Você está na página 1de 20

EARLY ONLINE RELEASE

Note: This article was posted on the Archives Web site as


an Early Online Release. Early Online Release articles
have been peer reviewed, copyedited, and reviewed by the
authors. Additional changes or corrections may appear in
these articles when they appear in a future print issue of
the Archives. Early Online Release articles are citable by
using the Digital Object Identifier (DOI), a unique number
given to every article. The DOI will typically appear at the
end of the abstract.

The DOI for this manuscript is doi: 10.5858/arpa.2016-0550-RA

The final published version of this manuscript will replace


the Early Online Release version at the above DOI once it is available.

© 2017 College of American Pathologists


Review Article

Application of Immunohistochemistry in the Diagnosis


of Pulmonary and Pleural Neoplasms
Jennifer S. Woo, MD; Opal L. Reddy, MD; Matthew Koo, MD; Yan Xiong, MD; Faqian Li, MD, PhD; Haodong Xu, MD, PhD

 Context.—A vast majority of neoplasms arising from lung pleura–based neoplasms obtained from small biopsy
or pleura are initially diagnosed based on the histologic samples.
evaluation of small transbronchial, endobronchial, or Data Sources.—A literature review of previously pub-
needle core biopsies. Although most diagnoses can be lished articles and the personal experience of the authors
determined by morphology alone, immunohistochemistry were used in this review article.
can be a valuable diagnostic tool in the workup of Conclusion.—Immunohistochemistry is a useful diag-
problematic cases. nostic tool in the workup of small biopsies from the lung
Objective.—To provide a practical approach in the and pleura sampled by small biopsy techniques.
interpretation and immunohistochemical selection of lung/ (Arch Pathol Lab Med. doi: 10.5858/arpa.2016-0550-RA)

B iopsies of lung and pleural neoplasms play an important


role in the characterization and staging of lung cancers,
particularly for patients with advanced disease (stages III
PRIMARY EPITHELIAL TUMORS OF LUNG
An 82-year-old nonsmoking man presented with a 4.7 3
3.6-cm right upper lobe mass by computed tomography
and IV) and for patients who are not candidates for surgical (CT) chest imaging. A CT-guided biopsy demonstrated a
procedures. Clinical and imaging correlation in conjunction sheetlike proliferation of poorly differentiated malignant
with morphologic assessment by routine hematoxylin-eosin epithelial cells (Figure 1, A). The tumor cells were positive
staining is crucial in the interpretation of small biopsies. for p40 (Figure 1, B) and cytokeratin 5/6 (CK5/6) (Figure 1,
However, immunohistochemistry (IHC) may be a valuable C), but negative for thyroid transcription factor 1 (TTF-1)
diagnostic tool in the workup of challenging cases. For (Figure 1, D) and napsin A (Figure 1, E). The final diagnosis
example, it can help differentiate between lung adenocar- was poorly differentiated SqCC.
cinoma and squamous cell carcinoma (SqCC), lung adeno- A 66-year-old Asian woman presented with a poorly
carcinoma and malignant mesothelioma (MM), primary and circumscribed, solid 2.6 3 2.5-cm right upper lobe mass by
metastatic carcinomas, and small cell lung carcinoma CT chest imaging. A CT-guided biopsy demonstrated a solid
(SCLC) and carcinoid tumor. It can also help detect growth pattern of poorly differentiated malignant epithelial
oncogenic mutants, such as endothelial growth factor cells (Figure 2, A). The tumor cells were positive for TTF-1
receptor (EGFR), anaplastic lymphoma kinase (ALK), and (Figure 2, B) and napsin A (Figure 2, C), but negative for p40
repressor of silencing 1 (ROS1), and immune checkpoint (Figure 2, D) and CK5/6 (Figure 2, E). The final diagnosis
was poorly differentiated adenocarcinoma with a solid
molecules, such as programmed death ligand-1 (PD-L1).
pattern of growth. Molecular studies showed that the tumor
This article provides an up-to-date review of the role of IHC
cells were positive for an EGFR exon 19 deletion.
in the workup of common entities seen in the small lung/
Historically, the most clinically significant distinction
pleural biopsy setting while using examples. among lung tumors was the distinction between non–small
cell lung carcinoma (NSCLC) and SCLC. However, because
Accepted for publication December 15, 2016. of markedly different prognostic and treatment implications
From the Department of Pathology and Laboratory Medicine, among different lung tumors, accurate subtyping with
David Geffen School of Medicine at UCLA, Los Angeles, California molecular characterization is critical. This is especially true
(Drs Woo, Reddy, Koo, and Xu); the Department of Pathology, Peking for the most common NSCLC types, lung adenocarcinoma
University First Hospital, Beijing, China (Dr Xiong); and the and SqCC, as treatment options and the role of ancillary
Department of Laboratory Medicine and Pathology, University of
molecular and cytogenetic studies widely differ for both
Minnesota, Minneapolis (Dr. Li).
The authors have no relevant financial interest in the products or tumors. Specifically, EGFR inhibitors such as erlotinib (OSI
companies described in this article. Pharmaceuticals, Melville, New York; Hoffmann-La Roche,
Presented at the First Chinese American Pathologists Association Basel, Switzerland; Genentech, South San Francisco, Cal-
(CAPA) Diagnostic Pathology Course: Best Practices in Immunohis- ifornia) and gefitinib (AstraZeneca, London, United King-
tochemistry in Surgical Pathology and Cytopathology; Flushing, New dom) have been shown to be effective in EGFR-mutated
York; August 22–24, 2015.
Reprints: Haodong Xu, MD, PhD, Department of Pathology and
tumors,1 and the ALK inhibitor crizotinib (Pfizer, New York,
Laboratory Medicine, David Geffen School of Medicine at UCLA, New York) has been shown to be effective in tumors with
10833 Le Conte Ave, CHS 13-145E, Los Angeles, CA 90095-1732 EML4-ALK fusion,2 which are both predominantly seen
(email: HaodongXu@mednet.ucla.edu). with adenocarcinomas. Additionally, several therapeutic
Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 1
Figure 1. Poorly differentiated squamous cell carcinoma of the lung. The needle core biopsy shows sheets of malignant large cell cells without
obvious squamous differentiation (A). The tumor cells are positive for p40 (B) and cytokeratin 5/6 (C). The tumor cells are negative for both thyroid
transcription factor 1 (D) and napsin A (E) (hematoxylin-eosin, original magnification 3400 [A]; original magnification 3400 [B through E]).

2 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al


Figure 2. Poorly differentiated adenocarcinoma of the lung. The needle core biopsy shows a solid pattern of malignant large cells without glandular
formation (A). The tumor cells are positive for thyroid transcription factor 1 (B) and napsin A (C). The tumor cells are negative for both p40 (D) and
cytokeratin 5/6 (E) (hematoxylin-eosin, original magnification 3400 [A]; original magnification 3400 [B through E]).

Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 3


Table 1. Common Immunohistochemical Stains Immunohistochemical markers for SqCC include p63,
Used to Differentiate Pulmonary Squamous Cell CK5/6, cytokeratin 34bE12 (CK903), and p40. The p63
Carcinoma From Adenocarcinoma antibody, with numerous studies showing excellent sensi-
tivity as a marker for SqCC,19–21 has long been the most
Squamous Cell commonly used nuclear marker for squamous origin.
Carcinoma Adenocarcinoma
However, p63 has been shown to be positive in 16% to
CK5/6 TTF-1–SPT24 (lower specificity) 65% of lung adenocarcinoma20,22–24 and in 55 of 172 cases
p40 (higher specificity) TTF-1–8G7G3/1 (higher specificity) (32%) of diffuse large B cell lymphoma.25
p63 (lower specificity) Napsin A
CK903/34bE12 p63 consists of several isoforms. The 2 major groups
include DNp63 and TAp63.26 DNp63 is the predominant p63
Abbreviations: CK, cytokeratin; TTF-1, thyroid transcription factor 1.
transcript in SqCC of lung and functions as an oncogene,27,28
whereas TAp63 has been shown to function as a p53-like
agents used in the treatment of adenocarcinoma are tumor suppressor.26 In the majority of pathology laborato-
contraindicated in SqCC, including pemetrexed (Alimta, ries, the routine p63 antibody is 4A4, which recognizes both
Eli Lilly and Company, Indianapolis, Indiana), because of TAp63 and DNp63 isoforms. The p40 antibody, which
the lack of effectiveness,3 and the vascular endothelial exclusively recognizes the DNp63 isoform, may have a
growth factor (VEGF) inhibitor bevacizumab (Avastin, superior specificity29 but inferior sensitivity30–32 compared
Genentech), because of the risk of life-threatening pulmo- with p63 in the diagnosis of pulmonary SqCC.
nary hemorrhage.4 Two genetic alterations have been Previous studies have demonstrated that a majority of
recently identified in pulmonary SqCC, discoidin domain poorly differentiated NSCLCs can be subclassified as
receptor tyrosine kinase 2 (DDR2) mutation5 and fibroblast adenocarcinoma or SqCC by IHC using a simple panel of
growth factor receptor 1 (FGFR1) amplification,6 both of IHC markers.19,33,34 For limited biopsy samples, we recom-
which have potential as novel therapeutic targets. mend an initial IHC panel of TTF-1 and p40. If needed,
In most instances, the morphologic distinction between additional squamous markers (p63 and CK5/6) or glandular
adenocarcinoma and SqCC is relatively straightforward: the markers (napsin A and CK7) may be added. The distinction
formation of glands and the production of cytoplasmic between adenocarcinoma and SqCC is generally straightfor-
mucin are characteristic of adenocarcinoma, whereas the ward when following this IHC approach. One or more
production of keratin and the presence of intercellular markers of glandular differentiation (TTF-1 or napsin A) with
bridges are characteristic of SqCC. However, the pathologist negative squamous markers is supportive of adenocarcinoma.
may run into diagnostic challenges in the setting of a poorly Conversely, one or more positive squamous markers (p40 or
differentiated tumor, or tumors with nonspecific morpho- p63), in the context of negative glandular markers, is
logic findings, especially in the biopsy setting. In this case, suggestive of SqCC. Cases of TTF-1– and/or napsin A–
the judicious use of IHC markers may be helpful for further positive adenocarcinoma may show focal p63 expression.19
characterization (Table 1). Interestingly, diffuse coexpression of TTF-1 and p63 may be
The most useful IHC markers for pulmonary adenocar- seen in adenocarcinomas with signet ring cell morphology
cinoma include TTF-1 and napsin A. TTF-1 is a homeodo- (many of which contain EML4-ALK translocations).35 Ad-
main-containing transcription factor that is predominantly enosquamous carcinoma requires the presence of 2 separate
found in normal type II alveolar pneumocytes.7 With a components demonstrating opposite and mirror-image
reported sensitivity ranging from 75% to 80%,8,9 TTF-1 has staining patterns (napsin A and TTF-1 positive and p40 and
long been the predominant nuclear IHC marker used to CK5/6 negative in the glandular component, with napsin A
identify cells of lung origin. However, TTF-1 expression has and TTF-1 negative and p40 and CK5/6 positive in the
been shown to decrease inversely with the degree of tumor squamous component).36 Nevertheless, in small biopsy
differentiation.8–10 Of the 2 main commercial TTF-1 mono- samples, the ability to definitively diagnose adenosquamous
clonal antibodies available for IHC staining (8G7G1/1 and carcinoma may be particularly challenging.
SPT24), the SPT24 clone has been shown to have a stronger Isolated p63 positivity in scattered cells is a nonspecific
affinity for TTF-1. However, the SPT24 TTF-1 clone may also staining pattern that does not contribute to the further
show an affinity for colorectal adenocarcinomas.11 classification of the lesion. Similarly, isolated CK5/6 staining
Napsin A, a relatively new cytoplasmic IHC marker for in scattered cells, in the absence of positivity for additional
pulmonary adenocarcinoma, is an aspartic proteinase squamous or glandular markers, is also a nonspecific
involved12 in the maturation of surfactant protein B that finding. In these cases, or in cases with nonreactivity to
has an expression thought to be regulated by TTF-1.13 the entire proposed IHC panel, the most suitable diagnosis
Napsin A has been shown to be superior to TTF-1 in would be NSCLC, not otherwise specified.37
distinguishing primary lung adenocarcinomas from other A major pitfall in interpreting IHC-stained slides of small
carcinomas (except for renal cell carcinoma,14,15 clear cell lung biopsy samples is the positivity of normal alveolar
carcinomas of the gynecologic tract,16,17 and thyroid epithelium for TTF-1 and/or napsin A. Therefore, the proper
carcinomas15). Napsin A, with a higher specificity but a interpretation of immunomarkers requires morphologic
lower sensitivity for adenocarcinoma than TTF-1, was also correlation. In addition, alveolar macrophages show cyto-
found to be a useful marker in cases of poorly differentiated plasmic napsin A staining. Prior to the availability of IHC
lung adenocarcinoma or an unknown primary tumor.17 stains, pathologists heavily relied on the histochemical
Another IHC marker for glandular origin may be the mucin stain to confirm a diagnosis of adenocarcinoma. Even
cytoplasmic immunostain cytokeratin 7 (CK7). However, today, despite a low sensitivity,38–40 special stains for
CK7 reactivity can be seen in most poorly differentiated cytoplasmic mucin may still prove to be useful in particularly
adenocarcinomas and has been reported in 9 of 15 challenging cases that show nonspecific staining patterns.
morphologically challenging cases of poorly differentiated Furthermore, in this age of targeted therapies, molecular
SqCCs (60%), which limits its diagnostic utility.18 and cytogenetic studies play a critical role in the workup of
4 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al
primary lung malignancies, especially in NSCLC. Although T-cell function. Recent clinical trials have shown that anti–
molecular and cytogenetic testing will remain the gold- PD-1 therapy in NSCLC demonstrates antitumor activity in
standard methodology for the detection of specific somatic patients with advanced NSCLC.60–64 The US Food and Drug
genetic mutations, IHC has emerged as a fairly effective and Administration has approved inhibitors targeting PD-1 for
rapid means for identifying these genetic variants. One of patients with NSCLC. Currently, a number of different anti–
the most significant mutations in NSCLC includes sensitiz- PD-L1 antibodies by various vendors have been formally
ing mutations of EGFR, particularly in exons 18–21. EGFR studied. In the future, pathologists may expect to become
mutations are important to identify, as neoplasms demon- more familiar with PD-L1 as an important NSCLC
strating these mutations may be susceptible to EGFR– predictive or companion biomarker. Figure 7, A, shows a
tyrosine kinase inhibitor (TKI) therapy (ie, erlotinib; pleomorphic carcinoma with a prominent spindle cell
gefitinib; afatinib, Boehringer Ingelheim, Ingelheim, Ger- component that is strongly positive for TTF-1 (Figure 7, B)
many]). The 2 most common EGFR mutations include in- and PD-L1 (Figure 7, C) by using a mouse monoclonal
frame deletions in exon 19 (E746_A750del) and a point antibody against PD-L1 (clone 22C3; DAKO, Carpinteria,
mutation at codon 858 in exon 21 (L858R), together California).
representing 85% to 90% of EGFR mutations in NSCLC
patients.41,42 Mutation-specific antibodies for these 2 genetic PLEURAL MM VERSUS LUNG CARCINOMA
mutations were developed43 in 2009, and numerous studies A 60-year-old man presented with multiple right pleural
have since demonstrated that IHC may be used as a reliable masses on CT imaging. A right pleural biopsy was
prescreening test for detecting EGFR mutations in performed at an outside institution and a diagnosis of
NSCLC.44–47 A moderately differentiated adenocarcinoma epithelioid MM was rendered. The patient was evaluated for
of the lung shows an acinar pattern of growth (Figure 3, A) a decortication of pleural mesothelioma and a rebiopsy was
and contains an EGFR mutant with E746-A750 (Figure 3, B)
performed. Biopsies of the right pleural masses demon-
detected by using rabbit monoclonal antibody against EGFR
strated a biphasic MM (75% epithelioid type, 25% sarco-
E746-A750 (clone 6B6; Cell Signaling Technology, Danvers,
matoid type) (Figure 8, A). The tumor cells were positive for
Massachusetts); a poorly differentiated adenocarcinoma of
AE1/AE3 (Figure 8, B), calretinin (Figure 8, C), and Wilms
the lung exhibits a solid pattern of growth (Figure 4, A) and
tumor 1 (WT-1) (Figure 8, D).
harbors an EGFR mutant with L835R (Figure 4, B) detected
Malignant mesothelioma of the pleura is a rare neoplasm
by using rabbit monoclonal antibodies against EGFR L835R
arising from mesothelial cells. Although known for a great
(clone 43B2; Cell Signaling Technology).
diversity of histologic patterns, MMs are generally divided
Chromosomal rearrangements of the ALK gene, although
found in a minority (0.4%–15%) of NSCLCs,48,49 are also into 3 major histologic types: epithelioid, sarcomatoid, and
important to identify, as these tumors may be susceptible to biphasic (mixed epithelioid and sarcomatoid).65 Given the
ALK inhibitors such as crizotinib. Recent studies have also wide range of morphologic features, the differential
shown that the use of ALK antibodies may also be an diagnosis of MM may significantly vary depending on the
effective prescreening tool to detect the presence of ALK histologic type. For epithelioid MM, the differential diag-
rearrangements in addition to the conventional ALK nosis may include carcinoma (including primary adenocar-
fluorescence in situ hybridization testing.50,51 A moderately cinoma, metastatic adenocarcinoma, or SqCC) as well as
differentiated adenocarcinoma of the lung shows an acinar other tumors with epithelioid features. Sarcomatoid MM
pattern of growth (Figure 5, A) and expresses ALK protein raises the differential diagnosis of sarcomatoid carcinoma,
(Figure 5, B) detected by using a rabbit monoclonal antibody malignant tumors with sarcomatoid features, and a variety
against ALK (clone D5F3; Cell Signaling Technology). of malignant sarcomas, including undifferentiated pleomor-
Immunohistochemical stains have also been developed to phic sarcoma (formerly malignant fibrous histiocytoma),
detect rearrangements of ROS1 (present in 1%–2% of osteosarcoma, and chondrosarcoma. Lymphoma, melano-
NSCLC),52,53 which encodes for a receptor tyrosine kinase. ma, angiosarcoma, and epithelioid hemangioendothelioma
Several studies suggest that ROS1 may represent another are also on the differential diagnosis, as are benign
therapeutic target of the ALK inhibitor crizotinib,52–54 with a mesothelial proliferations. Biphasic patterns should raise
recent study showing a marked antitumor activity by the differential diagnosis of other biphasic tumors such as
crizotinib in patients with ROS1-rearranged advanced synovial sarcoma.
NSCLC.55 Similar to IHC for ALK, IHC stains for ROS1 The most important consideration for the pathologist is
have been shown to be an effective and cost-effective means the patient’s clinical and radiologic features in correlation
to screen for ROS1 rearrangements.56–58 A poorly differen- with the hematoxylin-eosin morphology. However, IHC can
tiated adenocarcinoma of the lung shows acinar and be helpful in confirming MM, and the initial selection of
micropapillary patterns of growth (Figure 6, A) and stains will vary depending on the histologic type. In general,
expresses ROS1 (Figure 6, B), detected by using a rabbit nearly all mesothelial cells (including mesotheliomas) are
monoclonal antibody against ROS1 (clone D4D6; Cell positive for pancytokeratin (including AE1/AE3).66 However,
Signaling Technology). Therefore, the judicious use of sarcomatoid MM may show loss of keratin reactivity. In
mutation-specific IHC stains may be used as a practical these cases, a cocktail of keratins may improve detection.67
screening tool to detect certain actionable mutations (EGFR, Furthermore, sarcomatoid MM with either osteosarcoma-
ALK, and ROS1) amenable to targeted therapy. tous or chondrosarcomatous differentiation is known to be
In addition to the ROS1, ALK, and EGFR pathways, typically negative for keratin staining.
programmed death receptor-1 (PD-1), an immunoregula- Other mesothelial markers include calretinin (strong and
tory receptor expressed by activated T cells,59 and its ligand, diffuse nuclear and cytoplasmic staining), CK5/6 (cytoplas-
PD-L1 in cancer cells, have been shown to be promising mic staining), D2-40 (membranous staining), and WT-1
targets in NSCLC. Targeted therapies against PD-1 have (nuclear staining).66 It must be noted that because of the
been postulated to treat cancers by restoring proper effector variability of staining qualities among different antibody
Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 5
Figure 3. Moderately differentiated adenocarcinoma of the lung. The biopsy shows a moderately differentiated adenocarcinoma with an acinar
pattern (hematoxylin-eosin, A). The tumor cells express the EGFR mutant protein, E746-A750 (B) (hematoxylin-eosin, original magnification 3200
[A]; original magnification 3200 [B]).
Figure 4. Poorly differentiated adenocarcinoma of the lung. The biopsy shows poorly differentiated adenocarcinoma with a solid pattern (A). The
tumor cells express the EGFR mutant protein L858R (B) (hematoxylin-eosin, original magnification 3200 [A]; original magnification 3200 [B]).

clones and IHC laboratories, no single specific panel of IHC For the initial workup of an epithelioid MM, where the
antibodies can be recommended universally. In general, the main differential diagnoses are MM versus carcinoma, a
pathologist should become familiar with the staining pattern reasonable panel may include 2 mesothelial markers and 2
at his or her performing laboratory and take into account carcinoma markers (Table 2). An expanded panel may be
this knowledge when selecting the initial panel of stains. necessary if the initial panel reveals discordant results or if a
Immunohistochemistry laboratories should aim for a metastatic adenocarcinoma is suspected. Immunostains
sensitivity of at least 80% when validating immunostains positive for adenocarcinoma include TTF-1, napsin A, Ber-
for this purpose.68 EP4, carcinoembryonic antigen (CEA), Leu-M1, and MOC-
6 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al
Figure 5. Moderately differentiated adenocarcinoma of the lung. The biopsy shows a moderately differentiated adenocarcinoma with an acinar
pattern (A). The tumor cells express ALK protein (B) (hematoxylin-eosin, original magnification 3200 [A]; original magnification 3200 [B]).
Figure 6. Poorly differentiated adenocarcinoma of the lung. The biopsy shows a poorly differentiated adenocarcinoma with acinar and
micropapillary patterns (A). The tumor cells express ROS1 protein (B) (hematoxylin-eosin, original magnification 3200 [A]; original magnification
3200 [B]).

31. TTF-1 and napsin A have the additional benefit of positive in SqCC include p40, p63, MOC-31, and Ber-EP4.69
helping to confirm a primary lung origin. In differentiating Additional immunomarkers that may be helpful, depending
MM from adenocarcinoma, calretinin, CK5/6, D2-40, and on the differential diagnosis, include CD45 (for lymphoma);
WT-1 are very useful mesothelial markers. However, in S100, HMB-45, MART1, and SOX10 (for melanoma; both
differentiating MM from SqCC, the role of mesothelial S100 and SOX10 are more sensitive for spindle cell
markers is more limited, owing to the fact that SqCCs may melanoma)70; and CD31, CD34 and ERG1 (for angiosarco-
be positive for CK5/6, D2-40, and (to a lesser extent) ma and epithelioid hemangioendothelioma).
calretinin. Therefore, WT-1 is the most clinically useful For sarcomatoid MM, an initial panel may include
immunostain in differentiating MM from SqCC. Markers multiple cytokeratins, calretinin, and D2-40. Multiple
Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 7
keratin antibodies including AE1/AE3, CAM 5.2, and CK7
are recommended, as cytokeratin expression may be absent
or variable in sarcomatoid mesothelioma.71,72 However,
because sarcomatoid carcinoma and metastatic sarcomatoid
renal cell carcinoma may show the same staining pattern,
diffuse keratin positivity with sarcomatoid morphology is
not by itself diagnostic for sarcomatoid mesothelioma.
Metastatic renal cell carcinoma may be highlighted with
paired box gene 2 (PAX2) or PAX8 staining. Moreover, focal
keratin positivity must be interpreted with caution, as a
number of sarcomas may demonstrate focal keratin
positivity.67 For this reason, markers for sarcomatous
differentiation, such as smooth muscle actin (SMA), desmin,
and myoglobin, may be helpful.
Useful markers for sarcomatoid MM include calretinin
and D2-40,73,74 although the number of tumor cells positive
for these markers is variable. Additional immunostains to
distinguish between MM and other sarcomas may include
SMA, desmin, myoglobin and myogenin. Overall, no single
immunostain by itself is particularly helpful for the diagnosis
of sarcomatoid mesothelioma. A carefully chosen panel of
immunostains is the key in the workup of such cases.
Immunohistochemical staining with equivocal or nondiag-
nostic results may benefit from ultrastructural examination
by electron microscopy. However, the diagnostic utility of
electron microscopy may be more limited in the setting of
poorly differentiated tumors.75,76
Benign mesothelial proliferations are also in the differen-
tial diagnosis of MM. Morphologic features favoring
malignancy include dense cellularity, complex papillae,
tubules, cellular stratification, and necrosis.67 However,
because benign and malignant mesothelial cells may share
overlapping cytologic features, the presence of stromal
invasion is considered the most reliable feature for
separating benign versus MM proliferations.77–80 The inva-
sion of stroma or fat by atypical mesothelial cells may be
highlighted by immunostains including AE1/AE3 or calre-
tinin. In this case, the pathologist must determine whether
mesothelial cells represent entrapped benign cells or benign
cells cut en face versus true invasive malignancy. To
distinguish fatlike spaces that may be seen in organizing
pleuritis,81 S100, laminin, and collagen IV stains may be
performed to highlight adipocytes, whereas fatlike spaces
will be negative.
Immunohistochemical stains to distinguish between
benign and malignant mesothelial proliferations may be
helpful, but are often not entirely definitive for malignancy
or benignity (Table 3). Epithelial membrane antigen (EMA)
and p53 have been described as immunomarkers for
malignancy,82–85 whereas desmin has been described as an
indicator of benign mesothelial cells.84,85 A review by King et
al84 demonstrated sensitivity and specificity of desmin and
EMA to be less than 90%, which may be deemed not
sufficient enough when distinguishing benign from malig-
nant.
Several recent studies have reported insulin-like growth
factor II messenger ribonucleic acid–binding protein 3

Figure 7. Pleomorphic carcinoma with prominent spindle cell


component of the lung. The biopsy shows a proliferation of malignant
spindle cells (A), with tumor cells positive for thyroid transcription
factor 1 (B). The tumor cells are strongly positive for programmed death
receptor-1 (C) (hematoxylin-eosin, original magnification 3200 [A];
original magnification 3200 [B and C]).

8 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al


Figure 8. Biphasic pleural malignant mesothelioma. The biopsy shows a sheet of malignant epithelioid cell proliferation with focal malignant spindle
cells (A). The tumor cells are positive for AE1/AE2 (B), calretinin (C), and Wilms tumor 1 (D) (hematoxylin-eosin, original magnification 3400 [A];
original magnification 3400 [B through D]).

(IMP3) and glucose transporter 1 (GLUT1) to be useful benign and malignant proliferations has not been report-
biomarkers for differentiating MM from reactive mesothelial ed.88
cells. IMP3 is an oncofetal protein involved in embryogen- Overall, positive staining for IMP3 and/or GLUT1 may be
esis. GLUT-1 is a member of the GLUT family of passive helpful to confirm the diagnosis of malignancy; however,
carriers, which functions as an energy-independent system negative staining for IMP3 and/or GLUT1 does not
for transport of glucose. Both IMP3 and GLUT1 have been
completely rule out MM. BRCA1-associated protein 1
reported in a variety of carcinomas.86,87 Multiple studies
suggest that both epithelioid and sarcomatoid MMs are (BAP1) has most recently emerged as a promising biomark-
more likely to be positive for IMP3 (diffuse dark brown er for pleural MM, with loss of nuclear BAP1 staining seen
cytoplasmic staining), whereas reactive mesothelial prolif- in some mesotheliomas, but none of the benign cases.92,93
erations are more likely to negative for IMP3.88–91 A BAP1 IHC has been shown to have a relatively high
consistent difference in staining intensity of IMP3 between specificity (100%) but low sensitivity (27%).93
Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 9
Table 2. Common Immunohistochemical Stains Table 3. Common Immunohistochemical Stains
Used to Differentiate Pulmonary Malignant Used to Differentiate Pulmonary Malignant
Mesothelioma From Adenocarcinoma Mesothelioma From Reactive Mesothelial cells
Malignant Mesothelioma Adenocarcinoma Malignant Reactive
Mesothelioma Mesothelial Cells
CK5/6 TTF-1
Calretinin Napsin A p53 Desmin
D2-40 Ber-EP4 EMA IMP3—less frequently
WT-1 B72.3 positive
CEA IMP3—frequently positive GLUT1—less frequently
Leu-M1 positive
MOC-31 GLUT1—frequently positive
p16 gene homozygous
Abbreviations: CEA, carcinoembryonic antigen; CK, cytokeratin; TTF-1,
thyroid transcription factor-1; WT-1, Wilms tumor 1. deletion
BAP1 (loss of nuclear stain)
Abbreviations: BAP1, BRCA1 associated protein 1; EMA, epithelial
Homozygous p16 (CDKN2A) gene deletion identified membrane antigen; GLUT1, glucose transporter 1; IMP3, IG2BP3,
using fluorescence in situ hybridization techniques has also insulin-like growth factor mRNA-binding protein.
been described as an indicator of malignancies, including
MM.94,95 However, fluorescence in situ hybridization testing and thyroid, and is expressed in approximately 75% of lung
has a number of limitations, such as artifactual loss of p16 adenocarcinomas.99 However, it is also highly sensitive and
due to section thickness of formalin-fixed, paraffin-embed- specific for thyroid carcinomas, with the exception of
ded tissue (tumor cells may not be in the plane of section anaplastic carcinoma and a small proportion of other
and may appear to lose one or more copies of the gene) and extrapulmonary adenocarcinomas (including ovarian serous
difficulty of picking out individual cells of interest.80 carcinomas, endometrial and endocervical adenocarcino-
Therefore, a fresh nodule of cells selected for fluorescence mas, and colonic adenocarcinomas).99,100 Napsin A is
in situ hybridization studies may be more reliable. Addi- predominantly expressed in the lung and kidney, and
tionally, homozygous p16 deletion is present in only a demonstrates granular and cytoplasmic staining.101 It has a
subset of mesotheliomas. Therefore, the absence of p16 comparable sensitivity to that of TTF-1 in identifying lung
gene deletion cannot entirely exclude the diagnosis of adenocarcinomas. It is important to be aware that the 2
malignancy.96 markers may not always be coexpressed.101 Similar to TTF-1,
In summary, the role of IHC can be helpful to confirm a napsin A is not entirely specific for lung adenocarcinomas,
diagnosis of MM, and the precise IHC panel should be as its expression is seen in a majority of papillary renal cell
selected with a careful consideration of the differential carcinomas (75%–80%), a subset of clear cell renal cell
diagnosis. However, caution should be made when inter- carcinomas (approximately 30%), rare cases of papillary
preting immunostains, because of staining variability among thyroid carcinomas, and a significant proportion of ovarian
different antibody clones and individual IHC laboratories. and endometrial clear cell carcinomas.15,100,102 Therefore,
Because of this, IHC laboratories should aim for a sensitivity although TTF-1 and napsin A may be used to support a
of at least 80%.67 Electron microscopy and cytogenetic diagnosis of primary lung adenocarcinoma, in cases where
studies may be helpful in difficult circumstances, although morphology or clinical history may implicate an extrapul-
their diagnostic ability may be limited in the setting of monary primary, other organ-specific biomarkers may be
poorly differentiated tumors. needed.
The above clinical example describes the frequent
PRIMARY LUNG TUMORS VERSUS METASTASIS challenge of differentiating a primary lung adenocarcinoma
A 76-year-old woman with a remote history of breast from a metastatic breast carcinoma. Women with breast
carcinoma and bilateral lung adenosquamous carcinoma cancer have a 30% higher risk than the general population
presented with an enlarging pleural nodule at the lingula. A of developing a second primary malignancy, with approx-
CT-guided biopsy of the pleural nodule revealed adenocar- imately 4% to 9% developing lung cancer.103 In this case, a
cinoma (Figure 9, A) positive for GATA-binding protein 3 panel of TTF-1, napsin A, mammaglobin, GCDFP-15, and
(GATA3) (Figure 9, B), progesterone receptor (PR) (Figure 9, GATA3 was used to support the diagnosis of breast
C), estrogen receptor (ER) (Figure 9, D), mammaglobin carcinoma metastatic to the lung. Both TTF-1 and napsin
(Figure 9, E), and gross cystic disease fluid protein 15 A have been shown to be negative in breast adenocarcino-
(GCDFP-15) (focal), but negative for TTF-1 (Figure 9, F) and ma.103 Mammaglobin and GCDFP-15 are established
p40. The overall findings were consistent with metastatic cytoplasmic markers for metastatic breast carcinomas, with
breast carcinoma to the pleura. positivity in 85% and 53% of breast carcinomas and up to
Metastases from the extrapulmonary sites represent the 17% and 2% of lung carcinomas, respectively.9 GATA3, a
most common form of pulmonary neoplasm, as the lungs nuclear marker, was found to be expressed in 90 of 99 of
are one of the most common sites of distant metastases.97 breast ductal carcinomas (91%) and 48 of 48 breast lobular
Accordingly, although pathologists may more frequently carcinomas (100%), but was also seen in 62 of 72 urothelial
encounter primary lung neoplasms, differentiating primary carcinomas (86%),104 61 of 62 basal cell carcinomas of the
lung malignancies from metastatic neoplasms, especially skin (98%), 25 of 31 SqCCs of the skin (81%), 11 of 11
from poorly differentiated extrapulmonary adenocarcino- choriocarcinomas (100%), 6 of 6 endodermal sinus tumors
mas, can be particularly challenging. (100%), 37 of 64 MMs (58%), 18 of 22 extra-adrenal
TTF-1 and napsin A are 2 widely used markers to confirm paragangliomas (82%), and 22 of 24 pheochromocytomas
adenocarcinomas of pulmonary origin.15,98 TTF-1 is a (92%).105 GATA3 expression may also have prognostic
nuclear protein involved in the organogenesis of the lung implications in patients with breast cancer, where some
10 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al
Figure 9. Metastatic breast carcinoma of the lung. The biopsy shows a moderately differentiated adenocarcinoma with glandular formation (A). The
tumor cells are positive for GATA3 (B), progesterone receptor (C), estrogen receptor (D), and mammaglobin (E). The tumor cells are negative for
thyroid transcription factor 1 (F) (hematoxylin-eosin, original magnification 3200 [A]; original magnification 3200 [B through F]).

Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 11


Table 4. Common Immunohistochemical Stains negative, CK20 positive).100 TTF-1 and napsin A can also be
Used to Differentiate Primary Lung Carcinoma From included in the panel to support a lung primary, along with
Carcinoma of Breast, Prostate, and Colorectal Origin caudal type homeobox 2 (CDX2), a highly sensitive nuclear
marker of intestinal adenocarcinomas, to support a colo-
Lung Breast Prostatic Colorectal
Origin Origin Origin Origin
rectal origin.117 However, caution should be taken when
interpreting this panel on mucinous adenocarcinoma of the
TTF-1a Mammaglobin PSA CDX2b lung, as this subtype frequently shifts away from its lung
Napsin A GCDFP-15 PSAP
GATA3 NKX3.1
phenotype, and may be negative for TTF-1 and napsin A
ERG and positive for CK20 and CDX2.100,118 Moreover, TTF-1 has
also been found to be positive in a subset of colorectal
Abbreviations: ERG, v-ets erythroblastosis virus E26 oncogene homo-
logue; GCDFP-15, gross cystic disease fluid protein 15; PSA, prostate- carcinomas.11 In a study of 555 colorectal adenocarcinomas,
specific antigen; PSAP, prostate-specific acid phosphatase; TTF-1, TTF-1 was positive in 18 (3.2%) and 24 cases (4.3%), using
thyroid transcription factor 1. the 8G7G3/1 and SPT24 clones, respectively.119
a
TTF-1 may be positive in a subset of colorectal carcinomas. For other extrapulmonary adenocarcinomas, PAX8 has
b
CDX2 may be positive in mucinous adenocarcinoma of the lung. also been recently proposed as a third immunomarker used
alongside TTF-1 and napsin A to distinguish primary lung
studies have suggested that higher levels predict improved adenocarcinomas from metastatic neoplasms.120 PAX8 is a
survival.103,106 GATA3 may also stain background lympho- nuclear protein that is negative in primary lung adenocar-
cytes. Lastly, the use of this panel of immunostains can also cinomas, and has positive expression restricted to epithelial
aid with the diagnosis in cytology specimens. Mammaglobin tumors of the thyroid, kidney, Müllerian system, and
and GCDFP-15 have high specificities for breast carcinomas thymus.121
on cell block (88% and 96%, respectively), but have In summary, to distinguish a primary pulmonary adeno-
suboptimal sensitivities (26% and 14%, respectively), carcinoma from a metastatic malignancy, an initial panel of
especially relative to that of GATA3 (86%).107,108 A immunostains should include TTF-1, napsin A, CK7, and
combined double stain of TTF-1 and napsin A has also CK20. Organ-specific markers should be added according to
been proposed for these limited specimens, and it has been clinical suspicion and morphologic impression. Breast
demonstrated to be diagnostically useful in identifying cancer markers include GATA3, mammaglobin, and
pulmonary adenocarcinomas on cell block.109 GCDFP-15. For prostate cancer, prostate-specific antigen
In addition to breast cancer, prostate and colorectal and prostate-specific acid phosphatase are established
cancers are 2 of the most common malignancies that lead markers capable of identifying more than 90% of metastatic
to pulmonary metastases. The common IHC stains used to tumors. Newer markers for prostate cancer include NKX3.1,
differentiate primary lung carcinoma from carcinomas of as well as ERG, a highly specific marker in identifying
breast, prostate, and colorectal origin are summarized in prostate tumors affected by a TMPRSS2-ERG gene fusion,
Table 4. In the United States, prostatic adenocarcinoma is although it is a less sensitive marker overall. CDX2 may be
the most common type of cancer in males and is estimated added to identify metastatic colorectal adenocarcinomas,
to metastasize to the lungs in 46% of cases of metastatic although caution should be taken when primary mucinous
prostate cancer. In this setting, it is helpful to note that adenocarcinomas of the lung is in the differential, as this
metastatic prostate cancer rarely presents as an isolated subtype may mimic the immunophenotype of colorectal
metastasis.110,111 Cytokeratin 7 can be used to differentiate carcinoma. PAX8 offers high utility in identifying metastatic
lung adenocarcinoma from prostatic acinar carcinoma (more adenocarcinomas to the lung, because it is negative in
than 90% of which are negative for CK7 and cytokeratin 20 primary lung adenocarcinoma but positive in epithelial
[CK20]), with the exception of prostatic ductal carcinoma, tumors of the thyroid, kidney, Müllerian system, and
which can be CK7 positive.101 Prostate-specific antigen and thymus.
prostate-specific acid phosphatase are sensitive and specific
cytoplasmic markers for identifying more than 90% of LUNG NEUROENDOCRINE TUMORS
metastatic prostate adenocarcinomas, although both may A 72-year-old man with a 150–pack-year smoking history
demonstrate weaker expression in poorly differentiated presented with significant weight loss and CT chest imaging
tumors.100,112 Although NK3 homeobox 1 (NKX3.1), a highly that showed a right upper lobe lung mass and multiple
sensitive nuclear marker for primary prostatic adenocarci- enlarged mediastinal lymph nodes. Microscopic examina-
noma, was previously thought to be lost in a majority of tion of the endobronchial ultrasound biopsy revealed sheets
metastatic disease, newer antibodies have demonstrated and nests of loosely cohesive, small, round to fusiform
near-100% sensitivity and specificity for high-grade and hyperchromatic cells with finely granular chromatin, incon-
metastatic prostate adenocarcinomas.112 Lastly, v-ets avian spicuous nucleoli, and scant cytoplasm (Figure 10, A and B).
erythroblastosis virus E26 oncogene homolog (ERG) im- There was prominent crush artifact. In better preserved
munostain has been recently described as a surrogate areas, the mitotic count ranged from 6 to 8 per high-power
marker for the TMPRESS2-ERG fusion transcript that is field. The neoplastic cells were positive for CK7 (Figure 10,
reported in 40% to 70% of prostate cancer.113,114 Several C), synaptophysin (Figure 10, D), and TTF-1 (Figure 10, E);
studies have shown ERG antibodies to be highly accurate in they were negative for CK20 and chromogranin A. The Ki67
identifying prostate cancers with ERG overexpression, in labeling index was greater than 90% (Figure 10, F). The final
both primary and metastatic disease.113–115 diagnosis was SCLC.
Colorectal cancer metastasizes to the lungs in 10% to 20% With an incidence of more than 30 000 per year in the
of patients.116 CK7 and CK20 are frequently used as the United States, SCLC accounts for about 14% of all lung
first-line markers to differentiate lung (CK7 positive, CK20 cancers.122–125 Nearly all cases involve individuals with a
negative) from metastatic colorectal adenocarcinomas (CK7 history of smoking.126 Only a minority of cases present with
12 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al
Figure 10. Small cell lung carcinoma. The biopsy shows a sheet of small round hyperchromatic cells with peripheral crush artifact at low power (A).
At high power, the biopsy shows small round cells with scant cytoplasm, even and finely granular chromatin, inconspicuous nucleoli, and high
mitotic activity (B). The tumor cells are positive for cytokeratin 7 (C), synaptophysin (D), and thyroid transcription factor 1 (E). The Ki67 proliferative
index is greater than 90% of the tumor cells (F) (hematoxylin-eosin, original magnifications 340 [A] and 3400 [B]; original magnifications 3400 [D]
and 3100 [C, E, and F]).

Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 13


Table 5. Immunohistochemistry of Neuroendocrine Tumors of the Lung
Immunostain TC AC LCNEC SCLC
a
AE1/AE3 POS POS POS POS
CAM5.2 NEG/POS POS/NEG POS POS
CK7 NEG NEG POS NEG/POS
CK20 NEG NEG NEG NEG
CD56a POS POS POS/NEG POS
Synaptophysina POS POS POS POS/NEG
Chromogranin Aa POS POS POS/NEG NEG/POS
TTF-1a NEG NEG POS POS
Ki-67 (proliferation index)a 5% 20% 40%–80% 50%–100%
Abbreviations: AC, atypical carcinoid; CK, cytokeratin; LCNEC, large cell neuroendocrine carcinoma; NEG, negative; POS, positive; SCLC, small
cell lung carcinoma; TC, typical carcinoid; TTF-1, thyroid transcription factor 1.
a
Especially useful stains.

a small, localized lesion; most cases present with an neuroendocrine markers, a morphology highly characteristic
advanced stage and metastatic disease.126 Imaging studies of SCLC can support the diagnosis.128 Although TTF-1 can
can reveal a large mass demonstrating mediastinal invasion be positive in 70% to 90% of SCLCs,127,139–143 a study
or compression with regional lymphadenopathy; superior showed that 7 of 16 extrapulmonary small cell carcinomas
vena cava syndrome can occur in some instances.126 (44%) were also positive for TTF-1.144 Thus, TTF-1 has little
Small cell lung carcinoma can be easily recognized by its to no utility in determining the primary origin of the tumor.
distinct morphology.127,128 The neoplastic cells usually form A high Ki67 proliferation index can be very useful to
sheets and nests. Cytologically, they are small (less than 3 differentiate carcinoids from SLCL and LCNEC. Ki-67
times the diameter of a resting lymphocyte), hyperchro- proliferation index is usually up to 5% in typical carcinoid,
matic, and round to fusiform; they contain inconspicuous up to 20% in atypical carcinoid, 50% to 100% in SCLC, and
nucleoli, finely granular chromatin, and scant cyto- 40% to 80% in LCNEC.133 It should be noted, however, that
plasm.127,128 Mitotic figures may be difficult to identify on after chemotherapy, the high Ki-67 proliferation index of
small biopsies, but when present, they average about 80 SCLC can be reduced to levels more characteristic of
mitoses per 2 mm2.127,129,130 carcinoid tumors and thus be a source of confusion.128,145
Small cell lung carcinoma is one subclassification of The immunophenotypic profiles of SCLC and LCNEC are
neuroendocrine tumors of the lung, which also include highly similar. Thus, the distinction between the two
typical carcinoid, atypical carcinoid, and large cell neuroen- ultimately relies on morphologic examination. Small cell
docrine carcinoma (LCNEC). Typical carcinoid is character- lung carcinoma contains cells that are smaller (less than the
ized by a mitotic count of less than 2 per 2 mm2, atypical diameter of 3 small resting lymphocytes), have a higher
carcinoid is characterized by a mitotic count of 2 to 10 per 2 nuclear to cytoplasmic ratio, contain finely granular and
mm2, and high-grade neuroendocrine carcinomas (SCLC uniform chromatin, and have absent to inconspicuous
and LCNEC) are characterized by a mitotic count of more nucleoli, fusiform shape with scant cytoplasm, and crush
than 10 per 2 mm2. Necrosis is typically frequent, but it may artifact.128 In contrast, LCNEC is less uniform than SCLC
be missed because of limited sampling.128 Biopsy samples and contains polygonal-shaped cells with coarsely granular
may be subject to extensive crush artifact, which may limit to vesicular chromatin, prominent nucleoli, and abundant
the ability to render a definitive diagnosis. In these cases, pink cytoplasm. LCNEC is also less likely to demonstrate
cytology preparations may be better preserved and diag- nuclear molding.
nostic.128 In addition, in the larger resected specimens, Nonneuroendocrine mimickers of SCLC that are keratin
because of better fixation, the neoplastic cells tend to appear positive include Merkel cell carcinoma and various subtypes
bigger,131 which may pose confusion with LCNEC or other of SqCC (Table 6). Merkel cell carcinoma has a very similar
malignancies. immunophenotypic profile to that of SCLC; however,
In histologically challenging cases, especially in cases of
extensive crush artifact, a core panel of immunostains may
prove helpful to confirm the diagnosis: pan-cytokeratin
AE1/AE3 (AE1/AE3), CD56, chromogranin A, synaptophy- Table 6. Immunohistochemistry of Small Cell Lung
Carcinoma (SCLC) Versus Keratin-Positive Mimics
sin, TTF-1, and Ki-67 (Table 5).127,128,132,133
AE1/AE3 is useful to confirm an epithelial origin, because Immunostain SCLC MCC SqCC
a keratin-negative SCLC is extremely unusual.128 Although AE1/AE3 POS POS POS
CK7 is positive in about half of cases, CK20 is positive in less CD56 POS POS NEGa
than 10% of cases.134,135 Although CD56 is a highly sensitive Synaptophysin POS POS NEGa
marker (positive in 90%–100% of cases of SCLC),134,136,137 it Chromogranin A POS/NEG POS NEGa
TTF-1 POSa NEGa NEG
lacks specificity. Therefore, the diagnosis of SCLC in the CK20 NEGa POSa NEG
context of isolated CD56 positivity requires morphologic CK903 NEG ... POSa
correlation.128 Synaptophysin and chromogranin A are p63 NEG POS/NEG POSa
typically positive in SCLC, but in a study, no neuroendo- Abbreviations: CK, cytokeratin; MCC, Merkel cell carcinoma; NEG,
crine IHC activity was identified in 5 of 21 transbronchial negative; POS, positive; SqCC, squamous cell carcinoma; TTF-1,
biopsy specimens (24%) and 4 of 20 open lung biopsy thyroid transcription factor 1.
specimens (20%) of SCLC.138 In these cases negative for a
Especially useful results.
14 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al
Table 7. Immunohistochemistry of Small Cell Lung Carcinoma (SCLC) Versus Keratin-Negative Mimics
Immunostain SCLC Lymphoid Infiltrate/Lymphoma Ewing Sarcoma Melanoma
AE1/AE3 POS NEG NEG NEG
CD45 NEG POS NEG NEG
CD99 NEG ... POS ...
S100 NEG ... ... POS
Abbreviations: NEG, negative; POS, positive.

although Merkel cell carcinoma is typically positive for CK20 findings may include chronic inflammation, mast cells,
(perinuclear dotlike pattern) and negative for TTF-1, SCLC xanthomatous histiocytes, hemosiderin, calcifications, cho-
typically demonstrates the opposite profile (negative for lesterol clefts, and large lamellar structures.148 Cytologic
CK20 and positive for TTF-1).132,146 Although SqCC is atypia of the surface and round cells is usually not seen,
typically negative for neuroendocrine markers (CD56, although moderate to marked atypia can occur on rare
chromogranin A, and synaptophysin) and positive for occasions.150
CK903 and p63, SCLC also typically demonstrates the The largest IHC study of sclerosing pneumocytoma
opposite profile (positive for CD56, CG, and synaptophysin demonstrated that both surface and round cells stain for
and negative for CK903 and p63).132,146,147 TTF-1 and EMA.148 The surface cells are characteristically
Nonneuroendocrine mimickers of SCLC that are keratin positive for AE1/AE3 and also for CK7 and CAM 5.2. In
negative include lymphoid infiltrates/lymphoma, Ewing contrast, round cells are characteristically negative for AE1/
sarcoma, and melanoma (Table 7). CD45 can demonstrate AE3, with only a few cases showing scattered cells positive
hematolymphoid differentiation. CD99 and S100 can help for CK7 and CAM 5.2 (Table 8). Antibodies for surfactant
screen for Ewing sarcoma and melanoma, respectively. proteins A and B are also positive in surface cells and
negative in round cells.148
UNUSUAL LUNG NEOPLASMS The differential diagnosis of sclerosing pneumocytoma
A 33-year-old woman with a history of familial adeno- comprises both benign and malignant entities (Table 9).
matous polyposis and abdominal desmoid tumor presented Differentiating sclerosing pneumocytoma from malignant
with a heterogeneously enhancing 2.0 3 1.8-cm right upper lung tumors (eg, adenocarcinoma and carcinoid tumor) and
lobe nodule seen on chest CT. A CT-guided biopsy metastatic carcinomas (eg, metastatic papillary thyroid
demonstrated a low-grade epithelioid neoplasm (Figure carcinoma and metastatic renal cell carcinoma) is especially
11, A) positive for AE1/AE3 (focally positive in rare scattered critical.
cells) (Figure 11, B), Cam5.2 (Figure 11, C), and TTF-1 Distinguishing sclerosing pneumocytoma from lung
(Figure 11, D) and negative for CK7 (Figure 11, E). The adenocarcinoma may be particularly challenging in cases
neoplastic cells were also negative for synaptophysin, with subtle nuclear atypia. In this scenario, the presence of 2
chromogranin, HMB-45, MART1, S100, and PAX8. The distinct epithelial cell populations seen in the context of 1 of
histologic features and IHC staining results supported a final the 4 classic patterns of sclerosing pneumocytoma may be
diagnosis of sclerosing pneumocytoma. sufficient for a morphologic diagnosis. TTF-1 (staining both
Sclerosing pneumocytoma (formerly pulmonary scleros- surface and round cells) and AE1/AE3 (staining surface cells
ing hemangioma) is an uncommon lung tumor arising from only) can differentially highlight the 2 distinct epithelial cell
primitive lung epithelium.148 It is typically seen in middle-
populations. Sclerosing pneumocytoma is negative for the
aged adults, with a female predilection (5:1 female to male
neuroendocrine markers chromogranin and synaptophysin.
ratio).148 On imaging studies, sclerosing pneumocytoma
Because sclerosing pneumocytoma can assume a papillary
usually consists of a solitary (rarely multiple) nodule/mass
configuration, a metastatic papillary thyroid carcinoma or
that is well defined, oval, and homogeneous.149
renal cell carcinoma may enter the differential diagnosis.
By histology, sclerosing pneumocytoma consists of 2
Although cytologic features seen in papillary thyroid
major cell types: surface cells and round cells. Surface cells
resemble reactive type II pneumocytes and are cuboidal, carcinoma can be helpful, immunostains for thyroglobulin
whereas round cells consist of cells with well-defined and/or PAX8 can also be used to exclude the possibility of a
borders, fine chromatin, and inconspicuous nucleoli.150 metastatic papillary thyroid carcinoma. Similarly, the
Sclerosing pneumocytomas demonstrate 4 major patterns: marked cytologic atypia of a metastatic renal cell carcinoma
papillary, sclerotic, solid, and hemorrhagic, the former 3 may favor a malignant process. Immunohistochemical
patterns of which are the most common.148 Other histologic staining for CD10, carbonic anhydrase IX, and/or PAX8
(all typically positive in metastatic renal cell carcinoma) may
be helpful in these cases.
Table 8. Immunohistochemical Differences Benign tumors that enter the differential diagnosis of
Between Surface Cells and Round Cells sclerosing pneumocytoma include clear cell tumor, heman-
of Sclerosing Pneumocytoma gioma, and pulmonary hamartoma. Clear cell tumors
Surface Cells Round Cells typically demonstrate strong HMB-45 expression. Heman-
TTF-1 TTF-1 giomas typically stain positively for vascular markers such as
EMA EMA CD31, CD34, and ERG1. Pulmonary hamartoma, which
AE1/AE3 demonstrates varying degrees of mature cartilage, smooth
Abbreviations: EMA, epithelial membrane antigen; TTF-1, thyroid muscle, and/or adipose tissue, is usually diagnosed by
transcription factor 1. morphology alone. Immunohistochemical markers are
Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 15
Figure 11. Sclerosing pneumocytoma. The biopsy shows a low-grade epithelial neoplasm (A) positive for AE1/AE3 (focal) (B), Cam5.2 (C), and
thyroid transcription factor 1 (D) and negative for cytokeratin 7 (E) (hematoxylin-eosin, original magnification 3400 [A]; original magnification 3400
[B through E]).

16 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al


specific than thyroid transcription factor 1 in the differential diagnosis of primary
Table 9. Differential Diagnosis of Sclerosing pulmonary carcinoma: evaluation of 1674 cases by tissue microarray. Arch Pathol
Pneumocytoma Lab Med. 2012;136(2):163–171.
15. Bishop JA, Sharma R, Illei PB. Napsin A and thyroid transcription factor-1
Differential Diagnosis Immunostain expression in carcinomas of the lung, breast, pancreas, colon, kidney, thyroid,
Benign and malignant mesothelioma. Hum Pathol. 2010;41(1):20–25.
16. Fadare O, Desouki MM, Gwin K, et al. Frequent expression of napsin A in
Clear cell tumor HMB-45 clear cell carcinoma of the endometrium: potential diagnostic utility. Am J Surg
Hemangioma Vascular markers (CD31, Pathol. 2014;38(2):189–196.
CD34, ERG) 17. Kandalaft PL, Gown AM, Isacson C. The lung-restricted marker napsin A
Hamartoma S100 (mature is highly expressed in clear cell carcinomas of the ovary. Am J Clin Pathol. 2014;
chondrocytes) 142(6):830–836.
18. Mukhopadhyay S, Katzenstein AL. Subclassification of non-small cell lung
Malignant carcinomas lacking morphologic differentiation on biopsy specimens: utility of
Adenocarcinoma in situ TTF-1 an immunohistochemical panel containing TTF-1, napsin A, p63, and CK5/6. Am
Carcinoid tumor Synaptophysin, J Surg Pathol. 2011;35(1):15–25.
chromogranin A 19. Kargi A, Gurel D, Tuna B. The diagnostic value of TTF-1, CK5/6, and p63
Metastatic papillary Thyroglobulin, PAX8 immunostaining in classification of lung carcinomas. Appl Immunohistochem
Mol Morphol. 2007;15(4):415–420.
thyroid carcinoma
20. Rekhtman N, Ang DC, Sima CS, Travis WD, Moreira AL. Immunohisto-
Metastatic renal cell CD10, CA IX, PAX8 chemical algorithm for differentiation of lung adenocarcinoma and squamous
carcinoma cell carcinoma based on large series of whole-tissue sections with validation in
Abbreviations: CA IX, carbonic anhydrase IX; ERG, v-ets avian small specimens. Mod Pathol. 2011;24(10):1348–1359.
erythroblastosis virus E26 oncogene homolog; PAX8, paired box gene 21. Nicholson AG, Gonzalez D, Shah P, et al. Refining the diagnosis and
8; TTF-1, thyroid transcription factor 1. EGFR status of non-small cell lung carcinoma in biopsy and cytologic material,
using a panel of mucin staining, TTF-1, cytokeratin 5/6, and P63, and EGFR
mutation analysis. J Thorac Oncol. 2010;5:436–441.
22. Au NH, Gown AM, Cheang M, et al. P63 expression in lung carcinoma: a
generally not needed, but S100 may be used to highlight tissue microarray study of 408 cases. Appl Immunohistochem Mol Morphol.
2004;12(3):240–247.
mature chondrocytes. 23. Bishop JA, Benjamin H, Cholakh H, Chajut A, Clark DP, Westra WH.
Accurate classification of non-small cell lung carcinoma using a novel micro-
CONCLUSIONS RNA-based approach. Clin Cancer Res. 2010;16(2):610–619.
24. Pelosi G, Pasini F, Olsen Stenholm C, et al. P63 immunoreactivity in lung
In conclusion, the judicious use of IHC stains can be cancer: yet another player in the development of squamous cell carcinomas? J
helpful in the small-biopsy setting. In addition to helping Pathol. 2002;198(1):100–109.
25. Hedvat CV, Teruya-Feldstein J, Puig P, et al. Expression of p63 in diffuse
the pathologist further characterize the origin of the tumor large B-cell lymphoma. Appl Immunohistochem Mol Morphol. 2005;13(3):237–
cells in diagnostically challenging biopsies, IHC has also 242.
become increasingly important to identify diagnostic, 26. Crum CP, McKeon FD. P63 in epithelial survival, germ cell surveillance,
therapeutic, and prognostic biomarkers in cases of NSCLC. and neoplasia. Annu Rev Pathol. 2010;5:349–371.
27. Sniezek JC, Matheny KE, Westfall MD, Pietenpol JA. Dominant negative
References p63 isoform expression in head and neck squamous cell carcinoma. Laryngo-
1. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features scope. 2004;114(12):2063–2072.
associated with epidermal growth factor receptor gene mutations in lung cancers. 28. Massion PP, Taflan PM, Jamshedur Rahman SM, et al. Significance of p63
J Natl Cancer Inst. 2005;97(5):339–346. amplification and overexpression in lung cancer development and prognosis.
2. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase Cancer Res. 2003;63(21):7113–7121.
inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363(18):1693–1703. 29. Bishop JA, Teruya-Feldstein J, Westra WH, Pelosi G, Travis WD, Rekhtman
3. Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing N. p40 (DNp63) is superior to p63 for the diagnosis of pulmonary squamous cell
cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive carcinoma. Mod Pathol. 2012;25(3):405–415.
patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008; 30. Pelosi G, Sonzogni A, Papotti M, et al. Different prevalence of
26(21):3543–3551. transactivating (TA) p63 and nonTAp63 isoforms in pulmonary adenocarcinomas:
4. Johnson DF, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial a useful diagnostic tool. Mod Pathol. 2010;23(suppl 1): 411A–412A.
comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and 31. Pelosi G, Fabbri A, Rossi G, et al. A two-hit minimalist diagnostic
paclitaxel alone in previously untreated locally advanced or metastatic non- algorithm based on p40 (deltaNp63) and TTF-1 immunostaining upon small
small-cell lung cancer. J Clin Oncol. 2004;22(11):2184–2191. biopsy/cellblock samples for differentiating main subtypes of non-small cell lung
5. Hammerman PS, Sos ML, Ramos AH, et al. Mutations in the DDR2 kinase cancer and sparing material. J Thorac Oncol. 2011;6(6):S335–S336.
gene identify a novel therapeutic target in squamous cell lung cancer. Cancer 32. Righi L, Graziano P, Fornari A, et al. Immunohistochemical subtyping of
Discov. 2011;1(1):77–87. nonsmall cell lung cancer not otherwise specified in fine-needle aspiration
6. Weiss J, Sos ML, Seidel D, et al. Frequent and focal FGFR1 amplification cytology: a retrospective study of 103 cases with surgical correlation. Cancer.
associates with therapeutically tractable FGFR1 dependency in squamous cell 2011;117(15):3416–3423.
lung cancer. Sci Transl Med. 2010;2(62):62ra93. 33. Stoll LM, Johnson MW, Gabrielson E, Askin F, Clark DP, Li QK. The utility
7. Ikeda K, Clark JC, Shaw-White JR, et al. Gene structure and expression of of napsin-A in the identification of primary and metastatic lung adenocarcinoma
human thyroid transcription factor-1 in respiratory epithelial cells. J Biol Chem. among cytologically poorly differentiated carcinomas. Cancer Cytopathol.
1995;270(14):8108–8114. 201025;118(6):441–449.
8. Lau S, Luthringer D, Eisen R. Thyroid transcription factor-1: a review. Appl 34. Whithaus K, Fukuoka J, Prihoda TJ, Jagirdar J. Evaluation of napsin A,
Immunohistochem Mol Morphol. 2002;10(2):97–102. cytokeratin 5/6, p63, and thyroid transcription factor 1 in adenocarcinoma versus
9. Jagirdar J. Application of immunohistochemistry to the diagnosis of primary squamous cell carcinoma of the lung. Arch Pathol Lab Med. 2012;136(2):155–
and metastatic carcinoma to the lung. Arch Pathol Lab Med. 2008;132(3):384– 162.
396. 35. Yoshida A, Tsuta K, Watanabe S, et al. Frequent ALK rearrangement and
10. Chuman Y, Bergman A, Ueno T, et al. Napsin A, a member of the aspartic TTF-1/p63 co-expression in lung adenocarcinoma with signet-ring cell compo-
protease family, is abundantly expressed in normal lung and kidney tissue and is nent. Lung Cancer. 2011;72(3):309–315.
expressed in lung adenocarcinomas. FEBS Lett. 1999;462(1–2):129–134. 36. Zhang K, Hongbin D, Cagle PT. Utility of immunohistochemistry in the
11. Compérat E, Zhang F, Perrotin C, et al. Variable sensitivity and specificity diagnosis of pleuropulmonary and mediastinal cancers: a review and update.
of TTF-1 antibodies in lung metastatic adenocarcinoma of colorectal origin. Mod Arch Pathol Lab Med. 2014;138(12):1611–1628.
Pathol. 2005;18(10):1371–1376. 37. Travis W, Brambilla E, Noguchi M, et al. International Association for the
12. Ueno T, Linder S, Na C, Rice WR, Johansson J, Weaver TE. Processing of Study of Lung Cancer/American Thoracic Society/European Respiratory Society
pulmonary surfactant protein B by napsin and cathepsin H. J Biol Chem. 2004; international multidisciplinary classification of lung adenocarcinoma. J Thorac
279(16):16178–16184. Oncol. 2011;6(2):244–285.
13. DeFelice M, Siberschmidt D, DiLaura R, et al. TTF-1 phosphorylation is 38. Loo PS, Thomas SC, Nicolson MC, Fyfe MN, Kerr KM. Subtyping of
required for peripheral lung morphogenesis, perinatal survival, and tissue-specific undifferentiated non-small cell carcinomas in bronchial biopsy specimens. J
gene expression. J Biol Chem. 2003;278(37):35574–35583. Thorac Oncol. 2010;5(4):442–447.
14. Turner BM, Cagle PT, Sainz IM, Fukuoka J, Shen SS, Jagirdar J. Napsin A, a 39. Terry J, Leung S, Laskin J, Leslie KO, Gown AM, Ionescu DN. Optimal
new marker for lung adenocarcinoma, is complementary and more sensitive and immunohistochemical markers for distinguishing lung adenocarcinomas from

Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 17


squamous cell carcinomas in small tumor samples. Am J Surg Pathol. 2010; 68. Husain AN, Colby T, Ordonez N, et al. Guidelines for pathologic
34(12):1805–1811. diagnosis of malignant mesothelioma: 2012 update of the consensus statement
40. McGregor DH, Dixon AY, McGregor DK. Adenocarcinoma of the lung: a from the International Mesothelioma Interest Group. Arch Pathol Lab Med. 2013;
comparative diagnostic study using light and electron microscopy. Hum Pathol 137(5):647–667.
1988:19(8):910–913. 69. Ordonez NG. The diagnostic utility of immunohistochemistry in
41. Kosaka T, Yatabe Y, Endoh H, Kuwano H, Takahashi T, Mitsudomi T. distinguishing between epithelioid mesotheliomas and squamous carcinomas
Mutations of the epidermal growth factor receptor gene in lung cancer: biological of the lung: a comparative study. Mod Pathol. 2006;19(3):417–428.
and clinical implications. Cancer Res. 2004;64(24):8919–8923. 70. Weissinger SE, Keil P, Silvers DN, et al. A diagnostic algorithm to
42. Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth factor distinguish desmoplastic from spindle cell melanoma. Mod Pathol. 2014;27(4):
receptor mutations in non-small cell lung cancer. Clin Cancer Res. 2006;12(24): 524–534.
7232–7241. 71. Mayall FG, Goddard H, Gibbs AR. The diagnostic implications of variable
43. Yu J, Kane S, Wu J, et al. Mutation-specific antibodies for the detection of cytokeratin expression in mesotheliomas. J Pathol.1993;170(2):165–168.
EGFR mutations in non-small-cell lung cancer. Clin Cancer Res. 2009;15(9): 72. Chirieac LR, Pinkus GS, Pinkus JL, Godleski J, Sugarbaker DJ, Corson JM.
3023–3028. The immunohistochemical characterization of sarcomatoid malignant mesothe-
44. Xiong Y, Bai Y, Leong N, et al. Immunohistochemical detection of lioma of the pleura. Am J Cancer Res. 2011;1(1):14–24.
mutations in the epidermal growth factor receptor gene in lung adenocarcinomas 73. Chu AY, Litzky LA, Pasha TL, Acs G, Zhang PJ. Utility of D2-40, a novel
using mutation-specific antibodies. Diagn Pathol. 2013;8:27. mesothelial marker, in the diagnosis of malignant mesothelioma. Mod Pathol.
45. Seo AN, Park TI, Jin Y, et al. Novel EGFR mutation-specific antibodies for 2005;18(1):105–110.
lung adenocarcinoma: highly specific but not sensitive detection of an 74. Ordonez NG. D2-40 and podoplanin are highly specific and sensitive
E746_A750 deletion in exon 19 and an L858R mutation in exon 21 by immunohistochemical markers of epithelioid malignant mesothelioma. Hum
immunohistochemistry. Lung Cancer. 2014;83(3):316–323. Pathol. 2005;36(4):372–380.
46. Chen Z, Liu HB, Yu CH, Wang Y, Wang L, Song Y. Diagnostic value of 75. Dardick I, Al-Jabi M, McCaughey WT, et al. Ultrastructure of poorly
mutation-specific antibodies for immunohistochemical detection of epidermal differentiated diffuse epithelial mesotheliomas. Ultrastruct Pathol. 1984;7(2–3):
growth factor receptor mutations in non-small cell lung cancer: a meta-analysis. 151–160.
PLoS One. 2014;9(9):e105940. 76. Dardick I, Jabi M, McCaughey WT, et al. Diffuse epithelial mesothelioma:
47. Ragazzi M, Tamagnini I, Bisagni A, et al. Diamond: immunohistochem- a review of the ultrastructural spectrum. Ultrastruct Pathol. 1987;11(5–6):503–
istry versus sequencing in EGFR analysis of lung adenocarcinomas. J Clin Pathol. 533.
2016;69(5):440–447. 77. Churg A, Colby TV, Cagle P, et al. The separation of benign and malignant
48. Solomon B, Varella-Garcia M, Camidge DR. ALK gene rearrangements: a mesothelial proliferations. Am J Surg Pathol. 2000;24(9):1183–2000.
new therapeutic target in a molecularly defined subset of non-small cell lung 78. Cagle PT, Churg A. Differential diagnosis of benign and malignant
cancer. J Thorac Oncol. 2009;4(12):1450–1454. mesothelial proliferations on pleural biopsies. Arch Pathol Lab Med. 2005;
49. Pao W, Girard N. New driver mutations in non-small-cell lung cancer. 129(11):1421–1427.
Lancet Oncol. 2012;12(2):175–180. 79. Churg A, Cagle PT, Roggli VL. Tumors of the Serosal Membranes.
50. Selinger CI, Rogers TM, Russell PA, et al. Testing for ALK rearrangement in Washington, DC: American Registry of Pathology; 2006. Atlas of Tumor
lung adenocarcinoma: a multicenter comparison of immunohistochemistry and Pathology; 4th series, fascicle 3.
fluorescent in situ hybridization. Mod Pathol. 2013;26(12):1545–1553. 80. Churg A, Galateau-Salle F. The separation of benign and malignant
51. To KF, Tong JH, Yeung KS, et al. Detection of ALK rearrangement by mesothelial proliferations. Arch Pathol Lab Med. 2012;136(10):1217–1226.
immunohistochemistry in lung adenocarcinoma and the identification of a novel 81. Churg A, Cagle P, Colby TV, et al. The fake fat phenomenon in organizing
EML4-ALK variant. J Thorac Oncol. 2013;8(7):883–891. pleuritis: a source of confusion with desmoplastic malignant mesotheliomas. Am
52. Bergethon K, Shaw AT, Ou SH, et al. ROS1 rearrangements define a J Surg Pathol. 2011;35(12):1823–1829.
unique molecular class of lung cancers. J Clin Oncol. 2012;30(8):863–870. 82. Mayall FG, Goddard H, Gibbs AR. p53 immunostaining in the distinction
53. Davies KD, Le AT, Theodoro MF, et al. Identifying and targeting ROS1 between benign and malignant mesothelial proliferations using formalin-fixed
gene fusions in non-small cell lung cancer. Clin Cancer Res. 2012;18(17):4570– paraffin sections. J Pathol. 1992;168(4):377–381.
4579. 83. Cagle PT, Brown RW, Lebovitz RM. p53 immunostaining in the
54. Rimkunas VM, Crosby K, Kelly M, et al. Analysis of receptor tyrosine differentiation of reactive processes from malignancy in pleural biopsy
kinase ROS1 positive tumors in non-small cell lung cancer: identification of a specimens. Hum Pathol. 1994;25(5):443–448.
FIG-ROS1 fusion. Clin Cancer Res. 2012;18(16):4449–4457. 84. King J, Thatcher N, Pickering C, Hasleton P. Sensitivity and specificity of
55. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1-rearranged non- immunohistochemical antibodies used to distinguish between benign and
small-cell lung cancer. N Engl J Med. 2014;371(21):1963–1971. malignant pleural disease: a systematic review of published reports. Histopa-
56. Yoshida A, Tsuta K, Wakai S, et al. Immunohistochemical detection of thology. 2006;49(6):561–568.
ROS1 is useful for identifying ROS1 rearrangements in lung cancers. Mod Pathol. 85. Attanoos RL, Griffin A, Gibbs AR. The use of immunohistochemistry in
2014;27(5):711–720. distinguishing reactive from neoplastic mesothelium: a novel use for desmin and
57. Boyle TA, Masago K, Ellison KE, Yatabe Y, Hirsch FR. ROS1 immunohis- comparative evaluation with epithelial membrane antigen, p53, platelet-derived
tochemistry among major genotypes of non-small-cell lung cancer. Clin Lung growth factor-receptor, P-glycoprotein and bcl-2. Histopathology. 2003;43(3):
Cancer. 2015;16(2):106–111. 231–238.
58. Shan L, Lian F, Guo L, et al. Detection of ROS1 gene rearrangement in 86. Findeis-Hosey JJ, Xu H. The use of insulin like-growth factor II messenger
lung adenocarcinoma: comparison of IHC, FISH and real-time RT-PCR. PLoS RNA binding protein-3 in diagnostic pathology. Hum Pathol. 2010;42(3):303–
One. 2015;10(3):e0120422. 314.
59. Radziewicz H, Ibegbu CC, Fernandez ML, et al. Liver-infiltrating 87. Younes M, Lechago LV, Somoano JR, et al. Wide expression of the human
lymphocytes in chronic human hepatitis C virus infection display an exhausted erythrocyte glucose transporter Glut1 in human cancers. Cancer Res. 1996;56(5):
phenotype with high levels of PD-1 and low levels of CD127 expression. J Virol. 1164–1167.
2007;81(6):2545–2553. 88. Lee AF, Gown AM, Churg A. IMP3 and GLUT-1 immunohistochemistry
60. Gettinger S, Herbst RS. B7-H1/PD-1 blockade therapy in non-small cell for distinguishing benign from malignant mesothelial proliferations. Am J Surg
lung cancer: current status and future direction. Cancer J. 2014;20(4):281–289. Pathol. 2013;37(3):421–426.
61. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti–PD-L1 89. Shi M, Fraire AE, Chu P, et al. Oncofetal protein IMP3, a new diagnostic
antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455– biomarker to distinguish malignant mesothelioma from reactive mesothelial
2465. proliferation. Am J Surg Pathol. 2011;35(6):878–882.
62. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune 90. Ikeda K, Tate G, Suzuki T, Kitamura T, Mitsuya T. Diagnostic usefulness of
correlates of anti–PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443– EMA, IMP3, and GLUT-1 for the immunocytochemical distinction of malignant
2454. cells from reactive mesothelial cells in effusion cytology using cytospin
63. Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response preparations. Diagn Cytopathol. 2011;39(6):395–401.
to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014; 91. Minato H, Kurose N, Fukushima M, et al. Comparative immunohisto-
515(7528):563–567. chemical analysis of IMP3, GLUT1, EMA, CD146, and desmin for distinguishing
64. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of malignant mesothelioma from reactive mesothelial cells. Am J Clin Pathol. 2014;
non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–2028. 141(1):85–93.
65. Allen TC. Recognition of histopathologic patterns of diffuse malignant 92. Sheffield BS, Hwang HC, Lee AF, et al. BAP1 immunohistochemistry and
mesothelioma in differential diagnosis of pleural biopsies. Arch Pathol Lab Med. p16 FISH to separate benign from malignant mesothelial proliferations. Am J Surg
2005;129(11):1415–1420. Pathol. 2015;39(7):977–982.
66. Ordóñez NG. The immunohistochemical diagnosis of mesothelioma: a 93. Bott M, Brevet M, Taylor BS, et al. The nuclear deubiquitinase BAP1 is
comparative study of epithelioid mesothelioma and lung adenocarcinoma. Am J commonly inactivated by somatic mutations and 3p21.1 losses in malignant
Surg Pathol. 2003;27(8):1031–1051. pleural mesothelioma. Nat Genet. 2011;43(7):668–672.
67. Klebe S, Brownlee NA, Mahar A, et al. Sarcomatoid mesothelioma: a 94. Illei PB, Ladanyi M, Rusch VW, Zakowski MF. The use of CDKN2A
clinical-pathologic correlation of 326 cases. Mod Pathol. 2010;23(3):470– deletion as a diagnostic marker for malignant mesothelioma in body cavity
479. effusions. Cancer. 2003;99(1):51–56.

18 Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al


95. Chiosea S, Krasinskas A, Cagle PT, Mitchell KA, Zander DS, Dacic S. 124. Krug LM, Pietanza MC, Kris MG, et al. Small cell and other
Diagnostic importance of 9p21 homozygous deletion in malignant mesothelio- neuroendocrine tumors of the lung. In: DeVita VT, Lawrence TS, Rosenberg
mas. Mod Pathol. 2008;21(6):742–747. SA, eds. DeVita, Hellman and Rosenberg’s Cancer: Principles and Practice of
96. Monaco SE, Shuai Y, Bansal M, Krasinskas AM, Dacic S. The diagnostic Oncology. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2011:848–
utility of p16 FISH and GLUT-1 immunohistochemical analysis in mesothelial 870.
proliferations. Am J Clin Pathol. 2011;135(4):619–627. 125. Siegel R, Ward E, Brawley O, et al. Cancer statistics, 2011: the impact of
97. Raab SS, Allen TC, Leslie KO, Wick MR. Metastatic tumors in the lung. In: eliminating socioeconomic and racial disparities on premature cancer deaths. CA
Leslie KO, Wick MR, eds. Practical Pulmonary Pathology: A Diagnostic Cancer J Clin. 2011;61(4):212–236.
Approach. Philadelphia, PA: Elsevier; 2011:597–644. 126. Krug LM, Kris MG, Rosenzweig K, et al. Cancer of the lung: small cell and
98. Ye J, Findeis-Hosey JJ, Yang Q, et al. Combination of napsin A and TTF-1 other neuroendocrine tumors of the lung. In DeVita VT, Lawrence TS, Rosenberg
immunohistochemistry helps in differentiating primary lung adenocarcinoma SA, eds. DeVita, Hellman and Rosenberg’s Cancer: Principles and Practice of
from metastatic carcinoma in the lung. Appl Immunohistochem Mol Morphol. Oncology. 8th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2008:946–
2011;19(4):313–317. 971.
99. Ordóñez NG. Value of thyroid transcription factor-1 immunostaining in 127. Travis WD. Advances in neuroendocrine lung tumors. Ann Oncol. 2010;
tumor diagnosis. Appl Immunohistochem Mol Morphol. 2012;20(5):429–444. 21(suppl 7):vii65–vii71.
100. Lin F, Liu H. Immunohistochemistry in undifferentiated neoplasm/tumor 128. Travis WD. Update on small cell carcinoma and its differentiation from
of uncertain origin. Arch Pathol Lab Med. 2014;138(12):1583–1610. squamous cell carcinoma and other non-small cell carcinomas. Mod Pathol.
101. Ordóñez NG. Napsin A expression in lung and kidney neoplasia. Adv 2012;25(suppl 1):S18–S30.
Anat Pathol. 2012;19(1):66–73. 129. Travis WD, Brambilla E, Müller-Hermelink HK, et al. Pathology and
102. Iwamoto M, Nakatani Y, Fugo K, Kishimoto T, Kiyokawa T. Napsin A is Genetics: Tumors of the Lung, Pleura, Thymus and Heart. Vol 1. Lyon, France:
frequently expressed in clear cell carcinoma of the ovary and endometrium. Hum IARC; 2004.
Pathol. 2015;46(7):957–962. 130. Nicholson SA, Beasley MB, Brambilla E, et al. Small cell lung carcinoma
103. Yang M, Nonaka D. A study of immunohistochemical differential (SCLC): a clinicopathologic study of 100 cases with surgical specimens. Am J
expression in pulmonary and mammary carcinomas. Mod Pathol. 2010;23(5): Surg Pathol. 2002;26(9):1184–1197.
654–661. 131. Vollmer RT. The effect of cell size on the pathologic diagnosis of small
104. Liu H, Shi J, Wilkerson ML, Lin F. Immunohistochemical evaluation of and large cell carcinomas of the lung. Cancer. 1982;50(7):1380–1383.
GATA3 expression in tumors and normal tissues: a useful immunomarker for 132. Lin F, Prichard J. Handbook of Practical Immunohistochemistry:
breast and urothelial carcinomas. Am J Clin Pathol. 2012;138(1):57–64. Frequently Asked Questions. 2nd ed. New York, NY: Springer; 2015.
105. Miettinen M, McCue PA, Sarlomo-Rikala M, et al. GATA3: a multispecific 133. Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG. WHO
but potentially useful marker in surgical pathology: a systematic analysis of 2500 Classification of Tumors of the Lung, Pleura, Thymus and Heart. Lyon, France:
epithelial and nonepithelial tumors. Am J Surg Pathol. 2014;38(1):13–22. IARC; 2015.
106. Yoon NK, Maresh EL, Shen D, et al. Higher levels of GATA3 predict better 134. Bobos M, Hytiroglou P, Kostopoulos I, et al. Immunohistochemical
survival in women with breast cancer. Hum Pathol. 2010;41(12):1794–1801. distinction between merkel cell carcinoma and small cell carcinoma of the lung.
107. Yan Z, Gidley J, Horton D, Roberson J, Eltoum IE, Chhieng DC. Am J Dermatopathol. 2006;28:99–104.
135. Chu PG, Wu E, Weiss LM. Cytokeratin 7 and cytokeratin 20 expression in
Diagnostic utility of mammaglobin and GCDFP-15 in the identification of
epithelial neoplasms: a survey of 435 cases. Mod Pathol. 2000;13:962–972.
metastatic breast carcinoma in fluid specimens. Diagn Cytopathol. 2009;37(7):
136. Kontogianni K, Nicholson AG, Butcher D, Sheppard MN. CD56: a useful
475–478.
tool for the diagnosis of small cell lung carcinomas on biopsies with extensive
108. Braxton DR, Cohen C, Siddiqui MT. Utility of GATA3 immunohistochem-
crush artefact. J Clin Pathol. 2005;58(9):978–980.
istry for diagnosis of metastatic breast carcinoma in cytology specimens. Diagn
137. Hiroshima K, Iyoda A, Shida T, et al. Distinction of pulmonary large cell
Cytopathol. 2015;43(4):271–277.
neuroendocrine carcinoma from small cell lung carcinoma: a morphological,
109. Fatima N, Cohen C, Lawson D, Siddiqui MT. TTF-1 and napsin A double
immunohistochemical, and molecular analysis. Mod Pathol. 2006;19(10):1358–
stain. Cancer Cytopathol. 2011;119(2):127–133.
1368.
110. Bubendorf L, Schöpfer A, Wagner U, et al. Metastatic patterns of prostate
138. Guinee Jr DG, Fishback NF, Koss MN, Abbondanzo SL, Travis WD. The
cancer: an autopsy study of 1,589 patients. Hum Pathol. 2000;31(5):578–583.
spectrum of immunohistochemical staining of small cell lung carcinoma in
111. Wallis CJD, English JC, Goldenberg SL. The role of resection of pulmonary
specimens from transbronchial and open-lung biopsies. Am J Clin Pathol. 1994;
metastases from prostate cancer: a case report and literature review. Can Urol 102(4): 406–414.
Assoc J. 2011;5(6):E104–E108. 139. Folpe AL, Gown AM, Lamps LW, et al. Thyroid transcription factor-1:
112. Gurel B, Ali TZ, Montgomery EA, et al. NKX3.1 as a marker of prostatic immunohistochemical evaluation in pulmonary neuroendocrine tumors. Mod
origin in metastatic tumors. Am J Surg Pathol. 2010;34(8):1097–1105. Pathol. 1999;12(1):5–8.
113. Chaux A, Albadine R, Toubaji A, et al. Immunohistochemistry for ERG 140. Sturm N, Rossi G, Lantuéjoul S, et al. 34BetaE12 expression along the
expression as a surrogate for TMPRSS2-ERG fusion detection in prostatic whole spectrum of neuroendocrine proliferations of the lung, from neuroendo-
adenocarcinomas. Am J Surg Pathol. 2011;35(7):1014–1020. crine cell hyperplasia to small cell carcinoma. Histopathology. 2003;42(2):156–
114. van Leenders GJLH, Boormans JL, Vissers CJ, et al. Antibody EPR3864 is 166.
specific for ERG genomic fusions in prostate cancer: implications for pathological 141. Sturm N, Rossi G, Lantuejoul S, et al. Expression of thyroid transcription
practice. Mod Pathol. 2011;24(8):1128–1138. factor-1 in the spectrum of neuroendocrine cell lung proliferations with special
115. Braun M, Goltz D, Shaikhibrahim Z, et al. ERG protein expression and interest in carcinoids. Hum Pathol. 2002;33(2):175–182.
genomic rearrangement status in primary and metastatic prostate cancer—a 142. Travis WD. Neuroendocrine lung tumors. Pathol Case Rev. 2006;11:235–
comparative study of two monoclonal antibodies. Prostate Cancer Prostatic Dis. 242.
2012;15(2):165–169. 143. Travis WD. Lung tumours with neuroendocrine differentiation. Eur J
116. Zisis C, Tsakiridis K, Kougioumtzi I, et al. The management of the Cancer. 2009;45(suppl 1):251–266.
advanced colorectal cancer: management of the pulmonary metastases. J Thorac 144. Agoff SN, Lamps LW, Philip AT, et al. Thyroid transcription factor-1 is
Dis. 2013;5(suppl 4):S383–S388. expressed in extrapulmonary small cell carcinomas but not in other extrapulmo-
117. Barbareschi M, Murer B, Colby TV, et al. CDX-2 homeobox gene nary neuroendocrine tumors. Mod Pathol. 2000;13(3):238–242.
expression is a reliable marker of colorectal adenocarcinoma metastases to the 145. Brambilla E, Moro D, Gazzeri S, et al. Cytotoxic chemotherapy induces
lungs. Am J Surg Pathol. 2003;27(2):141–149. cell differentiation in small cell lung carcinoma. J Clin Oncol. 1991;9(1):50–61.
118. Inamura K, Satoh Y, Okumura S, et al. Pulmonary adenocarcinomas with 146. Asioli S, Righi A, Volante M, Eusebi V, Bussolati G. p63 expression as a
enteric differentiation. Am J Surg Pathol. 2005;29(5):660–665. new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110(3):640–647.
119. Dettmer M, Kim TE, Jung CK, Jung ES, Lee KY, Kang CS. Thyroid 147. Sturm N, Lantuejoul S, Laverriere MH, et al. Thyroid transcription factor 1
transcription factor-1 expression in colorectal adenocarcinomas. Pathol Res and cytokeratins 1, 5, 10, 14 (34betaE12) expression in basaloid and large-cell
Pract. 2011;207(11):686–690. neuroendocrine carcinomas of the lung. Hum Pathol. 2001;32(9):918–925.
120. Ye J, Hameed O, Findeis-Hosey JJ, et al. Diagnostic utility of PAX8, TTF-1 148. Devouassoux-Shisheboran M, Hayashi T, Linnoila RI, Koss MN, Travis
and napsin A for discriminating metastatic carcinoma from primary adenocar- WD. A clinicopathologic study of 100 cases of pulmonary sclerosing
cinoma of the lung. Biotech Histochem. 2012;87(1):30–34. hemangioma with immunohistochemical studies: TTF-1 is expressed in both
121. Ordonez NG. Value of PAX 8 immunostaining in tumor diagnosis: a round and surface cells, suggesting an origin from primitive respiratory
review and update. Adv Anat Pathol. 2012;19(3):140–151. epithelium. Am J Surg Pathol. 2000;24(7):906–916.
122. Altekruse SF, Kosary CL, Krapcho M, et al. SEER cancer statistics review, 149. Shin SY, Kim MY, Oh SY, et al. Pulmonary sclerosing pneumocytoma of
1975–2007, National Cancer Institute. http://seer.cancer.gov/csr/1975_2007/. the lung: CT characteristics in a large series of a tertiary referral center. Medicine
Accessed January 16, 2017. (Baltimore). 2015;94(4):e498.
123. Travis WD, Travis LB, Devesa SS. Lung cancer [published erratum appears 150. Keylock JB, Galvin JR, Franks TJ. Sclerosing hemangioma of the lung.
in Cancer. 1995;75(12):2979]. Cancer. 1995;75:191–202. Arch Pathol Lab Med. 2009;133(5):820–825.

Arch Pathol Lab Med Immunohistochemistry in Lung and Pleural Neoplasms—Woo et al 19

Você também pode gostar