can also have different grades, said no), and there was no agreement that the stain
but there was no agreement on this point regarding should be carried out in all cases, especially for resection
sporadic multicentric NETs. There was a consensus that specimens, in which an accurate mitotic count can be
no NETs should be reported as ‘‘benign’’ (other than carried out readily. However, it was agreed that Ki67
tightly defined subsets such as pancreatic or pulmo- staining can be highly useful for biopsy specimens. The
nary NETs less than 0.5 cm or gastric NETs less than vastly different labeling rates between well-differentiated
0.5 mm).35,37,38,83 NETs (with a labeling index typically in the 1% to 20%
The group recognized that different grading systems range) and high-grade neuroendocrine carcinomas (such
exist for well-differentiated NETs of different anatomic as small cell carcinoma and large cell neuroendocrine
sites, and although there was consensus that a uniform carcinoma, which typically show 50% to 95% labeling)
grading system applicable to all primary sites would make Ki67 staining very effective for distinguishing these
be desirable, the data to justify such a system do not groups.49,67 In addition, mitotic counting of small speci-
currently exist. The difficulty of determining which mens is difficult, as many of these samples will not contain
grading scheme to apply to metastases of unknown sufficient neoplastic tissue to enable 50 high-power fields
primary sites was acknowledged. Despite all of the to be counted. There was consensus; therefore, that Ki67
vagaries of grading NETs, there was unanimous agree- staining should be reported for biopsies of metastases or
ment that a grade should be stated in the report, with in other circumstances in which a larger resected portion
the particular grading system used to be specified. of the tumor is unlikely to become available.
Furthermore, the raw information used to derive the Another important consideration was that the
grade should also be reported to enable comparison optimal method to determine the Ki67 index has yet to
between different grading systems. It was recognized that be defined. Only 53% of participants believed that the
the proliferative index is inherently linked to the grade of Ki67 index was sufficiently reproducible between pathol-
NETs but it was also appreciated that the proliferative ogists to discriminate clinically relevant subsets of NETs.
fraction may not be homogeneous throughout a given Options to quantify Ki67 labeling include a general
NET and that there may be differences in proliferation ‘‘eyeballed’’ estimate of the percentage of positive cells,
between the primary tumor and metastatic sites. The systematically counting manually a defined number of
simplest method to determine the proliferative index is tumors cells (2000 as per the ENETS proposals75,76) and
to count mitotic figures, and this piece of data was calculating the positive percentage, or using a computer-
unanimously accepted to be necessary for complete ized digital image analysis system to measure the positive
pathology reports. The group agreed that the calculation percentage.18,48 Manual counting was felt by some to be
of the number of mitoses per unit area of the tumor was too time-consuming for general pathology practice, and it
the most reliable, and that the number of mitoses in 10 was agreed that image analysis, although attractive and in
high-power fields (2 mm2) was a standard format used in use at some institutions, was not yet sufficiently available
many classification and grading systems and should be for routine practice. There was no agreement that these 2
endorsed. It was pointed out that some grading schemes techniques could be advocated at this time. Seventy-five
use the number of mitoses in 50 high-power fields,22,30 percent felt that manual counting should not be done, and
and the group agreed that a total of 50 fields should be 63% believed that image analysis is not yet ready for
counted, but ultimately the mitotic rate should be general use. Thus, from a practical standpoint, providing
expressed based on the number in 10 high-power fields. an ‘‘eyeballed’’ estimate of the labeling percentage was
It is not adequate to count a single randomly chosen area agreed to be the only method that could be strongly
of the tumor; rather, the most active regions (‘‘hot spots’’) advocated at present. However, there was recognition of

304 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

many shortcomings to this approach. The interobserver The group acknowledged that the size of the pri-
(and likely intraobserver) variability was felt to be high, mary tumor may be associated with the mitotic rate and,
especially when a difference between 1% and 3% is used therefore, the grade, but it was agreed that the tumor size
to separate 2 grades in some systems.75,76 Furthermore, it was a parameter that helps define the stage of the tumor
was recognized that there is intratumoral heterogeneity rather than the grade. Thus, grade and stage should be
in Ki67 labeling (Fig. 1), meaning that a small sample separately determined and reported; the size of the NET
of a NET (eg, a biopsy of a metastasis) may not represent should not be used in the determination of the grade.
the most highly proliferative region of the tumor.15 The
group recommended to count the most densely staining
regions (‘‘hot spots’’) and to count a variety of areas Metastasis-specific Issues
within the tumor; it was specifically noted that counting Several issues specifically relate to the reporting
of random areas or single regions is inadequate. The of metastatic disease. For resected metastases, it was
result should be reported as a single percentage reflecting recommended to report the anatomic site, the number of
the average of the regions counted, rather than a range of lesions resected, and the size (in 1 dimension) of the
values. Although many issues were recognized regarding largest metastasis. Some comment about the percentage
the use of Ki67 staining, it was agreed that there currently of involvement of the resected tissue (such as the liver)
exists no better marker of proliferation for routine use. should be provided. Owing to the potential for hetero-
If multiple sites of disease are sampled, it was agreed geneity among different sites of metastasis, the group
that separate mitotic counts should be provided for each advised sampling more than 1 metastasis for histology,
anatomic site (for example the primary lesion, lymph if multiple metastases are resected. It was recognized
node metastasis, liver metastasis). There was no agree- that currently there is no system for substaging distant
ment whether separate Ki67 indices should be reported metastatic disease, meaning that patients with limited
for different sites of disease (61% said yes). spread outside the regional lymph nodes are grouped
Some grading systems for NETs use the presence of together with those having extensive, widely distributed
tumor necrosis (ie, nonischemic or infarct-like necrosis) as metastases. The desirability of a substaging system for
a criterion to increase the tumor grade.30,87 Thus, it was metastatic disease was acknowledged, as it would better
agreed that the presence of nonischemic tumor necrosis stratify the extent of disease for comparison of therapeutic
should be documented in the pathology report. trials.

FIGURE 1. Immunohistochemical staining for Ki67. Two images captured from a single pancreatic neuroendocrine tumor (NET)
reveal heterogeneity of labeling, from 1.4% in 1 region (A) to 21.5% in another (B).

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 305


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

Grading of metastases creates some special chal- DISCUSSION


lenges, as has been partially discussed above. Although Many different groups have explored the topic of
in most cases well-differentiated NETs can be distin- NET classification. Various systems of nomenclature,
guished from poorly differentiated (high grade) neuro- grading, and staging have been published, but none has
endocrine carcinomas, the distinction of low grade gained universal acceptance. Discussion of the systems
and intermediated-grade NETs can be very challenging proposed by the WHO, the ENETS, the AJCC, and other
based on biopsy material. Although the group agreed groups9,10,21,22,30,35,38,45,75,76 revealed that essentially all of
that an attempt should be made to grade NETs on these classifications are useful to stratify survival after
biopsy specimens, it was only felt to be realistic when resection of NETs and to predict the rate of progression
adequate neoplastic tissue is represented in the biopsy, of metastatic disease. A graphic comparison of major
and distinction of low and intermediate-grade NETs is grading, staging, and classification systems is presented in
not possible based on fine needle aspiration cytology Figure 2. This study was carried out in an effort to define
specimens. the essential pathologic information needed for clinical
decision-making that in fact constitutes the data under-
lying most of these NET classifications. There was no
Other Prognostic Factors and intent to introduce another classification system, as it was
Therapeutic Biomarkers perceived that adding to the plethora of available systems
already in use would only cause greater confusion.
Many different potential prognostic factors for
The application of the Delphic process19,25,77 in this
NETs have been reported. Histologic findings that
study seems to be novel for ‘‘consensus’’ groups preparing
were of recognized significance (and therefore, should
joint statements related to NETs. Many group efforts
be reported) include vascular and perineural invasion.
involve open and extensive discussions, allowing all
Immunohistochemical stains for endothelial markers
participants to present and defend their opinions. How-
(such as CD34 or D2-40) that can aid in identifying
ever, the decision-making process is often not anonymous
vascular invasion were recommended only for cases with
and introduces issues such as peer pressure, weight of
a histologic suspicion of vascular invasion. It was not felt
personal opinion, and vigor of personality into the
necessary to differentiate between lymphatic and blood
deliberations. The use of anonymous voting with an
vessel invasion, as this distinction often cannot be made
electronic system in this study ensured that all partici-
reliably on the basis of routine histologic sections. The
pants had an equal opportunity to express an independent
various architectural growth patterns of NETs were not
opinion. It is possible that this methodology prevented
considered to be prognostically relevant, nor was nuclear
agreement from being reached on some issues, as the
atypia. Unusual histologic features in NETs (including
holders of minority opinions could less readily be
clear cell or oncocytic morphology, and gland-formation)
swayed to change their votes. Nonetheless, agreement
should be mentioned in the report, however, as such
was achieved for 84% of the questions.
features may obscure the neuroendocrine nature of the Major areas of agreement were achieved related to
tumor. It was not thought necessary to quantify angio- terminology, use of diagnostic immunohistochemistry,
genesis or apoptosis. Although the immunoexpression of grading parameters, staging information, relevance of
cytokeratin 19 (CK19) reportedly has adverse prognostic other histologic prognostic features, and utility of ther-
implications in pancreatic NETs,17 the data were not felt apeutic biomarkers. The group agreed upon the inter-
to be sufficiently compelling to recommend the routine changeable use of ‘‘endocrine’’ and ‘‘neuroendocrine’’
use of this marker. terms to refer to these tumors and the desirability of
Advances in targeted therapy have risen the possi- eventually eliminating the term ‘‘carcinoid.’’12,82 The
bility that molecular testing or immunohistochemistry specific immunohistochemical stains useful to define
may become useful to determine the treatment of NETs. neuroendocrine differentiation were agreed upon, as were
However, at present no special immunohistochemical the limited circumstances under which staining for
stains or molecular tests are routinely recommended. bioactive peptide or amine secretory products (hormones)
may be helpful. As some novel therapeutic agents have
shown greater efficacy against NETs from specific
Other Information primary sites (eg, temozolamide for pancreatic NETs42),
It was agreed that abnormalities in the neuroendo- the need to attempt to determine the origin of meta-
crine cells in the resected organs away from the NET stases from occult primary NETs was recognized, and
should be described in the report. These include, for immunohistochemical staining for CDX2 and TTF1 was
example, gastric neuroendocrine cell hyperplasia asso- recommended as a diagnostic aid.32,78,84 Recent data also
ciated with Type 1 or Type 2 gastric well-differentiated suggest that staining for Isl1 may also be useful to define
NETs, or pancreatic endocrine microadenomas asso- pancreatic origin.79 The group also recognized many
ciated with well-differentiated pancreatic endocrine tu- potential immunohistochemical biomarkers of therapeu-
mors in patients with MEN1.2,3,37,68,83 Many NETs may tic responsiveness on the horizon, including somatostatin
exhibit stromal fibrosis, but it was agreed that there was receptor subtypes, MGMT, VEGF-r, and mTOR path-
no indication to attempt to quantify stromal fibrosis. way members26,34,42,53,73,94,97; however, none of these has

306 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

G r a d e

WD NET WD NET
Low Grade Intermediate Grade PD NE Carcinoma
Others

WD NE Carcinoma MD NE Carcinoma PD NE Carcinoma

HG NE Carcinoma
WHO Lung Typical Carcinoid Atypical Carcinoid Small Cell/Large Cell

ENETS G1 G2 G3
T1 T2 T3
Localized

Well Differentiated
(Neuro)endocrine Tumor
S t a g e

Poorly Differentiated
T4 N1

WHO WHO (Neuro)endocrine


Carcinoma
Regional Distant

M1

Well Differentiated
(Neuro)endocrine Carcinoma
SEER

AJCC

FIGURE 2. Comparison of various grading, staging, and classification systems for neuroendocrine tumors (NETs). The grading
systems are displayed along the x-axis (top) and the staging systems along the y-axis (left). The WHO systems for gastrointestinal
and pancreatic NETs include a combination of grading and staging information and are displayed with the box (lower right). The
various nomenclature of each system is used. The overlaps between the different systems are approximate. Abbreviations: AJCC
indicates American Joint Committee on Cancer; ENETS, European Neuroendocrine Tumors Society; HG, high grade; MD,
moderately differentiated; NE, neuroendocrine; NET, neuroendocrine tumor; PD, poorly differentiated; SEER, Surveillance,
Epidemiology, and End Results Program of the National Cancer Institute; WD, well differentiated; WHO, World Health
Organization.

been adequately validated to justify their routine use, and extent of disease as ‘‘localized,’’ ‘‘regional,’’ or ‘‘dis-
data showing the correlation between immunohistochem- tant.’’95 However, there is widespread recognition that
ical staining and therapeutic response are still emerging. NETs should be staged using the TNM system, and there
However, it seems likely that in the future, with increasing is currently an ENETS proposal for staging gastro-
advances toward specific molecular characterization of enteropancreatic NETs.75,76 The upcoming revision of the
NETs in terms of transcript identification (KiSS, MAGE, AJCC staging system will also include NETs.21 Although
NAPI) and functional biologic components (somatostatin there may be some subtle differences between the ENETS
receptor subtypes), that such information may become and AJCC systems, the fundamental staging landmarks
relevant.20 parallel those used for carcinomas of the corresponding
Formal TNM staging of NETs has been relatively organs, and the group recommended to include the
limited compared with the staging of carcinomas. For size of the tumor, the extent of invasion based on these
example, in the most recent AJCC staging classification, landmarks, the status of regional lymph nodes, and
gastroenteropancreatic NETs are specifically excluded the presence of distant metastatic disease (if known) in
from the TNM system.27 A relatively crude staging system the pathology reports, in addition to indicating a specific
exists in the Surveillance, Epidemiology, and End Results TNM stage. Reporting of margin status was also
Program database of the NCI, which categorizes the recommended.

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 307


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

There was universal recognition of the importance markers. Some of these more substantive disagreements
of grading NETs. The distinction of relatively slowly (particularly those related to terminology) reflect theore-
progressive well-differentiated NETs from highly aggres- tic concerns bolstered by ‘‘personal opinions’’ and long-
sive poorly differentiated NETs (small cell carcinoma and held traditions of practice. Whether to use ‘‘tumor’’ or
large cell neuroendocrine carcinoma) was highlighted ‘‘neoplasm’’ and whether the term ‘‘carcinoma’’ should be
as a critical pathologic determination.6,14,23,62 The rele- applied to well-differentiated NETs are important con-
vance of tumor cell proliferation for grading NETs was siderations that could not be resolved, but the basis for
also agreed. Mitotic rate was regarded as an essential the disagreement is unrelated to a lack of understanding
component of pathologic data, and specific recommen- of the disease or the absence of sufficient data. We
dations for the technique of mitotic counting were consider these to reflect the evolution of semantic
established. The existence of 3 grades of NETs was also terminology and not a real disagreement in terms of the
agreed upon, and it was accepted that inclusion of the histopathology or biology of NETs. It was clear to all
grade of the tumor, along with a reference to the specific that much of the terminology (tumor or carcinoid) reflects
grading system being used, should be included in all archaic medical lexicon and will with the passage of time
reports on NETs. Although there was complete agree- fade into medical history rather than be banned from
ment that grade can progress as NETs grow and usage.12,16,82 The routine use of immunohistochemical
disseminate,43,52,69,100 this was not considered a uniform staining for general neuroendocrine markers was also
event in the evolution of the disease. An important issue controversial, the divergent opinions reflecting varied
that was widely accepted was that intratumoral hetero- levels of comfort with diagnoses rendered solely on the
geneity of proliferative rate requires careful consideration basis of routine histology.
given its potential impact on overall assessment of the Some areas of disagreement were based on a lack of
NET aggressiveness. Thus, separate grading and mitotic sufficient data to support specific concepts or procedures.
rate counting of metastases should be carried out, and The concept of neoplastic progression in NETs was
each major site of disease should be assessed indepen- accepted, but the group could not agree whether well-
dently. The inclusion of information about nonischemic differentiated NETs can progress to fully high-grade
necrosis is used in some grading systems and therefore, neuroendocrine carcinomas.86 Some preliminary data
should be mentioned in the reports.30,87 The use of documenting this occurrence were presented, but there
immunohistochemical staining for Ki67 as a measure of have been few published examples, and it seems that, at
proliferative rate was endorsed by the group for specific best, this transformation is probably rare and does not
situations (see below for further discussion about Ki67 represent the pathway of development of a vast majority
staining). The stain is especially useful for the distinction of high-grade neuroendocrine carcinomas.
of high-grade neuroendocrine carcinoma from well- Another controversial area was the use of Ki67
differentiated NETs and should always be used when staining to determine the proliferative rate.33 Although
this differential diagnosis arises (particularly on biopsy this stain is widely used, particularly to distinguish well-
specimens).49,67 In addition, in cases with minimal tumor differentiated NETs from poorly-differentiated NETs
for which adequate mitotic counting is not possible, Ki67 such as small cell carcinoma,49,67 there was a division of
staining should be used to determine the proliferative opinion about when to use it and whether it should be
rate, as it labels a greater proportion of cells than those carried out for all specimens of NETs, especially those
with active mitotic figures. with sufficient material to allow mitotic rate counting.
Other prognostic factors that were agreed to have The ENETS grading system includes Ki67 labeling index
sufficient relevance for inclusion in reports included as 1 of the parameters, and oncologists in Europe rely
the presence of vascular and perineural invasion. Nuclear extensively on this information for treatment.33,71,75,76,90
grading was not felt to be relevant, based on the ob- In the US, however, the use of Ki67 has been more
servation that nuclear pleomorphism can be seen in limited, reflecting concerns with rigor of the assessment
some well-differentiated NETs that lack aggressive of the parameter. The interpretation of Ki67 staining was
behavior.98 Quantification of the extent of fibrosis, also debated. Although it was agreed that the most
angiogenesis, and apoptosis was not considered to be accurate assessment would involve counting nuclei to
necessary. Unusual histologic features, although unlikely determine the true labeling percentage, this was consid-
to have prognostic significance, were believed to be ered by some to be too time-consuming and cumbersome
worthy of documentation in the reports. for routine practice. Digital image analysis holds promise
The 17 questions for which no agreement could be for quantification of staining, but many pathologists do
reached (Supplementary Table 3, Supplemental Digital not currently have access to this technology. An ‘‘eye-
Content 3, http://links.lww.com/PAS/A51) include some balled’’ estimate was acknowledged to be inaccurate
relatively minor issues related to the precision with which owing to have high interobserver variability, but this
the extent of disease should be reported but also more technique was agreed to be the only one that could
substantive issues regarding terminology, the need for practically be recommended.18 Nevertheless, given the
routine immunohistochemical staining, the concept of issues with reproducibility of this technique and the
progression of well-differentiated NETs to high-grade inherent heterogeneity of staining within NETs (especially
neuroendocrine carcinoma, and the use of proliferation metastases, from which only limited biopsy samples are

308 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

often available15), it was not agreed that the Ki67 labeling


TABLE 1. Minimum Pathology Data Set: Information to be
index is sufficiently reproducible to allow separation of Included in Pathology Reports on NETs
clinically relevant subsets of NETs. It is clear that much
For resection of primary tumors:
of the debate about the use of Ki67 staining was related to
a lack of data regarding intraobserver and interobserver Anatomic site of tumor
Diagnosis (functional status need not be included in pathology
reproducibility, correlation of automated quantification report)
with clinical outcome, assessment of intratumoral hetero- Size (3 dimensions)
geneity, and comparison with mitotic rate as the ultimate Presence of unusual histologic features (oncocytic, clear cell, gland-
determinant of outcome and indication for specific pharma- forming, etc.)
Presence of multicentric disease
cologic therapy. The resolution of these critical issues was (OPTIONAL: immunohistochemical staining for general
agreed upon as a priority in the future investigation and neuroendocrine markers)
assessment of NETs. Chromogranin
The areas of consensus reached by this group were Synaptophysin
sufficient to allow the development of a Minimum Patho- Peptide hormones, IF a specific clinical situation suggests that the
correlation with a functional syndrome may be helpful
logy Data Set for the reporting of NETs (Table 1). This Grade (specify grading system used)
Minimum Pathology Data Set includes recommenda- Mitotic rate (number of mitoses per 10 high-power fields or 2 mm2;
tions for reporting both biopsy and resection specimens count 50 high-power fields in the most mitotically active regions, count
from primary tumors and metastases, and it applies to all multiple regions)
[OPTIONAL: Ki67 labeling index (count multiple regions with
NETs of the gastrointestinal tract and pancreas. In highest labeling density, report average percentage; ‘‘eyeballed’’
general, the group recommended reporting only those estimate is adequate)]
histologic features for which abundant data exist to Presence of nonischemic tumor necrosis
document their prognostic significance. In each of these Presence of other pathologic components (eg, nonneuroendocrine
sites, a specific diagnosis and grade are to be reported, components)
Extent of invasion (use anatomic landmarks for the AJCC T-staging
and staging is to be carried out when the information is of analogous carcinomas of the same anatomic sites)
available (ie, for resections and biopsies of metastases). Stomach: depth of invasion into/through gastric wall
However, there was no mandate to use any specific system Small bowel: depth of invasion into/through bowel wall
of nomenclature, grading, or staging, because the infor- Large bowel: depth of invasion into/through bowel wall
Appendix: depth of invasion into/through appendiceal wall;
mation that is used for these purposes is to be included presence and extent of mesoappendiceal invasion
elsewhere in the report. For example, the mitotic rate, Pancreas: the presence of extrapancreatic invasion or invasion of
with or without information about necrosis, is used to bile duct, duodenum, or ampulla
grade NETs in all major systems; and this information All sites: involvement of serosal/peritoneal surfaces; invasion of
should be routinely reported. The extent of invasion is to adjacent organs or structures
Presence of vascular invasion (OPTIONAL: perform
be reported, using similar landmarks to those used in the immunohistochemical stains for endothelial markers if needed)
AJCC staging systems for carcinomas of corresponding Presence of perineural invasion
organs. In fact, this should translate easily into the Lymph node metastases
upcoming AJCC staging system for NETs of the GI tract Number of positive nodes
Total number of nodes examined
and pancreas, which in almost every instance is based TNM Staging (specify staging system utilized)
on the corresponding carcinoma staging system. It is Resection margins (positive/negative/close) (OPTIONAL measure
important to indicate in the report which systems of distance from margin if within 0.5 cm)
nomenclature, grading, and staging are being used to Proliferative changes or other abnormalities in nonneoplastic
allow ready comparison among cases. neuroendocrine cells
A number of items were regarded as ‘‘optional’’ for For Biopsy of Primary Tumors:
inclusion in reports, based upon lack of agreement among Anatomic site of tumor
the participants about their importance. For example, Diagnosis (functional status need not be included in pathology report)
routine staining for general neuroendocrine markers Presence of unusual histologic features (oncocytic, clear cell, gland-
(chromogranin and synaptophysin) of resected NETs forming, etc.)
with classic histologic features was not agreed to be (OPTIONAL: immunohistochemical staining for general
neuroendocrine markers)
necessary, but can be carried out and reported if the Chromogranin
pathologist finds it necessary or the clinical situation Synaptophysin
requires it. Routine Ki67 staining was also deemed Peptide hormones, IF a specific clinical situation suggests that the
optional, unless the specimen in question is a biopsy correlation with a functional syndrome may be helpful
Grade (specify grading system used)
of a primary NET in which a high-grade neuroendocrine Mitotic rate (number of mitoses per 10 high-power fields or 2 mm2;
carcinoma could not be excluded or a biopsy of a meta- count up to 50 high-power fields)
stasis with inadequate tissue for accurate mitotic count- Ki67 labeling index, for biopsies in which a diagnosis of high-grade
ing. Measurement of the distance from the tumor to the neuroendocrine carcinoma cannot be excluded (count multiple regions
resection margins for cases grossly within less than 0.5 cm with highest labeling density, report average percentage; ‘‘eyeballed’’
estimate is adequate)
was also considered optional. It was clear from the Presence of nonischemic tumor necrosis
discussions, however, that some of this optional informa- Presence of other pathologic components (eg, nonneuroendocrine
tion might be regarded as mandatory in certain practice components)

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 309


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

significant factors, rather than data-driven subgroup-


TABLE 1. (continued)
ings based on extensive study of actual cases. The group
For Resection of Metastatic Tumors: recognized the need for improved clinicopathologic
Location of metastasis(es) studies of NETs based on large numbers of cases with
Diagnosis (functional status need not be included in pathology report) long-term follow-up, which almost certainly will need to
Number of metastases resected involve multiple institutions. The existing data actually
Extent of involvement of resected tissue (percentage)
Greastest dimension of largest metastasis
suggest that different grading parameters may optimally
Presence of unusual histologic features (oncocytic, clear cell, gland- separate prognostic groups from different anatomic
forming, etc.) sites.39,62 For example, some studies have shown that a
(OPTIONAL: immunohistochemical staining for general mitotic rate of 2/10 high-power fields optimally separates
neuroendocrine markers) low-grade NETs from intermediate-grade NETs in the
Chromogranin
Synaptophysin lung,88 whereas other investigators suggest that a much
Peptide hormones, IF a specific clinical situation suggests the lower rate of 2/50 high-power fields should be used
correlation with a functional syndrome may be useful for NETs in the pancreas and rectum.22,30 Many studies
Grade (specify grading system used) have used a mitotic rate of 10/10 high-power fields
Mitotic rate (number of mitoses per 10 high-power fields or 2 mm2;
count 50 high-power fields in the most mitotically active regions,
to distinguish well-differentiated (low and intermediate
provide separate mitotic rate for each major separate site of disease) grade) NETs from poorly-differentiated (high grade)
[OPTIONAL: Ki67 labeling index (count multiple regions with neuroendocrine carcinomas in sites such as the lung,
highest labeling density, report average percentage; ‘‘eyeballed’’ thymus, ampulla of Vater, and pancreas28,57,85,88; how-
estimate is adequate)] ever, according to the ENETS grading scheme, a rate
Presence of nonischemic tumor necrosis
Presence of other pathologic components of 20/10 high-power fields is preferable for this distinc-
Resection margins (positive/negative/close) (OPTIONAL measure tion.75,76 Unification of grading criteria among anatomic
distance from margin if within 0.5 cm) sites would have the advantage of allowing grading of
Identification of primary site metastatic NETs for which the primary site may not be
Immunohistochemistry for CDX2, TTF1
known. Until adequate data are obtained to determine
For Biopsy of Metastatic Tumors: whether a unified grading system can be developed, how-
ever, it is critical to continue to collect the specific data
Location of metastasis
Diagnosis (functional status need not be included in pathology report)
that underlie the grading schema being used. Namely, the
Presence of unusual histologic features (oncocytic, clear cell, gland- proliferative rate (mitotic rate, and/or Ki67 labeling
forming, etc.) index) should be specifically recorded for all NETs. This
Immunohistochemical staining for general neuroendocrine markers will allow translation between different systems and will
Chromogranin facilitate comparison of data from studies that may use
Synaptophysin
(OPTIONAL: peptide hormones, IF a specific clinical situation different cut-points for grading.
suggests the correlation with a functional syndrome may be useful) It must also be recognized that clinical or radio-
Grade for adequate biopsy specimens; FNA specimens may not be graphic information is useful in predicting the biology
adequate (specify grading system used) of NETs. For patients with metastases, a decision to
Mitotic rate (number of mitoses per 10 high-power fields or 2 mm2;
count up to 50 high-power fields)
treat can also be based on presence of symptoms,
Ki67 labeling index (count multiple regions with highest labeling whether hormone-mediated or based on the site and
density, report average percentage; ‘‘eyeballed’’ estimate is adequate) burden of disease. Longitudinal radiographic studies can
Presence of nonischemic tumor necrosis be used to determine the growth rate of metastases, which
Presence of other pathologic components (eg, nonneuroendocrine may vary among different sites of metastatic disease.73
components)
Identification of primary site This clinical and radiographic information must be
Immunohistochemistry for CDX2, TTF1 integrated with the pathology data for optimal thera-
peutic decision-making.
The development of a standardized approach to
the analysis of the pathology of neuroendocrine tumor
situations, and close collaboration with treating clinicians disease is a necessary step to provide a secure platform
is suggested to ensure the necessary data are included in upon which the evolving therapeutic landscape can be
the reports. based, and it is vital to establish clear parameters within
Although there was some disagreement about the which the disease can be uniformly evaluated and rational
importance of various pathologic findings, and no therapeutic strategies developed. A clear advance was
universal grading or staging system arose through this the development of a Minimum Pathology Data Set as
consensus meeting, there was general agreement that it proposed by this group. The routine application and use
would be preferable to develop such a system. Ideally, the of such information will result in the accumulation of
grading parameters for well-differentiated NETs would consistent and globally applicable information about the
be the same irrespective of the site of origin, and the pathology of NETs. With time, this will translate into an
terminology would also be uniform. One of the reasons improved capability to define the disease of individual
for the variety of different systems is that most are patients and facilitate provision of a uniform basis to
proposals based on a reasonable assessment of potentially prognosticate and treat patients. Widespread adoption of

310 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

this strategy will assist the more fluid translation of novel metastatic colorectal cancer using a Delphi process. J Surg Res.
therapeutic strategies from the research arena into routine 2009;156:32–38.
practice. 20. Drozdov I, Kidd M, Nadler B, et al. Predicting neuroendocrine
tumor (carcinoid) neoplasia using gene expression profiling and
supervised machine learning. Cancer. 2009;115:1638–1650.
21. Edge SE, Byrd DR, Carducci MA, et al. AJCC Cancer Staging
REFERENCES Manual. 7th ed. New York: Springer; 2009. (in press).
1. Ahmed A, Turner G, King B, et al. Midgut neuroendocrine 22. Fahy BN, Tang LH, Klimstra D, et al. Carcinoid of the rectum risk
tumours with liver metastases. Results of the UKI NETS study. stratification (CaRRS): a strategy for preoperative outcome
Endocr Relat Cancer. 2009;16:885–894. assessment. Ann Surg Oncol. 2007;14:396–404.
2. Anlauf M, Perren A, Kloppel G. Endocrine precursor lesions and 23. Fenoglio-Preiser CM. Gastrointestinal neuroendocrine/neuroecto-
microadenomas of the duodenum and pancreas with and without dermal tumors. Am J Clin Pathol. 2001;115(suppl):S79–S93.
MEN1: criteria, molecular concepts and clinical significance. 24. Ferrone CR, Tang LH, Tomlinson J, et al. Determining prognosis
Pathobiology. 2007;74:279–284. in patients with pancreatic endocrine neoplasms: can the
3. Anlauf M, Schlenger R, Perren A, et al. Microadenomatosis of the WHO classification system be simplified? J Clin Oncol. 2007;25:
endocrine pancreas in patients with and without the multiple 5609–5615.
endocrine neoplasia type 1 syndrome. Am J Surg Pathol. 2006;30: 25. Fink A, Kosecoff J, Chassin M, et al. Consensus methods:
560–574. characteristics and guidelines for use. Am J Public Health. 1984;
4. Arnold R. Endocrine tumours of the gastrointestinal tract. 74:979–983.
Introduction: definition, historical aspects, classification, staging, 26. Granberg D. Investigational drugs for neuroendocrine tumours.
prognosis and therapeutic options. Best Pract Res Clin Gastro- Expert Opin Investig Drugs. 2009;18:601–608.
enterol. 2005;19:491–505. 27. Green FL, Page DL, Fleming ID, et al. AJCC Cancer Staging
5. Beasley MB, Thunnissen FB, Brambilla E, et al. Pulmonary Manual. 6th ed. New York: Springer; 2002.
atypical carcinoid: predictors of survival in 106 cases. Hum Pathol. 28. Heitz PU, Komminoth P, Perren A, et al. Pancreatic endocrine
2000;31:1255–1265. tumours: introduction. In: DeLellis RA, Lloyd RV, Heitz PU, et al,
6. Bordi C, D’Adda T, Azzoni C, et al. Criteria for malignancy in eds. Pathology and Genetics of Tumours of Endocrine Organs. Lyon:
gastrointestinal endocrine tumors. Endocr Pathol. 2006;17:119–129. IARC Press; 2004:177–182.
7. Burke AP, Thomas RM, Elsayed AM, et al. Carcinoids of the
29. Heymann MF, Joubert M, Nemeth J, et al. Prognostic and
jejunum and ileum: an immunohistochemical and clinicopathologic
immunohistochemical validation of the capella classification of
study of 167 cases. Cancer. 1997;79:1086–1093.
pancreatic neuroendocrine tumours: an analysis of 82 sporadic
8. Capella C, Solcia E, Sobin LH, et al. Endocrine tumours of the
cases. Histopathology. 2000;36:421–432.
colon and rectum. In: Hamilton SR, Aaltonen LA, eds. Pathology
30. Hochwald SN, Zee S, Conlon KC, et al. Prognostic factors in
and Genetics of Tumours of the Digestive System. Lyon: IARC
pancreatic endocrine neoplasms: an analysis of 136 cases with
Press; 2000:137–139.
a proposal for low-grade and intermediate-grade groups. J Clin
9. Capella C, Solcia E, Sobin LH, et al. Endocrine tumours of the
Oncol. 2002;20:2633–2642.
small intestine. In: Hamilton SR, Aaltonen LA, eds. Pathology and
Genetics of Tumours of the Digestive System. Lyon: IARC Press; 31. Hofer MD, Chang MC, Hirko KA, et al. Immunohistochemical
2000:77–82. and clinicopathological correlation of the metastasis-associated
10. Capella C, Solcia E, Sobin LH, et al. Endocrine tumours of the gene 1 (MTA1) expression in benign and malignant pancreatic
stomach. In: Hamilton SR, Aaltonen LA, eds. Pathology and endocrine tumors. Mod Pathol. 2009;22:933–939.
Genetics of Tumours of the Digestive System. Lyon: IARC Press; 32. Jaffee IM, Rahmani M, Singhal MG, et al. Expression of the
2000:53–57. intestinal transcription factor CDX2 in carcinoid tumors is a marker
11. Chatzipantelis P, Konstantinou P, Kaklamanos M, et al. The role of midgut origin. Arch Pathol Lab Med. 2006;130:1522–1526.
of cytomorphology and proliferative activity in predicting biologic 33. Jamali M, Chetty R. Predicting prognosis in gastroentero-
behavior of pancreatic neuroendocrine tumors: a study by pancreatic neuroendocrine tumors: an overview and the value of
endoscopic ultrasound-guided fine-needle aspiration cytology. ki-67 immunostaining. Endocr Pathol. 2008;19:282–288.
Cancer Cytopathol. 2009;117:211–216. 34. Janson ET. Somatostatin analogs in the treatment of neuro-
12. Chetty R. Requiem for the term ‘‘carcinoid tumour’’ in the endocrine gastroenteropancreatic and intrathoracic tumors.
gastrointestinal tract? Can J Gastroenterol. 2008;22:357–358. J Endocrinol Invest. 2005;28:137–140.
13. Cheuk W, Kwan MY, Suster S, et al. Immunostaining for thyroid 35. Klimstra D, Perren A, Oberg K, et al. Pancreatic endocrine
transcription factor 1 and cytokeratin 20 aids the distinction of tumours: non-functioning tumours and microadenomas. In:
small cell carcinoma from Merkel cell carcinoma, but not DeLellis RA, Lloyd RV, Heitz PU, et al, eds. Pathology and
pulmonary from extrapulmonary small cell carcinomas. Arch Genetics of Tumours of Endocrine Organs. Lyon: IARC Press;
Pathol Lab Med. 2001;125:228–231. 2004:201–204.
14. Cho CS, Labow DM, Tang L, et al. Histologic grade is correlated 36. Kloppel G, Anlauf M. Epidemiology, tumour biology and
with outcome after resection of hepatic neuroendocrine neoplasms. histopathological classification of neuroendocrine tumours of the
Cancer. 2008;113:126–134. gastrointestinal tract. Best Pract Res Clin Gastroenterol. 2005;19:
15. Couvelard A, Deschamps L, Ravaud P, et al. Heterogeneity of 507–517.
tumor prognostic markers: a reproducibility study applied to liver 37. Kloppel G, Anlauf M, Perren A. Endocrine precursor lesions
metastases of pancreatic endocrine tumors. Mod Pathol. 2009;22: of gastroenteropancreatic neuroendocrine tumors. Endocr Pathol.
273–281. 2007;18:150–155.
16. DeLellis RA. The neuroendocrine system and its tumors: an 38. Kloppel G, Perren A, Heitz PU. The gastroenteropancreatic
overview. Am J Clin Pathol. 2001;115(suppl):S5–S16. neuroendocrine cell system and its tumors: the WHO classification.
17. Deshpande V, Fernandez-del Castillo C, Muzikansky A, et al. Ann N Y Acad Sci. 2004;1014:13–27.
Cytokeratin 19 is a powerful predictor of survival in pancreatic 39. Kloppel G, Rindi G, Anlauf M, et al. Site-specific biology and
endocrine tumors. Am J Surg Pathol. 2004;28:1145–1153. pathology of gastroenteropancreatic neuroendocrine tumors.
18. Dhall D, Frishberg DP, Galliano G, et al. Interobserver varia- Virchows Arch. 2007;451(suppl 1):S9–S27.
bility in assessing Ki67 proliferative index in gastrointestinal 40. Koura AN, Giacco GG, Curley SA, et al. Carcinoid tumors of the
well-differentiated neuroendocrine neoplasms. Mod Pathol. 2009; rectum: effect of size, histopathology, and surgical treatment on
22:116A. metastasis free survival. Cancer. 1997;79:1294–1298.
19. Dixon E, Armstrong C, Maddern G, et al. Development of quality 41. Kulke MH. New developments in the treatment of gastrointestinal
indicators of care for patients undergoing hepatic resection for neuroendocrine tumors. Curr Oncol Rep. 2007;9:177–183.

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 311


Klimstra et al Am J Surg Pathol  Volume 34, Number 3, March 2010

42. Kulke MH, Hornick JL, Frauenhoffer C, et al. O6-methylguanine 65. Papotti M, Bongiovanni M, Volante M, et al. Expression of
DNA methyltransferase deficiency and response to temozolomide- somatostatin receptor types 1-5 in 81 cases of gastrointestinal and
based therapy in patients with neuroendocrine tumors. Clin Cancer pancreatic endocrine tumors. A correlative immunohistochemical
Res. 2009;15:338–345. and reverse-transcriptase polymerase chain reaction analysis.
43. Kytola S, Hoog A, Nord B, et al. Comparative genomic Virchows Arch. 2002;440:461–475.
hybridization identifies loss of 18q22-qter as an early and specific 66. Pelosi G, Bresaola E, Bogina G, et al. Endocrine tumors of the
event in tumorigenesis of midgut carcinoids. Am J Pathol. 2001; pancreas: Ki-67 immunoreactivity on paraffin sections is an
158:1803–1808. independent predictor for malignancy: a comparative study with
44. La Rosa S, Sessa F, Capella C, et al. Prognostic criteria in proliferating-cell nuclear antigen and progesterone receptor protein
nonfunctioning pancreatic endocrine tumours. Virchows Arch. immunostaining, mitotic index, and other clinicopathologic vari-
1996;429:323–333. ables. Hum Pathol. 1996;27:1124–1134.
45. Landry CS, Brock G, Scoggins CR, et al. A proposed staging 67. Pelosi G, Rodriguez J, Viale G, et al. Typical and atypical
system for gastric carcinoid tumors based on an analysis of 1543 pulmonary carcinoid tumor overdiagnosed as small-cell carcinoma
patients. Ann Surg Oncol. 2009;16:51–60. on biopsy specimens: a major pitfall in the management of lung
46. Landry CS, Woodall C, Scoggins CR, et al. Analysis of 900 cancer patients. Am J Surg Pathol. 2005;29:179–187.
appendiceal carcinoid tumors for a proposed predictive staging 68. Perigny M, Hammel P, Corcos O, et al. Pancreatic endocrine
system. Arch Surg. 2008;143:664–670. (discussion 670). microadenomatosis in patients with von Hippel-Lindau disease:
47. Leja J, Essaghir A, Essand M, et al. Novel markers for characterization by VHL/HIF pathway proteins expression. Am
enterochromaffin cells and gastrointestinal neuroendocrine carci- J Surg Pathol. 2009;33:739–748.
nomas. Mod Pathol. 2009;22:261–272. 69. Perren A, Roth J, Muletta-Feurer S, et al. Clonal analysis of
48. Lejeune M, Jaen J, Pons L, et al. Quantification of diverse sporadic pancreatic endocrine tumours. J Pathol. 1998;186:
subcellular immunohistochemical markers with clinicobiological 363–371.
relevancies: validation of a new computer-assisted image analysis 70. Phan AT, Yao JC, Evans DB. Treatment options for metastatic
procedure. J Anat. 2008;212:868–878. neuroendocrine tumors. Surgery. 2008;144:895–898.
49. Lin O, Olgac S, Green I, et al. Immunohistochemical staining of 71. Plockinger U, Rindi G, Arnold R, et al. Guidelines for the
cytologic smears with MIB-1 helps distinguish low-grade from diagnosis and treatment of neuroendocrine gastrointestinal
high-grade neuroendocrine neoplasms. Am J Clin Pathol. 2003;120: tumours. A consensus statement on behalf of the European
209–216. neuroendocrine tumour society (ENETS). Neuroendocrinology.
50. Lloyd RV. Practical markers used in the diagnosis of neuro- 2004;80:394–424.
endocrine tumors. Endocr Pathol. 2003;14:293–301. 72. Plockinger U, Wiedenmann B. Neuroendocrine tumors. Biother-
51. Lu J, Xue LY, Lu N, et al. Superficial primary small cell carcinoma apy. Best Pract Res Clin Endocrinol Metab. 2007;21:145–162.
of the esophagus: clinicopathological and immunohistochemical 73. Reidy DL, Tang LH, Saltz LB. Treatment of advanced disease
analysis of 15 cases. Dis Esophagus. In press in patients with well-differentiated neuroendocrine tumors. Nat
52. Modlin IM, Kidd M, Latich I, et al. Current status of Clin Pract Oncol. 2009;6:143–152.
gastrointestinal carcinoids. Gastroenterology. 2005;128:1717–1751. 74. Rigaud G, Missiaglia E, Moore PS, et al. High resolution
53. Modlin IM, Kidd M, Drozdov I, et al. Pharmacotherapy of allelotype of nonfunctional pancreatic endocrine tumors: identifi-
neuroendocrine cancers. Expert Opin Pharmacother. 2008;9: cation of two molecular subgroups with clinical implications.
2617–2626. Cancer Res. 2001;61:285–292.
54. Modlin IM, Moss SF, Chung DC, et al. Priorities for improving 75. Rindi G, Kloppel G, Alhman H, et al. TNM staging of foregut
the management of gastroenteropancreatic neuroendocrine tumors. (neuro)endocrine tumors: a consensus proposal including a grading
J Natl Cancer Inst. 2008;100:1282–1289. system. Virchows Arch. 2006;449:395–401.
55. Modlin IM, Oberg K, Chung DC, et al. Gastroenteropancreatic 76. Rindi G, Kloppel G, Couvelard A, et al. TNM staging of midgut
neuroendocrine tumours. Lancet Oncol. 2008;9:61–72. and hindgut (neuro) endocrine tumors: a consensus proposal
56. Modlin IM, Shapiro MD, Kidd M, et al. Siegfried oberndorfer and including a grading system. Virchows Arch. 2007;451:757–762.
the evolution of carcinoid disease. Arch Surg. 2007;142:187–197. 77. Rowe G, Wright G. The Delphi technique as a forecasting tool:
57. Nassar H, Albores-Saavedra J, Klimstra DS. High-grade neuroen- issues and analysis. Intl J Forecasting. 1999;15:353–375.
docrine carcinoma of the ampulla of vater: a clinicopathologic and 78. Saqi A, Alexis D, Remotti F, et al. Usefulness of CDX2 and TTF-1
immunohistochemical analysis of 14 cases. Am J Surg Pathol. 2005; in differentiating gastrointestinal from pulmonary carcinoids. Am
29:588–594. J Clin Pathol. 2005;123:394–404.
58. Nikou GC, Lygidakis NJ, Toubanakis C, et al. Current diagnosis 79. Schmitt AM, Riniker F, Anlauf M, et al. Islet 1 (Isl1) expression is
and treatment of gastrointestinal carcinoids in a series of 101 a reliable marker for pancreatic endocrine tumors and their
patients: the significance of serum chromogranin-A, somatostatin metastases. Am J Surg Pathol. 2008;32:420–425.
receptor scintigraphy and somatostatin analogues. Hepatogastro- 80. Sen N, Calli Demirkan N, Aksoy Altinboga A, et al. Synchronous
enterology. 2005;52:731–741. endocrine tumors of small intestine: report of a case. Turk
59. Oberg K. Somatostatin analog octreotide LAR in gastro-entero- J Gastroenterol. 2008;19:193–196.
pancreatic tumors. Expert Rev Anticancer Ther. 2009;9:557–566. 81. Shia J, Tang LH, Weiser MR, et al. Is nonsmall cell type high-
60. Oberg K, Eriksson B. Nuclear medicine in the detection, staging grade neuroendocrine carcinoma of the tubular gastrointestinal
and treatment of gastrointestinal carcinoid tumours. Best Pract Res tract a distinct disease entity? Am J Surg Pathol. 2008;32:719–731.
Clin Endocrinol Metab. 2005;19:265–276. 82. Soga J. The term ‘‘carcinoid’’ is a misnomer: the evidence based on
61. Oberg K, Jelic S. Neuroendocrine gastroenteropancreatic tumors: local invasion. J Exp Clin Cancer Res. 2009;28:15.
ESMO clinical recommendations for diagnosis, treatment and 83. Solcia E, Fiocca R, Villani L, et al. Hyperplastic, dysplastic, and
follow-up. Ann Oncol. 2008;19(suppl 2):ii104–ii105. neoplastic enterochromaffin-like-cell proliferations of the gastric
62. Panzuto F, Nasoni S, Falconi M, et al. Prognostic factors and survival mucosa. Classification and histogenesis. Am J Surg Pathol. 1995;
in endocrine tumor patients: comparison between gastrointestinal and 19(suppl 1):S1–S7.
pancreatic localization. Endocr Relat Cancer. 2005;12:1083–1092. 84. Srivastava A, Hornick JL. Immunohistochemical staining for
63. Pape UF, Berndt U, Muller-Nordhorn J, et al. Prognostic factors CDX-2, PDX-1, NESP-55, and TTF-1 can help distinguish
of long-term outcome in gastroenteropancreatic neuroendocrine gastrointestinal carcinoid tumors from pancreatic endocrine and
tumours. Endocr Relat Cancer. 2008;15:1083–1097. pulmonary carcinoid tumors. Am J Surg Pathol. 2009;33:
64. Pape UF, Jann H, Muller-Nordhorn J, et al. Prognostic relevance 626–632.
of a novel TNM classification system for upper gastroentero- 85. Suster S, Moran CA. Neuroendocrine neoplasms of the media-
pancreatic neuroendocrine tumors. Cancer. 2008;113:256–265. stinum. Am J Clin Pathol. 2001;115(suppl):S17–S27.

312 | www.ajsp.com r 2010 Lippincott Williams & Wilkins


Am J Surg Pathol  Volume 34, Number 3, March 2010 Pathology Reporting of Neuroendocrine Tumors

86. Tang L, Shia J, Vakiani E, et al. High grade transformation of 93. Wick MR, Graeme-Cook FM. Pancreatic neuroendocrine neo-
differentiated neuroendocrine neoplasms (NENs) of the entero- plasms: a current summary of diagnostic, prognostic, and
pancreatic system-a unique entity distinct from de novo high grade differential diagnostic information. Am J Clin Pathol. 2001;
neuroendocrine carcinoma (HGNECa) in pathogenesis and clinical 115(suppl):S28–S45.
behavior. Mod Pathol. 2008;21:137A. 94. Yao JC. Neuroendocrine tumors. Molecular targeted therapy for
87. Travis WD. The concept of pulmonary neuroendocrine tumours. carcinoid and islet-cell carcinoma. Best Pract Res Clin Endocrinol
In: Travis WD, Brambilla E, Muller-Hermelink HK, et al, eds. Metab. 2007;21:163–172.
Pathology and Genetics of Tumours of the Lung, Pleura, Thymus 95. Yao JC, Hassan M, Phan A, et al. One hundred years after
and Heart. Lyon: IARC Press; 2004:19–20. ‘‘carcinoid’’: epidemiology of and prognostic factors for neuro-
88. Travis WD, Rush W, Flieder DB, et al. Survival analysis of 200 endocrine tumors in 35,825 cases in the United States. J Clin Oncol.
pulmonary neuroendocrine tumors with clarification of criteria 2008;26:3063–3072.
for atypical carcinoid and its separation from typical carcinoid. 96. Yao JC, Hoff PM. Molecular targeted therapy for neuroendocrine
Am J Surg Pathol. 1998;22:934–944. tumors. Hematol Oncol Clin North Am. 2007;21:575–581; x.
89. Van Essen M, Krenning EP, De Jong M, et al. Peptide receptor 97. Yao JC, Phan AT, Chang DZ, et al. Efficacy of RAD001
radionuclide therapy with radiolabelled somatostatin analogues in (everolimus) and octreotide LAR in advanced low- to intermedi-
patients with somatostatin receptor positive tumours. Acta Oncol. ate-grade neuroendocrine tumors: results of a phase II study. J Clin
2007;46:723–734. Oncol. 2008;26:4311–4318.
90. Vilar E, Salazar R, Perez-Garcia J, et al. Chemotherapy and role of 98. Zee SY, Hochwald SN, Conlon KC, et al. Pleomorphic pancreatic
the proliferation marker Ki-67 in digestive neuroendocrine tumors. endocrine neoplasms: a variant commonly confused with adeno-
Endocr Relat Cancer. 2007;14:221–232. carcinoma. Am J Surg Pathol. 2005;29:1194–1200.
91. Volante M, Brizzi MP, Faggiano A, et al. Somatostatin receptor 99. Zhang L, Smyrk TC, Oliveira AM, et al. KIT is an independent
type 2A immunohistochemistry in neuroendocrine tumors: a prognostic marker for pancreatic endocrine tumors: a finding
proposal of scoring system correlated with somatostatin receptor derived from analysis of Islet cell differentiation markers. Am
scintigraphy. Mod Pathol. 2007;20:1172–1182. J Surg Pathol. 2009. In press
92. Wang W, Epstein JI. Small cell carcinoma of the prostate. 100. Zhao J, Moch H, Scheidweiler AF, et al. Genomic imbalances in
A morphologic and immunohistochemical study of 95 cases. the progression of endocrine pancreatic tumors. Genes Chromo-
Am J Surg Pathol. 2008;32:65–71. somes Cancer. 2001;32:364–372.

r 2010 Lippincott Williams & Wilkins www.ajsp.com | 313

Potrebbero piacerti anche