4 Pleural, Pericardial, and Peritoneal Fluid

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

4

Pleural, Pericardial, and


Peritoneal Fluids
Edmund S. Cibas

SPECIMEN COLLECTION, PREPARATION, Diffuse Malignant Mesothelioma


AND REPORTING TERMINOLOGY Primary Effusion Lymphoma
Metastatic Tumors
ACCURACY
Adenocarcinoma
BENIGN ELEMENTS Squamous Cell Carcinoma
Small Cell Carcinoma
NON-NEOPLASTIC CONDITIONS Melanoma
Acute Serositis Non-Hodgkin Lymphoma
Eosinophilic Effusions Hodgkin Lymphoma
Lymphocytic Effusions Multiple Myeloma
Rheumatoid Pleuritis Acute and Chronic Leukemias
Lupus Pleuritis Sarcomas
Other Non-Neoplastic Conditions Germ Cell Tumors
MALIGNANT EFFUSIONS
Primary Tumors

The pleural, pericardial, and peritoneal cavities are lined


by a single layer of flat mesothelial cells called the serosa. Specimen Collection,
Normally, these cavities are collapsed and contain only Preparation, and Reporting
a small amount of fluid, enough to lubricate the adja- Terminology
cent surfaces as they move over each other with respi-
ration, heartbeats, and intestinal peristalsis. In disease Specimens are obtained by inserting a needle into the
states, a greater amount of fluid—an effusion—accu- pleural space (thoracentesis), pericardial space (peri-
mulates. Effusions are classified clinically as transuda- cardiocentesis), and peritoneal cavity (paracentesis). No
tive or exudative. Transudates result from an imbalance more than 1500 mL of pleural fluid should be removed
of hydrostatic and oncotic pressures. Common causes at any given time because larger evacuations can cause
are congestive heart failure (CHF), cirrhosis, and the postexpansion pulmonary edema.1 By contrast, large–
nephrotic syndrome. Transudates have a low lactate volume paracentesis (e.g., 4 to 6 L) is relatively safe,
dehydrogenase (LDH) and low total protein concentra- and there are even reports of draining 20 L of ascites at
tion. Exudates result from injury to the mesothelium, as one time.2 Although peritoneal fluid is usually obtained
occurs with malignancy, pneumonia, lupus, rheumatoid through the abdominal wall, in women it can also be
pleuritis, pulmonary infarction, or trauma. Exudates aspirated from the cul-de-sac through the vagina (cul-
have a relatively high LDH and total protein concentra- docentesis). Less commonly, fluid is obtained by suction
tion. The distinction is made by protein concentration during thoracic, abdominal, or cardiac surgery.
measurements performed in the clinical laboratory. The Fluid is collected in clean containers and sent unfixed
distinction is important because pleural involvement by to the laboratory. To prevent clotting, which widely dis-
a malignancy causes an exudate, and therefore cytologic perses cells, thus hindering their evaluation, fluid can
examination is not needed for a transudate.1 Malignant be collected in heparinized bottles containing 3 units of
tumors are a common cause of exudates because the heparin per milliliter of capacity.3 If heparinized bottles
serosal surfaces are a frequent site of metastasis for are not available, the heparin can be poured into the bot-
many tumors and the site of origin for the asbestos- tle before the fluid is collected; contact with glass results
related tumor malignant mesothelioma. in rapid clotting.

129
130 Pleural, Pericardial, and Peritoneal Fluids

Fluid is refrigerated at 4°C until the time of slide Most laboratories report results using descriptive
­ reparation. An effusion specimen is remarkably
p terminology accompanied by simple primary headings
hardy—it can be refrigerated for 2 weeks or longer such as “no malignant cells identified,” “suspicious cells
without compromising cellular morphology or anti- present,” and “positive for malignant cells.” A case is
genicity for immunostains.4 A variety of slide prepa- called suspicious when the abnormal cells are too poorly
ration methods are available. Slide preparation often preserved or too few to support a definite diagnosis of
begins by shaking the container to disperse the cells, malignancy; this occurs in about 5% of specimens.19,20
then centrifuging a 50 mL aliquot (or the entire speci- In this situation, if the patient does have a malignancy
men if less than 50mL). The supernatant is discarded, involving the serosal cavity, fluid is likely to reaccumu-
and the sediment used to prepare direct smears, cyto- late and the subsequent specimens may be diagnostic
centrifuge preparations,5 thinlayer slides5,6 or filter of malignancy. Criteria for the adequacy of an effusion
preparations. The slides are usually alcohol fixed and specimen have not been established.
Papanicolaou stained. If a hematologic malignancy is
suspected, air-dried cytocentrifuge preparations are
helpful. One is stained with a Romanowsky-type stain Accuracy
and the rest can be reserved, if needed, for immuno-
cytochemical studies for lymphocyte surface mark- Cytology is more sensitive than blind biopsy for detect-
ers.7 So-called “cell blocks” are especially useful as ing serosal malignancy (71% versus 45%),21 presumably
adjuncts to the “cytologic” preparations previously because fluid provides a more representative sample.
listed. To prepare a cell block, the remainder of the Estimates of the sensitivity of cytology for diagnosing
sediment is wrapped in filter paper, placed in a cas- serosal malignancy range from 58% to 71%.20,21 The can-
sette, embedded in paraffin, and cut and stained in cer detection rate by cytology is increased by 2% to 38%
the manner of histologic sections. Before placing it in when multiple specimens are examined.19,22,23 This still
a cassette, however, it is helpful to coagulate the sed- leaves a substantial false-negative rate. Thoracoscopy is
iment by adding a few drops of plasma and several the procedure of choice for patients with a strong clini-
drops of a thrombin solution.8-10 Clotting the speci- cal suspicion of pleural disease but a negative cytology
men in this fashion does not disperse the diagnostic result.24
cells as does a spontaneously formed clot, but rather The specificity of a cytologic diagnosis is quite high;
congeals the sediment into a compact mass. If the false-positive diagnoses occur in less than 1% of cases.19,25
fluid was not heparinized and clots are present, they When they occur, false-positive and false-suspicious
should be removed and placed in cassettes for pro- diagnoses are caused by mesothelial cell atypia in the
cessing as cell block material. setting of pulmonary infarction,21 tuberculosis,19 chemo-
To improve sensitivity, most laboratories use two or therapy,26 acute pancreatitis,25,26 ovarian fibroma,19 and
more preparation methods for effusions. Smears alone cirrhosis.25 In children, false-positives result from mis-
identify about 67% of malignant fluids, whereas cyto- interpreting benign lymphoid cells as lymphoma or
centrifuge, filter, and cell block preparations detect 83% neuroblastoma.27
to 85%.11 Cell block sections are especially useful for spe- Immunocytochemistry is an essential adjunct to cyto-
cial stains and immunohistochemistry because of the morphology in selected cases and substantially improves
ease with which multiple duplicate slides can be pre- diagnostic accuracy.
pared, the relative absence of obscuring background
staining, and the standardization of the preparation When to use immunohistochemistry for
for control slides.12 Cell blocks also make for excellent effusions:
morphologic comparison with histopathologic sections
• confirming malignancy when morphology alone
(when, for example, the patient has had a prior breast
is equivocal
biopsy) because they are fixed and stained in an identi-
• screening an effusion for lobular breast cancer
cal manner.
• distinguishing adenocarcinoma from
In some laboratories, an unfixed wet smear stained
mesothelioma
with toluidine blue is prepared first to identify any fluid
• establishing the primary site of a malignant
that contains large numbers of malignant cells. It is help-
effusion; a patient with:
ful to separate such fluids from the routine staining cycle
• an occult primary
to prevent cross-contamination.10
• multiple primaries
Leftover fluid is stored in the refrigerator in case addi-
tional slides are needed. Fresh fluid is sometimes useful
for other studies, including flow cytometry,13-15 electron The circumstances outlined in the box represent
microscopy,16 and cytogenetic or molecular genetic common applications for immunohistochemistry.
analysis.17,18 Antibodies against carcinoembryonic antigen (CEA),
Benign Elements 131

B72.3, and a number of other markers have high sensi- copy. Two or more mesothelial cells in groups are often
tivity and specificity for malignancy and are extremely separated by a narrow space or “window.” Less com-
useful in a variety of settings, particularly for resolving monly, mesothelial cells have one or more cytoplasmic
cases that are cytologically equivocal.28 Detailed dis- vacuoles.
cussion of specific applications is found in the sections With acute or chronic injury, mesothelial cells
that follow. undergo hyperplasia and hypertrophy and can have sig-
nificant nuclear atypia, but they remain predominantly
dispersed as isolated cells. Such “reactive” mesothelial
Benign Elements cells generally comprise a spectrum of cells that range
from normal to “atypical,” with variation in nuclear size,
Benign effusions contain mesothelial cells, histiocytes, a coarse chromatin texture, irregular nuclear contours,
and lymphocytes in varying proportions. Because some or prominent nucleoli (Fig. 4.2).
bleeding is common during specimen collection, red
and white blood cells are common contaminants. Differential diagnosis of reactive
mesothelial cells:
Cytomorphology of mesothelial cells:
• mesothelioma
• often numerous • metastatic malignancy
• dispersed as isolated cells
• occasional small clusters with “windows”
• round cells
Malignant mesothelioma should be considered
• round nucleus
if there is marked atypia, particularly if the cells are
• single nucleolus
much larger than normal, or if the fluid contains
• dense cytoplasm with clear outer rim (“lacy skirt”)
numerous clusters with 12 or more mesothelial cells,
even if the cells themselves are not particularly atypi-
cal. Such large groups are uncommon in benign effu-
Mesothelial cells can be sparse or numerous in sions. Clinical correlation is important because it
benign effusions (Fig. 4.1). They are mainly dispersed may account for the atypia; some medical conditions,
as isolated cells or occasional small clusters. Large clus- including anemia, cirrhosis, lupus, pulmonary infarc-
ters composed of more than 12 cells are highly unusual tion, renal failure, and AIDS,29 are notorious causes of
in benign effusions. Binucleation and multinucleation mesothelial atypia. On the other hand, if the patient
are common, and mesothelial cells in mitosis can be has a large, unexplained, unilateral effusion, particu-
seen in benign effusions. The dense cytoplasm reflects larly with radiographic evidence of pleural thickening,
the abundance of tonofilaments, and the clear outer additional evaluation (pleural biopsy, cytogenetics or
rim (“lacy skirt” or “halo”) corresponds to long, slen- molecular genetics) should be considered to exclude
der microvilli, better visualized with electron micros- mesothelioma.17,18

A B
Figure 4.1  Mesothelial cells. A, Many characteristic features of mesothelial cells are seen here: the peripheral lucent zone, or “lacy
skirt” (arrow); the dense perinuclear zone; the occasional binucleation; and the slitlike separation (“window”) between adjacent cells.
A few histiocytes (arrowheads), with folded nuclei and vacuolated cytoplasm, are also present. B, Peripheral halos are well seen in this
cell block section. Note also the large multinucleated mesothelial cell, a nonspecific finding.
132 Pleural, Pericardial, and Peritoneal Fluids

Some effusions contain abundant histiocytes


(Fig. 4.3A). A particularly marked histiocytic reaction to
irritation of the serosal surfaces has been termed nodular
histiocytic hyperplasia.30,31 This is a nonspecific chronic
inflammatory reaction that should not be misconstrued
as a malignancy in cytologic or histologic specimens.10,31
When abundant, histiocytes can form aggregates on
smears and liquid-based preparations,31 and they
tend to sediment together in cell block preparations,
­forming mass-like aggregates that mimic malignancy.
Immunohistochemistry can be useful to distinguish
histiocytes from mesothelial cells and metastatic
­carcinoma; histiocytes are immunoreactive for CD68
and negative for keratin proteins (Fig. 4.3B); the reverse
is true for mesothelial cells and metastatic carcinoma.

Figure 4.2  Reactive mesothelial atypia (peritoneal fluid, Non-Neoplastic Conditions


cirrhosis). Some benign fluids contain a population of ­moderately
enlarged mesothelial cells with large, hyperchromatic, irregular In many benign disorders, effusions give a nonspe-
nuclei. cific cytologic picture. Thus, pleural fluid in con-
gestive heart failure or pulmonary infarction is
Metastatic malignancy should be considered when a morphologically indistinguishable from pericardial
second population of cells is identified that is morpho- fluid caused by renal failure and peritoneal fluid as a
logically distinct from the mesothelial cells. In a minor- result of cirrhosis. Fortunately, the features of some
ity of malignant effusions, a second population of cells benign conditions are sufficiently characteristic to
is not evident. This is particularly true with lobular car- narrow the differential diagnoses or even indicate
cinoma of the breast and melanoma, the cells of which the specific etiology. To give an unusual example,
mimic normal histiocytes or mesothelial cells. Special finding undigested meat and vegetable matter in
stains are then needed to resolve the case. pleural fluid strongly suggests esophageal rupture
(Boerhaave’s syndrome). 32
Cytomorphology of histiocytes:
• smaller nucleus than that of mesothelial cells Acute Serositis
• nucleus often folded
• cytoplasm granular or vacuolated Acute pleuritis, pericarditis, and peritonitis are usually
• no “windows” between adjacent cells the result of a bacterial infection. Bacterial infection of
• dense aggregates (cell block sections) the pleura occurs in the setting of pneumonia, which
secondarily involves the overlying pleura and results in a

A B
Figure 4.3  Histiocytes (pleural fluid, congestive heart ­failure). A, Like mesothelial cells, histiocytes are usually dispersed as isolated
cells, but centrifugation for cell block sections compresses them into large groups. Compare the histiocytes, which have an oval or folded
nucleus, to the mesothelial cell (arrow). B, Only the mesothelial cell is immunoreactive for cytokeratins.
Non-Neoplastic Conditions 133

pleural empyema. Acute infection of the peritoneal cav- stained slides. Charcot-Leyden crystals (see Fig. 2.10)
ity is often secondary to inflammation of or injury to the are present in some cases, and curiously, are more com-
bowel, as in spontaneous bacterial peritonitis. mon in fluids that have been refrigerated for more than
The fluid is a creamy pale yellow (purulent) and often 24 hours.36
foul-smelling. Cytologic preparations are highly cellu-
lar and composed almost exclusively of polymorphonu-
clear leukocytes. Bacteria are demonstrated with special
Lymphocytic Effusions
stains in some cases. A pleural effusion consisting mostly of small lympho-
It is important to screen such cases carefully for malig- cytes is a relatively common but nonspecific find-
nant cells because acute infection can be a complication ing. Cytologic preparations are often highly cellular
of metastatic malignancy. and composed almost exclusively of dispersed, small
lymphocytes.37 Mesothelial cells and histiocytes are either
conspicuously absent or present in small numbers.
Eosinophilic Effusions
A pleural effusion is considered “eosinophilic” when Differential diagnosis of
eosinophils account for 10% or more of the nucleated lymphocytic effusions:
cells present. Between 5% and 16% of exudative effusions
• malignancy
are eosinophilic effusions.33 The most common causes
• tuberculosis
are pneumothorax and hemothorax.24 The introduction
• status post coronary artery bypass
of air or blood into the pleural space, so often the reason
behind an eosinophilic effusion, can occur simply with
repeated thoracenteses. Less common causes include
drug reactions, parasitic infections, pulmonary infarc- Despite the absence of malignant cells, a malig-
tion, and the Churg-Strauss syndrome.24,34 In about one nancy is a common cause of a lymphocytic effusion.
third of cases the origin remains obscure.35 Most cases The malignancy may be nearby (e.g., in the lung) and
resolve spontaneously. may be obstructing lymphatic outflow but may not
Eosinophilic pericardial and peritoneal effusions are have spread to the pleural surfaces. Alternatively, a
less common than eosinophilic pleural effusions. pleural malignancy may be evoking a peritumoral lym-
Cytologic preparations are usually cellular and phocytic response, but the tumor itself is not shedding
remarkable for a high concentration of eosinophils. On cells into the effusion.30 It is not uncommon for the ini-
alcohol-fixed Papanicolaou-stained slides, the defining tial pleural fluids in patients with a pleural mesotheli-
eosinophilic cytoplasmic granules are either orangeo- oma to consist only of lymphocytes.38 Effusions caused
philic or pale-green and inconspicuous, and the cells by small lymphocytic lymphoma and chronic lympho-
are identified more on the basis of their bilobed nuclei cytic leukemia are quite uncommon. Because these
(Fig. 4.4). The granules are brightly eosinophilic on are B-cell neoplasms, immunocytochemical or flow
cell block preparations stained with hematoxylin and cytometric evaluation of lymphocyte surface mark-
eosin (H & E), however, and on air-dried Romanowsky- ers is helpful in confirming the diagnosis. In a patient
with chronic lymphocytic leukemia and a peripheral
lymphocytosis, however, contamination of the effu-
sion by peripheral blood during a traumatic tap should
be excluded before diagnosing pleural involvement.
Even a small amount of blood containing leukemic
cells can result in a false-positive diagnosis. The diag-
nosis of tuberculosis can be confirmed by microbio-
logic studies or pleural biopsy, which reveals caseating
granulomas and acid-fast organisms. The differential
diagnosis includes other benign effusions of nontu-
berculous origin, as in patients after coronary artery
bypass surgery.24

Rheumatoid Pleuritis
Less than 5% of patients with rheumatoid arthritis
Figure 4.4  Eosinophilic pleural effusion. Numerous eosin- develop pleural involvement by the same necrotiz-
ophils in pleural fluid are more commonly associated with
benign conditions, like a pneumothorax (as in this case) or ing granulomatous inflammation that causes joint
hemothorax. disease. In almost all cases, joint disease precedes
134 Pleural, Pericardial, and Peritoneal Fluids

the development of pleuritis, but occasionally pleuri-


tis precedes or is synchronous with the onset of joint
Lupus Pleuritis
disease.39,40 The pleural effusion can be unilateral or About one third of patients with systemic lupus ery-
bilateral, and some patients have a synchronous peri- thematosus (SLE) develop a pleural or pericardial effu-
cardial effusion. Radiographic studies reveal pulmo- sion. Peritoneal effusions are less common but do occur.
nary nodules in a minority of patients; presumably, Rarely, an effusion is the initial manifestation.
these are rheumatoid nodules. The effusion can last The characteristic cell is the lupus erythematosus
for days, months, or sometimes years. (LE) cell, a neutrophil or macrophage that contains an
The cytologic picture is so characteristic that it has ingested cytoplasmic particle called a hematoxylin body.
been termed pathognomonic.39 Examination of pleural The hematoxylin body may be green, blue, or purple with
fluid, therefore, can be extremely useful to confirm the the Papanicolaou stain, and magenta with Romanowsky-
diagnosis of rheumatoid pleuritis and exclude the possi- type stains, and has a glassy, homogeneous appearance
bility of coincident disease, especially a malignancy. (Fig. 4.6). Filling the cytoplasm of the neutrophil or mac-
rophage, it often pushes the nucleus to one side, indent-
Cytomorphology of rheumatoid ing it into a crescent-like shape. Hematoxylin bodies are
pleuritis: thought to represent degenerated nuclei. Similar cells
that contain ingested nuclei with a visible chromatin
• abundant clumps of granular debris
structure (rather than the glassy, structureless hematox-
• macrophages
ylin body) are called tart cells after the patient in whom
they were first described.
Lupus erythematosus cells are present in just 27%
Cytologic preparations are sparsely or moder- of effusions in patients with systemic lupus erythe-
ately cellular. An abundant granular material domi- matosus, and only in those with a known diagnosis of
nates the picture (Fig. 4.5A). It can stain green, pink, systemic lupus erythematosus.41
red, or orange with the Papanicolaou stain, and aggre-
gates into small and large clumps with irregular edges.
Large, island-like masses can be appreciated in cell
Other Non-Neoplastic Conditions
block material. The predominant cell is the macro- Most viral pneumonias associated with a pleural
phage, which is round or spindle-shaped; multinucle- ­ ffusion result in a nonspecific cytologic picture. The
e
ated macrophages are seen in most but not all cases cytopathic changes characteristic of the herpes viruses
(Fig. 4.5A and B). Lymphocytes and polymorphonu- and cytomegalovirus (CMV) are rarely seen in serous
clear leukocytes may be seen. Mesothelial cells are effusions. Although fungal infections are common in
noticeably absent. patients who are immunocompromised, organisms
The characteristic granular debris is different from are rarely seen in pleural, pericardial, and peritoneal
fibrin, which is usually strandlike rather than coarsely fluids. Candida species, Cryptococcus neoformans,
granular. Although the elongated macrophages resem- Coccidioides immitis, Blastomyces dermatitidis, and
ble the spindle cells seen in squamous and other can- Aspergillus niger have been described in fluids in rare
cers, their nuclei are normochromatic. instances.10

A B
Figure 4.5  Rheumatoid pleuritis. A, Scattered multinucleated histiocytes and clumped granular debris in the background are
characteristic of pleural fluids in patients with rheumatoid pleuritis. B, A pleural biopsy has the appearance of an “opened out”
­rheumatoid nodule, with epithelioid histiocytes, giant cells, and fibrinoid debris lining the pleural space.
Malignant Effusions 135

Table 4.1  The most Common Tumors that


cause Malignant Effusions by Site and Sex*
Site Men Women
Pleural lung breast
lymphoma or leukemia lung
gastrointestinal tract lymphoma or leukemia
sarcoma ovary
mesothelioma gastrointestinal tract
genitourinary (kidney, endometrium
prostate, bladder) sarcoma
melanoma mesothelioma
Peritoneal lymphoma or leukemia ovary
gastrointestinal tract breast
pancreas endometrium
Figure 4.6  Hematoxylin body (lupus pleuritis). The lobes of lung stomach
the nucleus are pushed against the side of the neutrophil by a sarcoma lymphoma or leukemia
large, homogeneous, intracytoplasmic body (Wright-Giemsa).
prostate colon and rectum
melanoma pancreas
germ cell tumors mesothelioma
Pneumocystis carinii has been identified in pleural
mesothelioma
and peritoneal effusions from patients who are immu-
nocompromised.42-44 Papanicolaou stains show foamy
* Data from Sears D, Hajdu SI: The cytologic diagnosis of malignant
exudates similar to those seen in respiratory specimens.
neoplasms in pleural and peritoneal effusions. Acta Cytol 1987;31:85-97;
The trophozoites measure 2.5 to 5.0 μm and have pale Johnston WW: The malignant pleural effusion: A review of cytopathologic
cytoplasm and a dot-like nucleus; they may be intra- diagnoses of 584 specimens from 472 consecutive patients. Cancer
1985;56:905-909.
cellular (within macrophages) or extracellular, and
are well seen on air-dried preparations stained with a
Romanowsky-type stain. Cyst forms measure 4 to 7 μm
and can be seen with special stains like the methena-
mine silver stain. is less than 6 months.48 Certain tumors, like estrogen-
­positive breast cancers and well-differentiated mucinous
adenocarcinomas of the appendix, have a slightly better
Malignant Effusions prognosis.
Systemic chemotherapy fails to alleviate most recur-
Some tumors have a greater tendency than others to rent malignant effusions, with a few notable exceptions
spread to the pleura, pericardium, or peritoneum. (e.g., those caused by lymphoma and small cell lung can-
The most common are listed in Table 4.1. In children, cer). Because most malignant pleural effusions recur and
the most common cause of a malignant pleural or impede respiration, chest tube placement or pleurodesis
peritoneal effusion is non-Hodgkin lymphoma. 27 (sclerosis of the pleural cavity by injecting talc, doxycy-
Most patients with a malignant effusion have a pre- cline, or bleomycin) is often performed as a palliative
viously documented primary neoplasm. In some cases, measure.2 For patients with recurrent malignant ascites,
however, a malignant effusion is the first manifestation palliative treatment may consist of either repeated para-
of an occult malignancy. The most common occult pri- centeses, intraperitoneal chemotherapy, placing a drain-
mary in women and men who present with a malignant age catheter, or implanting a peritoneovenous shunt
pleural effusion is lung cancer. It is extremely uncom- (usually into the superior vena cava).2,48 Surprisingly,
mon for breast cancer to manifest itself initially as a there is no evidence that disseminating tumor cells via a
malignant effusion.23,45,46 The most common occult peritoneovenous shunt decreases survivial.2 The various
sources of a malignant peritoneal effusion are intes- treatment options have their advantages and disadvan-
tinal and pancreatic cancer in men and ovarian can- tages; selecting the best treatment option focuses on the
cer in women.46,47 Other tumors that can present as a patient’s desires and improving the quality of life.
malignant effusion include lymphoma, melanoma, and
mesothelioma.46 In some patients, the primary site is Tips for detecting malignant cells in effusions:
never discovered.23,46
• “second population”
Malignant cells in pleural, pericardial, or peritoneal
• numerous large clusters
fluid betoken a grim prognosis. The median survival for
• lacunae (cell block sections)
patients with a positive pleural or peritoneal effusion
136 Pleural, Pericardial, and Peritoneal Fluids

A good way to identify malignant cells in effusions is to nant effusions.30,50 Strongly linked in most cases to asbes-
first locate some benign mesothelial cells. With these as a tos exposure, it arises most commonly in the pleura and
benchmark, one searches for a “second population” of cells less commonly in the peritoneum; primary tumors of
(not counting, of course, any lymphocytes or histiocytes) the pericardium or tunica vaginalis of the testis are rare.
that is clearly different. Malignant cells are not necessarily The latency (time from first asbestos exposure to clini-
larger than the mesothelial cells. Some are about the same cal disease presentation) is extremely long, with an aver-
size but are recognized because of their high nuclear-to- age of 30 to 40 years. The peak incidence in the United
cytoplasmic ratio, nuclear hyperchromasia, or macronu- States appears to have happened in the 1990s, but cases
cleoli. Exceptions to this rule occur, notably mesothelioma, are still increasing in other countries like Great Britain
for which a sharp distinction between benign and neo- and Australia.30
plastic mesothelial cells is not appreciated. Malignant mesothelial cells grow as multiple plaques
Normal mesothelial cells virtually never form large cell that coalesce into larger nodules visible radiographi-
clusters. Effusions with numerous large cell aggregates cally as a thickening of the pleura. Histologically, these
are easily spotted as malignant. Care must be taken not tumors are classified as epithelial, sarcomatoid, or
to confuse loosely clustered cells, which are a common mixed (biphasic) types. The epithelial type comes in
artifact of cytocentrifugation and liquid-based prepara- a dizzying variety of histologic patterns: epithelioid,
tions. Reliably malignant clusters are tightly cohesive. deciduoid, tubulopapillary, microcystic, small cell, des-
Malignant cells in cell block sections are frequently sit- moplastic, and high grade (pleomorphic), but most of
uated in lacunae (Fig. 4.7). These clear spaces surround- these variants are rare.30 The most common patterns of
ing individual cells or groups of cells are seen in 75% of cell epithelial mesothelioma are the epithelioid and tubulo-
blocks of malignant effusions, mostly adenocarcinomas, papillary. Most mesotheliomas are, in fact, well-differen-
but the finding is not specific; lacunae are also seen in one tiated tumors and cytologically remarkably, deceptively
third of benign effusions.49 Lacunae are helpful in locat- bland. Mesotheliomas, like benign mesothelial cells, are
ing suspicious cells at low magnification, but inspection immunoreactive for cytokeratins (including CK 5 and
at high magnification is needed for definitive diagnosis. 6), desmin, vimentin, calretinin, and Wilms tumor pro-
tein 1 (WT1).
Common symptoms are chest pain and shortness of
Primary Tumors breath. Establishing the diagnosis is not always straight-
Primary tumors of the serosal surfaces are uncommon, forward, with a median time from the onset of symptoms
being far outnumbered by secondary involvement by to diagnosis of 8 weeks.38 Most patients have an effusion,
tumors from other locations. The two primary serosal usually unilateral, at the time of presentation, and the
malignancies considered here are malignant mesothe- fluid is often described as having the color and consis-
lioma and primary effusion lymphoma. tency of honey. When suspicious and positive results
are combined, the sensitivity of effusion cytology for the
diagnosis of mesothelioma is only 32%.38
Diffuse Malignant Mesothelioma
Diffuse malignant mesothelioma (for simplicity, referred Cytomorphology of mesothelioma:
to as mesothelioma) accounts for less than 2% of malig-
• two principal patterns:
• large clusters with scalloped (“knobby”) edges
• “noncohesive” (isolated cells)
• cytomegaly
• round, centrally placed nucleus
• prominent nucleolus
• binucleation and multinucleation
• dense cytoplasm with peripheral “halo”
• normal nuclear-to-cytoplasmic ratio
• windows

Only the epithelial and mixed (biphasic) types of


mesothelioma are likely to exfoliate malignant cells;
the pure sarcomatoid type rarely exfoliates. When the
Figure 4.7  Cell block lacunae (pleural fluid). In cell block sec- malignant cells exfoliate, the most common cytologic
tions, malignant cells are often situated in an empty space (lacuna);
the reason for this artifact is unknown. It is commonly seen with pattern is of numerous large clusters (morulae; Fig.
adenocarcinomas, rarely with lymphomas and melanoma. 4.8A). The clusters are composed of up to hundreds
Malignant Effusions 137

A B

C D
Figure 4.8  Malignant mesothelioma (pleural fluid). A,. Solid, morulae-like spheres, some of them elongated, are composed of
cells that resemble normal mesothelial cells. A fluid composed of many large clusters is virtually always malignant. B, A branching
pattern is seen in some cases. Note the knobby contours. C, In most mesotheliomas, the nuclear-to-cytoplasmic ratio of normal
­mesothelial cells is recapitulated. D, In other cases, the nuclear-to-cytoplasmic ratio is significantly increased.

of cells that are recognizably mesothelial in ori- Differential diagnosis of


gin, with round nuclei, prominent nucleoli, and mesothelioma:
dense cytoplasm with a pale rim. The morulae have
a knobby contour (“mulberry” clusters), and some • reactive mesothelial cells
show branching (Fig. 4.8B). In most cases, the malig- • metastatic tumor
nant mesothelial cells are larger than normal meso- • adenocarcinoma
thelial cells, sometimes markedly so. Cytoplasm is • squamous cell carcinoma
abundant in most cases, and therefore the nuclear- • epithelioid hemangioendothelioma
to-cytoplasmic ratio is often deceptively normal (Fig.
4.8C), but cytoplasm can be scant in some cases (Fig.
4.8D). Occasionally, microvilli can be appreciated Mesothelioma versus Reactive Mesothelial Cells. Because
(Fig. 4.9). Nuclear atypia is mild in most cases. On cell reactive mesothelial cells of the pleura and peritoneum do
block sections, the clusters are a solid mass of cells, or not form numerous large morulae, the diagnosis of meso-
they may contain a collagenous or acid mucopolysac- thelioma is straightforward when the specimen is highly
charide core (Fig. 4.10). cellular and contains many large clusters of enlarged
Not all fluids have the mulberry-cluster pattern. In mesothelial cells (see Figs. 4.8A). Such a specimen can be
another rather common pattern, the malignant cells are called, at the very least, suspicious for malignancy. In the
not cohesive but instead are dispersed as isolated cells appropriate clinical context (unilateral effusion, asbestos
(see Fig. 4.8C).51 More unusual variants include tumors exposure, and pleural thickening) it can be argued that
composed predominantly of vacuolated cells (Figs 4.11A they can reliably be called positive for malignancy.
and B), tumors that show small cell differentiation, those As mentioned previously, the striking morular pat-
with an abundant lymphohistiocytic infiltrate, and those tern is not seen in all mesotheliomas. In such cases,
accompanied by psammoma bodies.52 the distinction from reactive mesothelial cells is more
138 Pleural, Pericardial, and Peritoneal Fluids

be epithelial membrane antigen (EMA), but not all


investigators report good ­specificity. Although some
find excellent specificity,53-56 especially for EMA clone
E29,53 others do not.57,58
Although time consuming and not available in most
laboratories, cytogenetic analysis has high sensitivity and
specificity for the distinction between reactive mesothelial
cells and mesothelioma. In almost all cases, mesotheliomas
show clonal cytogenetic aberrations indicative of malig-
nancy, the most common being deletions of 1p, 3p, 6q, 9p,
and 22q.17 With a combination of appropriate probes, these
deletions can be detected more easily by fluorescence in
situ hybridization ([FISH]; Fig. 4.12).18,59

Figure 4.9  Malignant mesothelioma (pleural fluid). In some Mesothelioma versus Adenocarcinoma. Some adeno-
cases, the characteristic microvilli can be appreciated. carcinomas spread to involve the serosal ­surfaces in a
diffusely infiltrative, “pseudomesotheliomatous” pat-
tern that mimics mesothelioma. The similarity extends
to histopathology and cytopathology—exfoliated cells of
mesotheliomas and adenocarcinomas can be arranged
in cohesive clusters or dispersed in a noncohesive pat-
tern, and both can have vacuolated cytoplasm. A few
morphologic clues exist, however. Mesothelioma cells
form a morphologic continuum with benign-appearing
mesothelial cells at one end, whereas fluids that harbor
metastatic adenocarcinoma generally contain two dis-
tinct cell populations. Tumor cells that are separated by
slitlike “windows” and have abundant, dense cytoplasm
are more likely to be mesothelial in origin. On cell block
sections, a core of edematous collagen and stromal
cells, surrounded by neoplastic cells, is more commonly
Figure 4.10  Malignant mesothelioma (pleural fluids). In seen in mesothelioma than in adenocarcinoma (see Fig.
some cases, cell block sections show that clusters of malignant 4.10). Conversely, ringlike structures with hollow cores,
cells surround a collagenous core. seen in some adenocarcinomas, are uncommon in meso-
thelioma. Clusters with a knobby (mulberry-like) contour,
rather than the smooth, cannonball-like edge of many
problematic. There are precious few (if any) immu- adenocarcinomas, are characteristic of mesotheliomas
nohistochemical markers on which one can rely for (see Fig. 4.8B).
this distinction. The best marker for distinguishing These morphologic clues, unfortunately, are not
benign from malignant mesothelial cells appears to always reliable. Exceptions occur frequently enough that,

A B
Figure 4.11  Malignant mesothelioma. A, B, Rarely, the tumor cells show striking cytoplasmic vacuolization.
Malignant Effusions 139

9p21
CEP 9

Figure 4.12  Malignant mesothelioma. Mesotheliomas are characterized genetically by clonal chromosomal deletions. Fluorescence
in situ hybridization of pleural fluid shows a normal cell on the right and a mesothelioma cell on the left. The preparation has been incu-
bated with probes for the centromeric region (green) and the deleted region (red) of chromosome 9. Both cells show two centromeric
regions, but the mesothelioma cell is missing a ­segment of the short arm of chromosome 9. (Courtesy of Paola Dal Cin, Brigham and
Women’s Hospital, Boston.)

in a given case, morphology alone cannot be depended mesotheliomas, demonstrating a nuclear and cytoplas-
on for an unequivocal classification. Histochemical mic staining pattern (Fig. 4.13A). By contrast, only a
and immunocytochemical stains can make the distinc- small number of adenocarcinomas are positive, virtu-
tion in almost all cases and are indispensible in this ally always in a predominantly cytoplasmic pattern.60,61
regard (Table 4.2). Two so-called mesothelial cell mark- Similarly, the WT1 protein, the product of the WT1
ers are particularly useful in this distinction. Calretinin, gene, located on chromosome 11p and implicated in
a ­calcium-binding protein, is strongly positive in most the pathogenesis of Wilms tumors and mesotheliomas,
is strongly expressed in most mesotheliomas.62,63 The
staining is nuclear, not cytoplasmic (Fig. 4.13B). With
the exception of serous carcinomas, most adenocarci-
Table 4.2  Common Histochemical and
Immunohistochemical Staining Patterns for nomas are negative for WT1.
Mesothelioma and Metastatic Adenocarcinoma Mesotheliomas are typically negative for intracy-
toplasmic mucin with the mucicarmine and periodic
Expected Result*
acid-Schiff diastase (PAS-D) stains and negative for the
Stain Adenocarcinoma Mesothelioma carcinoma markers CEA, MOC-31, Ber-EP4, Leu-M1
PAS-D + − (CD15), and B72.3.64-66 In contrast, at least half of ade-
mucicarmine + − nocarcinomas in effusions are positive for cytoplasmic
CEA + − mucin, and most are immunoreactive for one or more
MOC-31 + − of the carcinoma markers CEA, MOC-31, Ber-EP4,
Ber-EP4 + − Leu-M1, and B72.3 (Fig. 4.14A). Thyroid transcription
Leu M-1 + − factor-1 (TTF-1) is particularly useful in the frequent
B72.3 + − distinction between lung adenocarcinoma and meso-
TTF-1 +(nuclear)† − thelioma. Strong nuclear staining is common in lung
calretinin − + (nuclear and adenocarcinomas and absent in mesotheliomas (Fig.
cytoplasmic) 4.14B).63,67
WT1 − + (nuclear)‡ A few caveats are in order. First, a critical analysis of
CEA, carcinoembryonic antigen; PAS-D, periodic acid-Schiff and dictase;
the mucin stains is important. Fine cytoplasmic gran-
TTF-1, thyroid transcription factor-1; WT1, Wilms tumor 1 protein. ules seen with the PAS-D stain, the interposition of
* Result observed in most but not all cases positively stained basement membrane material, and

 Positive in adenocarcinomas of the lung and thyroid only. Also
positive in small cell carcinomas.
an extracellular localization of staining should not

 Also positive in serous carcinomas of the ovary. be misinterpreted as a positive reaction for mucin.
140 Pleural, Pericardial, and Peritoneal Fluids

A calretinin B WT1

Figure 4.13  Immunoprofile of malignant mesothelioma. A, Mesotheliomas show nuclear and cytoplasmic staining for calretinin.
B, There is usually nuclear immunoreactivity for Wilms tumor 1 protein (WT1).

A CEA B TTF-1

Figure 4.14  Immunoprofile of metastatic adenocarcinomas. A, Most adenocarcinomas are immunoreactive for one or more of the
“carcinoma markers” like carcinoembryonic antigen ([CEA] shown here), but often only some of the cells are positive. B, Most adeno-
carcinomas of the lung are immunoreactive for thyroid transcription factor-1 (TTF-1), as are most thyroid cancers.

Table 4.3  Recommended Immunohistochemical


A ­positive staining reaction is nevertheless encountered
Panels Based on Differential Diagnosis*
in rare instances of mesothelioma.30,68 For this reason,
it is wise to use a panel of histochemical and immu- Differential Diagnosis Panel
nocytochemical stains from those listed in Table 4.2. Pleural mesothelioma Mesothelial markers calretinin,
A panel of four or five markers is sufficient in most versus lung WT1 and carcinoma
adenocarcinoma markers MOC-31, Ber-EP4,
cases and can be tailored to the particular differential
B72.3, CEA, TTF-1
diagnosis (Table 4.3).66
Peritoneal mesothelioma Mesothelial marker calretinin
Mesothelioma can also be distinguished from met- versus serous carcinoma and carcinoma markers
astatic adenocarcinoma by electron microscopy; the MOC-31 Ber-EP4, estrogen
microvilli of mesothelioma cells have a length to diam- receptors
eter ratio of 15:1 or greater, whereas those of adeno- Mesothelioma versus Mesothelioma markers
carcinoma have a smaller ratio.16 This method requires squamous carcinoma calretinin, WT1 and carcinoma
markers p63, MOC-31
special fixation of the specimen for optimal results,
Mesothelioma versus renal Mesothelioma markers
however, and is rarely used.
cell carcinoma calretinin, WT1 and carcinoma
markers LeuM1 (CD15), renal
Mesothelioma versus Squamous Cell Carcinoma. In cell carcinoma antigen
effusions as in histologic sections, certain mesothe-
liomas can resemble a squamous cell carcinoma, CEA, carcinoembryonic antigen; TTF-1, thyroid transcription factor-1;
WT1, Wilms tumor protein 1.
­particularly because both typically have dense cyto- * Modified from Ordonez NG: What are the current best
plasm. Immunohistochemistry can be helpful in this immunohistochemical markers for the diagnosis of epithelioid
distinction (see Table 4.3) mesothelioma? A review and update. Hum Pathol 2007;38(1):1-16.
Malignant Effusions 141

Mesotheliomas versus Vascular Tumors. Primary vas-


Cytomorphology of primary effusion
cular tumors of the lung (angiosarcomas and epithe-
lymphoma:
lioid hemangioendotheliomas) are rare but they can
present with a pseudomesotheliomatous growth pat- • dispersed large cells
tern that mimics mesotheliomas not just clinically • round or irregular nucleus
and radiologically but also cytologically.69 They arise • prominent nucleoli
in the lung but spread to involve the pleural surface • abundant basophilic cytoplasm (Romanowsky
in a diffuse pattern. Epithelioid hemangioendothe- stain)
liomas bear an uncanny resemblance to epithelial
mesotheliomas (Fig. 4.15). A panel of immunostains
that includes vascular markers (CD31 and CD34) and The cells are always large, ranging from plasmablas-
mesothelial markers (calretinin and WT1) are helpful tic or immunoblastic (round nucleus with prominent
in this distinction. nucleolus) to anaplastic (large, irregular, multilobated
nucleus) in morphology (Fig. 4.16A). Cytoplasm is
abundant and may have a perinuclear hof or vacuoles.
Primary Effusion Lymphoma
Mitoses and apoptotic bodies are present and can be
Primary effusion (body-cavity based) lymphoma (PEL) numerous.71 The diagnosis of malignancy is straightfor-
is a rare subtype of diffuse large B-cell lymphoma that ward. Given the usual clinical setting of an immunode-
is associated with human herpes virus 8 (HHV-8) and ficiency, a lymphoma should be suspected; this can be
presents as a pleural, pericardial, or peritoneal effu- confirmed by immunocytochemistry. PEL cells usually
sion.70 Most cases have a null-cell immunophenotype; express CD45 (leukocyte common antigen) but are neg-
B-cell clonality is usually demonstrated by molecular ative for the B-cell markers CD19 and CD20. Other lym-
studies. Rare PELs have a T-cell immunophenotype.71,72 phoid markers, like the activation marker CD30 and the
All cases are positive for HHV-8; indeed, detection of plasma cell markers CD38 and CD138, are often pres-
HHV-8 is an essential step in confirming the diagnosis of ent.73 Demonstrating the presence of human HHV-8 is
PEL. By definition, this is a fluid-based malignancy with- essential for diagnosis (see Fig. 4.16B).72 In most cases
out an associated mass lesion, lymphadenopathy, or the neoplastic cells are co-infected with Epstein-Barr
organomegaly.73 Most cases arise in the setting of human virus (EBV ), best demonstrated by ­EBV-encoded RNA
­immuno­deficiency virus (HIV) infection, but rare cases (EBER) in situ hybridization.72
in transplant recipients have also been reported. The prog- PEL cells are markedly atypical and obviously
nosis is poor: median survival is less than 6 months.73,74 malignant. It is usually simply a question of classifying

Figure 4.15  Epithelioid hemangioendothelioma of the lung (pleural fluid). This tumor is a good mimic of mesothelioma because
the cells form large aggregates and have round, centrally placed nuclei and abundant cytoplasm.
142 Pleural, Pericardial, and Peritoneal Fluids

A B HHV-8

Figure 4.16  Primary effusion lymphoma. A, The malignant cells are large, with thick nuclear membranes, irregularly distributed
chromatin, and prominent nucleoli. Apoptotic bodies are present. B, The presence of human herpes virus 8 (HHV-8), demonstrated
here by immunohistochemistry, is a sine qua non of primary effusion lymphomas.

Differential diagnosis of primary Metastatic Tumors


effusion lymphoma:
Adenocarcinoma
• diffuse large B-cell lymphoma (not PEL)
Metastatic carcinomas are by far the most common
• pyothorax-associated lymphoma
tumors found in effusions. Of these, adenocarcinomas
• anaplastic large cell lymphoma
are more common than squamous and small cell cancers.
• post-transplant lymphoproliferative disorder
In most cases the carcinoma cells are easily recognized
• carcinoma
because, in one way or another, they are morphologi-
• melanoma
cally distinct from mesothelial cells (Fig. 4.17). This is the
key to identifying them on cytologic preparations.
the malignancy. PEL is morphologically similar to the The cells of metastatic carcinoma are often but not
immunoblastic type of diffuse large B-cell lymphoma necessarily larger, more pleomorphic, and more hyper-
(DLBL). If the patient has a mass lesion, lymphade- chromatic than mesothelial cells. They can, in fact, be
nopathy, or organomegaly along with the effusion, the smaller and more uniform in size than the mesothelial
lymphoma is not classified as a PEL but is instead a cells around them. Most commonly, they are ­distinguished
conventional DLBL. Immunohistochemistry can also
distinguish these entities because DLBL expresses
B-cell markers and is negative for HHV-8. Pyothorax-
associated lymphoma (PAL) is a rare, EBV-associated
subtype of DLBL that develops after longstanding
chronic pleural inflammation, and there is usually an
associated pleural mass. Like other DLBLs, it expresses
B-cell markers and is negative for HHV-8. Anaplastic
large cell lymphoma (ALCL) sometimes presents with
a pleural effusion. The “hallmark cells” (with horse-
shoe-shaped nuclei) typical of anaplastic large cell
lymphoma (see Fig. 11.26) can be seen in some cases
of PEL, but anaplastic large cell lymphoma is nega-
tive for HHV-8. In transplant recipients, the possibil-
ity of a post-transplant lymphoproliferative disorder
(PTLD) should be considered (see Chapter 11). PTLDs
are associated with EBV but are negative for HHV-8.75
Figure 4.17  Adenocarcinoma of the lung (pleural fluid).
Carcinomas and melanoma can be excluded by the Clusters of large, highly atypical cells like these are easily spotted
absence of immunoreactivity for keratins, S-100 pro- and identified as malignant, but in the absence of a known pri-
tein, and HMB-45. mary, special stains might be needed for precise classification.
Malignant Effusions 143

from benign mesothelial cells by their tendency to form Adenocarcinomas exfoliate as large spheres or as iso-
large clusters (see Fig. 4.7). Other carcinomas, however, lated cells. Cytoplasmic vacuoles are often present, but
exfoliate in a noncohesive way, as isolated cells instead they are not specific for adenocarcinomas; they are seen
(Fig. 4.18). In such cases, cell size, increased nuclear-to- in some mesotheliomas and other tumors. Some mor-
cytoplasmic ratio, irregular nuclear contours, prominent phologic clues can point to the site of origin of an ade-
nucleoli, or coarsely textured chromatin distinguish nocarcinoma. For example, abundant hollow spheres
them from mesothelial cells. (“cannonballs”) are a common pattern in metastatic
breast cancer (Fig. 4.19A and B). Malignant signet ring
Cytomorphology of adenocarcinomas: cells in an effusion point to an origin from stomach can-
cer, but other primary sites, such as the breast, are also
• large spheres or single cells
possible (Fig. 4.20). Most colorectal cancers are com-
• cytoplasmic vacuolization
posed of elongated cells with hyperchromatic nuclei
• signet ring cells (gastric, breast)
arranged in acinar formations; individual cell necrosis is
a common finding. Large cells with prominent nucleoli
and abundant lacy, vacuolated cytoplasm are typical of
clear cell carcinomas of the kidney and the female gen-
ital tract (Fig. 4.21). Most metastatic prostate cancers
exfoliate as isolated cells or loosely cohesive groups of
cells with prominent nucleoli. Some high-grade meta-
static prostate cancers resemble small cell carcinoma.76
Psammoma bodies are encountered in effusions
caused by serous neoplasms of the ovary, fallopian tube,
and endometrium; papillary carcinomas of the thyroid;
adenocarcinomas of the lung; and mesotheliomas (Fig.
4.22). They are also seen in some benign proliferative
reactions of the mesothelium, especially in the perito-
neum,77 but also the pericardium,29 and by themselves
should not be taken as evidence of malignancy.
When some mucinous adenocarcinomas, particu-
larly those of the appendix, spread to involve the peri-
toneal surfaces, they produce a slow but relentless
accumulation of extracellular mucin that eventually
distends the peritoneal cavity, a condition known as
Figure 4.18  Adenocarcinoma of the lung (pleural fluid). In pseudomyxoma peritonei. Peritoneal fluid from such
cases like this, the malignant cells can be more difficult to recog- patients is gelatinous and composed predominantly
nize because they are dispersed as isolated cells. Careful exam-
ination reveals markedly enlarged nucleoli and a cytoplasmic of mucin, which stains blue-green or purple with the
texture that is frothier and less dense than that of most meso- Papanicolaou stain. Such specimens are often sparsely
thelial cells. cellular and may contain only vacuolated histiocytes

A B
Figure 4.19  Ductal carcinoma of the breast (pleural fluid). Ductal cancers often exfoliate as large spheres of malignant cells (A,
thinlayer slide), which may be hollow (B, cell block).
144 Pleural, Pericardial, and Peritoneal Fluids

Figure 4.22  Metastatic papillary carcinoma of the thyroid


Figure 4.20  Adenocarcinoma of the stomach (pleural fluid). (pleural fluid). This large irregular cluster of malignant cells is
Large numbers of isolated signet ring cells are characteristic of associated with a psammoma body.
many gastric cancers.
called muciphages (Fig. 4.23A). The tumor cells in the
peritoneum that produce the mucin (Fig. 4.23B) often
comprise just a tiny fraction of the volume of the spec-
imen and may not be present in the slides examined.
When there are no definite malignant cells (just mucin
and muciphages), only a presumptive diagnosis of
malignancy can be offered.

Differential diagnosis of metastatic


adenocarcinoma:
• reactive mesothelial cells
• mesothelioma

It is usually easy to distinguish metastatic adenocar-


cinoma from reactive mesothelial cells. Malignant cells
in cell block sections are frequently arranged in large
clusters or are situated in lacunae (see Fig. 4.7). In some
Figure 4.21  Clear cell carcinoma of the kidney (pleural
fluid). Large cells with round nuclei, prominent nucleoli, and cases, however, metastatic adenocarcinoma cells are
abundant granular and vacuolated cytoplasm are typical of renal not easily recognized. A clear second population (mor-
cell carcinoma. phologically distinct from mesothelial cells) may not

A B
Figure 4.23  Pseudomyxoma peritonei (peritoneal fluid). A, Extracellular mucin is present in abundance, and stains blue or pur-
ple with the Papanicolaou stain. Malignant cells are often not seen; the few cells in this field are histiocytes. B, In this case, rare strips
of neoplastic epithelium were identified in cell block sections.
Malignant Effusions 145

be apparent; the tumor cells might mimic mesothelial The differential diagnosis of adenocarcinoma and
cells, or the suspicious cells might be few in number or mesothelioma has been discussed at length previously.
poorly preserved. In any doubtful case, special stains Immunostains are indispensable for a definitive diagno-
for mucin and immunohistochemistry for carcinoma sis (see Table 4.3)
markers (see Table 4.2) are helpful for distinguishing
reactive mesothelial cells (negative) from adenocarcino- Squamous Cell Carcinoma
mas (positive).28,65 A panel of four or five immunomark-
Squamous cell carcinomas (SQCs) rarely metastasize to
ers is sufficient in most cases. The panel can be selected
the pleura, pericardium, or peritoneum. Those that do
based on clinical history (Table 4.3 can be applied for
are most commonly carcinomas of the lung, larynx, and
this purpose) and might include, in addition to carci-
female genital tract.78 The primary tumor is known in vir-
noma markers, two mesothelial markers like calretinin
tually all cases before an effusion develops.
and WT-1.66
Lobular carcinoma of the breast is perhaps the most
Cytomorphology of squamous cell
subtle of all the adenocarcinomas; with only rou-
carcinoma:
tine stains, the cells of this tumor can be impossible
to distinguish from histiocytes and mesothelial cells • large clusters or isolated cells
(Fig. 4.24A). For this reason, it is advisable to do spe- • keratinized or nonkeratinized
cial stains for mucin (mucicarmine and PAS-D) and a • dense cytoplasm
limited panel of immunostains (e.g., CEA, gross cystic
disease fluid protein, estrogen and progesterone recep-
tors) on any effusion from a patient with lobular breast The cells of squamous cell carcinoma are arranged
cancer (Fig. 4.24B). either in large clusters (Figs 4.25A and B, 4.26) or as iso-
Immunohistochemical markers are also helpful in lated cells. The cytologic appearance differs depend-
patients with a tumor of unknown primary or a his- ing on the degree of keratinization of the tumor.
tory of two or more neoplasms. Useful tissue-spe- The ­cytoplasm is usually dense and occasionally
cific antigens include TTF-1 (for adenocarcinoma and ­orangeophilic. Cells with a ­tadpole or spindle shape are
small cell carcinoma of the lung and thyroid cancers, uncommon. Nuclei are enlarged, hyperchromatic, and
see Fig. 4.14B),67 estrogen and progesterone receptors coarsely granular; nucleoli are usually not prominent.
(for breast and gynecologic malignancies), thyroglob- Nuclear pyknosis and karyorrhexis are seen in some
ulin, prostate-specific antigen (PSA), and prostatic cases. Anucleated squames may be present. Cytoplasmic
acid phosphatase (PAP). Negative staining for both vacuolization is sometimes seen, and should not be
prostate-specific antigen and prostatic acid phos- interpreted as indicative of glandular differentiation.78
phatase does not exclude metastatic prostatic cancer, A panel of immunostains that includes p63, MOC-31,
however, because less than 50% of metastatic prostate and the mesothelial markers calretinin and WT-1 can be
cancers in pleural effusions are immunoreactive for useful to distinguish squamous cell carcinoma from reac-
these markers.76 tive mesothelial cells and mesothelioma (see Table 4.3).

A B
Figure 4.24  Lobular carcinoma of the breast (pleural fluid). A, These malignant cells are extremely difficult to recognize because
they resemble histiocytes or mesothelial cells (Papanicolaou stain). B, A mucicarmine stain reveals focal intracytoplasmic mucin.
146 Pleural, Pericardial, and Peritoneal Fluids

A B
Figure 4.25  Squamous cell carcinoma of the cervix (pericardial fluid). Nonkeratinizing squamous cell cancers shed large spheres
of malignant cells. A, Cytocentrifuge preparation. B, Cell block preparation.

The differential diagnosis includes lymphoma. The


cells of small cell carcinoma are distinguished by their
tendency to form clusters. Although the nuclei of lym-
phoma cells can be irregular in shape, they are rarely
sharply angular like the cells of small cell carcinoma.
Most small cell lung cancers, unlike lymphomas, are
immunoreactive for TTF-1, but a negative result does
not exclude the diagnosis. The differential diagnosis also
includes other small cell malignancies like small cell car-
cinoma of the prostate76 and Merkel cell carcinoma. In
most cases, the clinical history of a specific malignancy
points in the correct direction. Immunohistochemistry
can be helpful if needed: For example, small cell carcino-
mas are immunoreactive for CK7 and negative for CK20;
Figure 4.26  Squamous cell carcinoma of the lung (pleural the reverse is true for Merkel cell carcinoma.
fluid). The malignant cells have coarsely textured chromatin and
plate-like cytoplasm.
Melanoma
Small Cell Carcinoma Most malignant melanomas arise in the skin, but extra-
Small cell carcinoma of the lung, despite its predilection cutaneous tumors, such as ocular melanomas, do occur.
for widespread metastases, causes a pleural effusion in In 5% of cases, patients present with metastatic disease
less than 3% of patients.79

Cytomorphology of small cell


carcinoma:
• small cells (isolated, in chains and clusters)
• nuclear molding
• scant cytoplasm

The cells are dispersed as isolated cells and arranged


in clusters and chains. They are small, with a diam-
eter approximately two to three times that of small
lymphocytes. Cytoplasm is scant. The nuclei are dark
and have a finely granular chromatin texture; nucle-
oli are inconspicuous. Karyorrhexis is common. When
Figure 4.27  Small cell carcinoma of the lung (pleural fluid).
arranged in groups, the cells are crescent shaped Some of the small malignant cells are dispersed as isolated cells,
and angulated as they wrap themselves around one whereas others are molded together in clusters. The mesothelial
another (Fig. 4.27). cell on the right provides a size comparison.
Malignant Effusions 147

without a known primary or with only a remote ­history and cleaved nuclei and scant cytoplasm. Lymphoblastic
of a pigmented cutaneous lesion. To compound the lymphomas are also composed of relatively small cells
problem, the diagnosis of melanoma can be subtle in with round or irregularly shaped nuclei, finely dispersed
effusions. The malignant cells resemble mesothelial chromatin, and scant or moderate amounts of cyto-
cells: they are often dispersed as isolated round cells plasm (Fig. 4.30). Small lymphocytic lymphoma rarely
with prominent nucleoli (Fig. 4.28A). In some cases, involves serosal cavities. Burkitt and Burkitt-like lympho-
cells show a fine brown cytoplasmic pigmentation or mas are high-grade neoplasms composed of lymphoid
intranuclear pseudoinclusions. Cell clusters are uncom- cells of intermediate size with round nuclei, prominent
mon (Fig. 4.28B), and cell blocks rarely show pericellu- nucleoli, and coarsely textured chromatin. Mitoses are
lar lacunae. The distinction from reactive mesothelial numerous.
cells is difficult in cases that show little nuclear pleo- Karyorrhexis is a conspicuous feature of many lym-
morphism and hyperchromasia and no cytoplasmic phomas (see Fig. 4.29). It is seen in both treated and
pigmentation.80 Immunocytochemistry is helpful; mel- untreated patients and is an uncommon finding in
anomas are reactive for S-100 protein and HMB-45, and benign effusions or malignant effusions resulting from
negative for keratin proteins. other tumors.
Precise classification of the lymphoma based on its
morphology is rarely necessary with effusions because
Non-Hodgkin Lymphoma
most patients have biopsy-proven disease before the
A malignant effusion complicates the course of dis- effusion develops. Nevertheless, some degree of subclas-
ease in many patients with non-Hodgkin lymphoma. sification is usually possible based on the size of the cells,
In fact, 10% to 15% of malignant effusions are caused the degree of nuclear membrane irregularity (cleaved or
by lymphoma; the proportion is significantly higher in noncleaved), and whether the cells resemble lympho-
the pediatric population.23,81 Most represent secondary blasts or Burkitt cells. Such features can be appreciated
involvement of serosal surfaces; few are PELs. Certain especially well on air-dried Romanowsky-stained prepa-
subtypes have a greater propensity for involving serosal rations (Fig. 4.30).
surfaces (e.g., lymphoblastic lymphoma).
Cytologic preparations are highly cellular and com-
posed of dispersed lymphoid cells. Lacunar spaces are Differential diagnosis of secondary
not seen in cell block sections with lymphomas. In many involvement by lymphoma:
cases, mesothelial cells are conspicuously absent. The • PEL
cells of DLBLs are easiest to recognize because they are • benign lymphocytic effusion
larger than histiocytes and their nucleoli are usually • PTLD
prominent (Fig. 4.29). Nuclei are round or highly irregu- • carcinoma
lar, and the chromatin is coarsely textured. Cytoplasm is • mesothelioma
often abundant, pale, and vacuolated. The cells of small • melanoma
cell lymphomas are only slightly larger than normal lym- • small round blue-cell tumors
phocytes. Those of follicular lymphomas have irregular

A B
Figure 4.28  Melanoma (pleural fluid). A, Nonpigmented melanoma cells are usually isolated, not clustered, and resemble meso-
thelial cells. B, Less often, they aggregate in large clusters.
148 Pleural, Pericardial, and Peritoneal Fluids

Figure 4.29  Non Hodgkin lym-


phoma: diffuse large B-cell type
(peritoneal fluid). The lymphoma
cells have predominantly round
nuclei with prominent nucleoli.
Note the karyopyknosis and
karyorrhexis, characteristic of
most lymphomas.

Figure 4.30  Lymphoblastic


lymphoma. Because lympho-
blastic lymphoma cells are only
slightly larger than normal lym-
phocytes, they are easily misdi-
agnosed. They are recognized
by their more finely dispersed
chromatin texture, better appre-
ciated on air-dried preparations
(Wright-Giemsa stain).

Secondary involvement by a large-cell non-Hodgkin phoma) includes a benign lymphocytic effusion like
lymphoma like DLBL or anaplastic large cell lymphoma that resulting from tuberculosis. The diagnosis of
can be morphologically indistinguishable from a PEL. tuberculosis should be considered if there are suspi-
The absence of a documented primary or of any mass cious clinical findings and the fluid is composed pre-
lesion, lymphadenopathy, or organomegaly in a patient dominantly of small mature lymphocytes. The cells of
who is immunocompromised should raise the possi- small lymphocytic lymphoma are virtually impossible
bility of PEL, which can be confirmed by demonstrat- to distinguish from small, mature lymphocytes with
ing the presence of HHV-8. The differential diagnosis Papanicolaou-stained preparations, even with the help
of secondary involvement by a lymphoma composed of computer-assisted morphometry.82 Given that sero-
predominantly of small cells (e.g., lymphoblastic lym- sal involvement by small lymphocytic lymphoma (or its
Malignant Effusions 149

A B
Figure 4.31  Non-Hodgkin lymphoma. In doubtful cases, immunostains for immunoglobulin light chains, performed on air-dried
cytocentrifuge preparations, can be helpful. Here the malignant lymphoid cells are immunoreactive for κ light chains A, and negative
for λ light chains B.

leukemic counterpart, chronic lymphocytic leukemia)


is a highly rare event (some authors who have reviewed
Hodgkin Lymphoma
large series of malignant lymphomas could find no Patients with Hodgkin lymphoma can develop benign
autopsy-documented cases),83 it is wise to ­document and malignant effusions. The more common benign
immunoglobulin light chain restriction before ren- effusions are likely a result of thoracic duct obstruction
dering a diagnosis of malignancy. κ and λ light chain or impaired lymphatic drainage resulting from a tumor
expression can be examined on air-dried cytocentri- that is nearby but not directly involving the serosal sur-
fuge preparations using immunocytochemistry7 or by face. Malignant effusions are relatively uncommon and
flow cytometry.13 Clonal excess is determined in many are almost never the initial manifestation of the disease.
cases by a simple inspection of the proportion of B The cytologic hallmark is the Reed-Sternberg cell,
cells that express κ or λ light chains (Fig. 4.31A and B). a large, multinucleated cell with huge inclusion-like
This method is a useful adjunct for the cytologic eval- nucleoli (Fig. 4.32). Mononuclear variants are often pres-
uation of lymphocyte-rich effusions that are cytologi- ent, together with a mixed population of inflammatory
cally equivocal for malignancy.14 In the case of the less cells that includes lymphocytes, plasma cells, eosino-
common T-cell lymphoma, a T-cell phenotype with phils, neutrophils, and histiocytes.
aberrant markers can be demonstrated by immunocy- In a patient with a history of Hodgkin lymphoma, a
tochemistry or flow cytometry, confirming a malignant fluid composed of a mixed population of inflammatory
diagnosis.7 cells but without Reed-Sternberg cells is considered sug-
In a transplant recipient, the possibility of a PTLD gestive of malignancy.
must be considered whenever an effusion contains a
prominent lymphoid population. PTLDs are associated
with EBV, best demonstrated by EBV-encoded RNA in
situ hybridization and are negative for HHV-8.
Lymphomas are rarely confused with other malignan-
cies because other tumors tend to form cell clusters in
effusions. In some preparations, a crowding of the lym-
phoma cells mimics cluster formation, but such arti-
factual clustering is usually recognizable for what it is.
Conversely, some carcinomas, mesotheliomas, and mel-
anomas shed in a noncohesive pattern and can resemble
the cells of a large-cell lymphoma (see Figs. 4.8C, 4.18).
If there is any doubt concerning the lymphoid nature of
a malignant effusion, a panel of immunomarkers (e.g.,
leukocyte common antigen, keratin proteins, calretinin,
WT1, S-100, HMB-45) can be helpful. Similarly, small
round blue cell tumors (e.g., neuroblastoma, Ewing Figure 4.32  Hodgkin lymphoma (pleural fluid). The classic
sarcoma/PNET) can be ruled out with the appropriate Reed-Sternberg cell is a large, multincleated cell, with promi-
immunohistochemical panel. nent nucleoli.
150 Pleural, Pericardial, and Peritoneal Fluids

The cells of chronic lymphocytic leukemia are indistin-


Multiple Myeloma
guishable from small, mature lymphocytes. Thus, immu-
Multiple myeloma rarely involves the serosal cavities. The nophenotyping is necessary to establish this diagnosis.
pleura is involved more commonly than the peritoneum In patients with circulating blasts, the possibility that the
or pericardium. Pleural involvement is sometimes a fluid was contaminated by peripheral blood during a trau-
direct extension of the tumor from an erosive rib lesion. matic tap is likely if the specimen contains red blood cells.
The degree of plasmacytoid differentiation varies from
one tumor to another. The cells are large and dispersed
Sarcomas
as isolated cells. Nuclei are round and have coarsely tex-
tured chromatin with prominent nucleoli. Cytoplasm is Virtually any sarcoma can metastasize to the sero-
usually abundant, nuclei are eccentrically positioned sal surfaces, although they do so much less frequently
within the cell, and there is a perinuclear clear zone than other tumors. When they do, it is usually late in the
(Fig. 4.33A). In poorly differentiated tumors the cells course of the disease. Those that have been reported in
have less cytoplasm and vary in size and shape.84 effusions range in morphology from spindle cell sarco-
Immunocytochemistry reveals monotypic expres- mas, like leiomyosarcoma, synovial sarcoma, and malig-
sion of either κ or λ light chains. Cell block sections are nant nerve sheath tumors, to small round cell sarcomas,
particularly well suited because the immunoglobulin like Ewing sarcoma/PNET, neuroblastoma, and embryo-
is cytoplasmic in these tumors. Curiously, although the nal rhabdomysarcoma.46
cells are negative for keratin proteins, they often express The cytomorphology of sarcomas in effusions is highly
epithelial membrane antigen.84 They are also positive for variable, depending on the sarcoma involved. The cells
the plasma cell marker CD138 (Fig. 4.33B). may be cohesive, exfoliating as tissue fragments (see Fig.
4.15), or they may be dispersed as isolated cells (Fig. 4.34),
and the individual cells can be small or large. Small round
Acute and Chronic Leukemias
cell sarcomas, like embryonal rhabdomyosarcoma, exfo-
Acute lymphoblastic and myeloblastic leukemias occa- liate predominantly as dispersed cells, mimicking the pat-
sionally involve the serosal surfaces. Blasts are round, tern of a non-Hodgkin lymphoma. Immunocytochemistry
two or three times the diameter of lymphocytes, and dis- is helpful in resolving equivocal cases.
persed as isolated cells. The nuclei are round or irregu-
larly shaped. The chromatin is pale and finely dispersed,
Germ Cell Tumors
and nucleoli are usually prominent.
Lymphoblasts cannot be distinguished from myelo- Like sarcomas, germ cell tumors rarely cause malignant
blasts on Papanicolaou-stained preparations, but this is effusions. When they do, it is usually late in the course of
rarely if ever important because the type of leukemia has widely disseminated disease.
usually been determined long before the patient devel- The seminoma of the testis, and its counterpart, the
ops an effusion. Auer rods, pink rodlike structures in the dysgerminoma of the ovary, are composed of ­dyshesive
cytoplasm, are diagnostic of myeloid differentiation, and uniform cells that resemble enlarged mesothelial
can be appreciated on air-dried Romanowsky-stained cells. They have less cytoplasm than mesothelial cells,
preparations. ­however, and more prominent nucleoli.

A B CD138

Figure 4.33  Multiple myeloma (pleural fluid). A, The fluid is composed almost exclusively of malignant plasma cells with abundant
cytoplasm and eccentrically placed nuclei. B, The malignant cells are immunoreactive for CD138.
references 151

Nonseminomatous germ cell tumors, which include


embryonal carcinoma, endodermal sinus (yolk sac)
tumor, choriocarcinoma, and malignant teratoma, are
composed of large pleomorphic cells with pale, finely
granular chromatin and prominent, often multiple,
nucleoli. The cells are morphologically indistinguish-
able from poorly differentiated large cell carcinomas.
Immunocytochemistry for placental alkaline phospha-
tase is helpful in distinguishing nonseminomatous germ
cell tumors (positive) from large cell carcinomas (neg-
ative). Special studies, however, are rarely necessary
because these tumors rarely manifest themselves ini-
tially as a malignant effusion.
Rarely, sex cord-stromal tumors can also involve sero-
sal surfaces and cause an effusion, sometimes many
years after treatment of the primary tumor (Fig. 4.35).
Figure 4.34  Pleomorphic rhabdomyosarcoma. The malignant
cells are isolated. Binucleation, eccentrically placed nuclei, and Immunohistochemistry for inhibin, a marker of many
dense cytoplasm are typical of many rhabdomyosarcomas. sex cord-stromal tumors, along with comparison of the
effusion specimen with sections from the original tumor,
can be indispensible for correct interpretation.

A B
Figure 4.35  Adult granulosa cell tumor (ascites). A, The malignant cells are uniform and bland and resemble benign mesothe-
lial cells. The arrangement in large clusters was suspicious, as was the prominence of nuclear grooves and focal follicle formation. B,
Comparison with the original histologic sections was helpful in establishing the diagnosis.

References
1. Thomsen TW, DeLaPena J, Setnik GS: Videos in clinical 6. Gabriel C, Achten R, Drijkoningen M: Use of liquid-
medicine. Thoracentesis. N Engl J Med 2006;355(15):16e. based cytology in serous fluids: A comparison with
2. Covey AM: Management of malignant pleural effusions and conventional cytopreparatory techniques. Acta Cytol
ascites. J Support Oncol 2005;3(2):169-173 76. 2004;48(6):825-835.
3. Roffe BD, Wagner FH, Derewicz HJ, Gill GW: Heparinized 7. Martin SE, Zhang HZ, Magyarosy E, et al.: Immunologic
bottles for the collection of body cavity fluids in cytopathology. methods in cytology: Definitive diagnosis of non-Hodgkin’s
Am J Hosp Pharm 1979;36:211-214. lymphomas using immunologic markers for T- and B-cells.
4. Manosca F, Schinstine M, Fetsch PA, et al.: Diagnostic effects of Am J Clin Pathol 1984;82:666-673.
prolonged storage on fresh effusion samples. Diagn Cytopathol 8. De Girolami E: Applications of plasma-thrombin cell block in
2007;35(1):6-11. diagnostic cytology. II: Digestive and respiratory tracts, breast
5. Ylagan LR, Zhai J: The value of ThinPrep and cytospin and effusions. Pathol Annu 1977;12(Part 2):91-110.
pre­paration in pleural effusion cytological diagnosis of 9. De Girolami E: Applications of plasma-thrombin cell block in
mesothelioma and adenocarcinoma. Diagn Cytopathol diagnostic cytology. I: Female genital and urinary tracts. Pathol
2005;32(3):137-144. Annu 1977;12(Part 1):251-275.
152 Pleural, Pericardial, and Peritoneal Fluids

10. Naylor B: Pleural, peritoneal, and pericardial fluids. In Bibbo M 33. Kalomenidis I, Light RW: Pathogenesis of the eosinophilic
(ed): Comprehensive Cytopathology, 2nd ed. Philadelphia, WB pleural effusions. Curr Opin Pulm Med 2004;10(4):289-293.
Saunders, 1997, pp. 551-621. 34. Romero Candeira S, Hernandez Blasco L, Soler MJ, et al.:
11. Starr RL, Sherman ME: The value of multiple preparations in Biochemical and cytologic characteristics of pleural
the diagnosis of malignant pleural effusions: A cost-benefit effusions secondary to pulmonary embolism. Chest 2002;
analysis. Acta Cytol 1991;35:533-537. 121(2):465-469.
12. Fetsch PA, Simsir A, Brosky K, Abati A: Comparison of 35. Veress JF, Koss LG, Schreiber K: Eosinophilic pleural effusions.
three commonly used cytologic preparations in effusion Acta Cytol 1979;32:40-44.
immunocytochemistry. Diagn Cytopathol 2002;26(1):61-66. 36. Naylor B, Novak PM: Charcot-Leyden crystals in pleural fluids.
13. Czader M, Ali SZ: Flow cytometry as an adjunct to Acta Cytol 1985;29:781-784.
cytomorphologic analysis of serous effusions. Diagn Cytopathol 37. Spieler P: The cytologic diagnosis of tuberculosis in pleural
2003;29(2):74-78. effusions. Acta Cytol 1979;23:374-378.
14. Ibrahim RE, Teich D, Smith BR, et al.: Flow cytometric surface 38. Renshaw AA, Dean BR, Antman KH, et al.: The role of cytologic
light chain analysis of lymphocyte rich effusions: A useful evaluation of pleural fluid in the diagnosis of malignant
adjunct to cytologic diagnosis. Cancer 1989;63:2024-2029. mesothelioma. Chest 1997;111:106-109.
15. Zarbo RJ: Flow cytometric DNA analysis of effusions: a new test 39. Naylor B: The pathognomonic cytologic picture of rheumatoid
seeking validation. Am J Clin Pathol 1991;95:2-4. pleuritis. Acta Cytol 1990;34:465-473.
16. Kobzik L, Antman KH, Warhol MJ: The distinction of 40. Zuffery P, Ruzicka J, Gerster JC: Pleural fluid cytology as
mesothelioma from adenocarcinoma in malignant effusions an indication of an effusion of rheumatoid origin [Letter].
by electron microscopy. Acta Cytol 1985;29:219-225. J Rheumatol 1993;20:1449-1451.
17. Granados R, Cibas ES, Fletcher JA: Cytogenetic analysis of 41. Naylor B: Cytological aspects of pleural, peritoneal, and
effusions for malignant mesothelioma: A diagnostic adjunct to pericardial fluids from patients with systemic lupus
cytology. Acta Cytol 1994;38:711-717. erythematosus. Cytopathology 1992;3:1-8.
18. Factor RE, Dal Cin P, Fletcher JA, Cibas ES: The value of 42. Balachandran I, Jones DB, Humphrey DM: A case of
cytogenetics and molecular cytogenetics as adjuncts to Pneumocystis carinii in pleural fluid with cytologic, histo-
cytology in the diagnosis of malignant mesothelioma in pleural logic, and ultrastructural documentation. Acta Cytol 1990;
and peritoneal fluids. Mod Pathol 2008;21:72A. 34:486-490.
19. Motherby H, Nadjari B, Friegel P, et al.: Diagnostic accuracy of 43. Elwood LJ, Dobrzanski D, Feuerstein IM, Solomon D:
effusion cytology. Diagn Cytopathol 1999;20:350-357. Pneumocystis carinii in pleural fluid: The cytologic appearance.
20. Spriggs AI, Boddington MM: Atlas of serous fluid cytopathology: Acta Cytol 1991;35:761-764.
A guide to cells of pleural, pericardial, peritoneal and hydro- 44. Magliocco A, Miller K, Robertson DI, Gill MJ: Occurrence of
cele fluids. In Gresham GA (ed): Current Histopathology Series. Pneumocystis carinii organisms in a peritoneal effusion from a
Dordrecht, The Netherlands, Kluwer Academic Publishers, patient with the acquired immunodeficiency syndrome. Diagn
1989, p. 14. Cytopathol 1991;7:540-542.
21. Nance KV, Shermer RW, Askin FB: Diagnostic efficacy of pleural 45. Monte SA, Ehya H, Lang WR: Positive effusion cytology as the
biopsy as compared with that of pleural fluid examination. initial presentation of malignancy. Acta Cytol 1987;31:448-452.
Mod Pathol 1991;4:320-324. 46. Sears D, Hajdu SI: The cytologic diagnosis of malignant
22. Garcia LW, Ducatman BS, Wang HH: The value of multiple fluid neoplasms in pleural and peritoneal effusions. Acta Cytol
specimens in the cytological diagnosis of malignancy. Mod 1987;31:85-97.
Pathol 1994;7:665-668. 47. Ringenberg QS, Doll DC, Loy TS, Yarbro JW: Malignant ascites
23. Johnston WW: The malignant pleural effusion: A review of unknown origin. Cancer 1989;64:753-755.
of cytopathologic diagnoses of 584 specimens from 472 48. Gushchin V, Demmy TL, Kane JM 3rd: Surgical management
consecutive patients. Cancer 1985;56:905-909. of metastatic peritoneal or pleural disease. Semin Oncol
24. Light RW: Clinical practice. Pleural effusion. N Engl J Med 2007;34(3):215-225.
2002;346(25):1971-1977. 49. Price BA, Ehya H, Lee JH: Significance of pericellular lacunae in
25. Kutty CPK, Remeniuk E, Varkey B: Malignant-appearing cells in cell blocks of effusions. Acta Cytol 1992;36:333-337.
pleural effusion due to pancreatitis: Case report and literature 50. Ehya H: The cytologic diagnosis of mesothelioma. Semin Diagn
review. Acta Cytol 1981;25:412-416. Pathol 1986;3(3):196-203.
26. Hsu C: Cytologic detection of malignancy in pleural effusion: A 51. Kho-Duffin J, Tao LC, Cramer H, et al.: Cytologic diagnosis
review of 5255 samples from 3811 patients. Diagn Cytopathol of malignant mesothelioma, with particular emphasis on
1987;3:8-13. the epithelial noncohesive cell type. Diagn Cytopathol
27. Hallman JR, Geisinger KR: Cytology of fluids from pleural, 1999;20(2):57-62.
peritoneal and pericardial cavities in children: A comprehensive 52. Sherman ME, Mark EJ: Effusion cytology in the diagnosis of
survey. Acta Cytol 1994;38:209-217. malignant epithelioid and biphasic pleural mesothelioma.
28. Esteban JM, Yokota S, Husain S, Battifora H: Arch Pathol Lab Med 1990;114:845-851.
Immunocytochemical profile of benign and carcinomatous 53. Saad RS, Cho P, Liu YL, Silverman JF: The value of epithelial
effusions. Am J Clin Pathol 1990;94:698-705. membrane antigen expression in separating benign mesothelial
29. Zakowski M, Ianuela-Shanerman A: Cytology of pericardial proliferation from malignant mesothelioma: A comparative
effusions in AIDS patients. Diagn Cytopathol 1993;9:266-269. study. Diagn Cytopathol 2005;32(3):156-159.
30. Churg A, Cagle PT, Roggli VL (eds): Tumors of the Serosal 54. Dejmek A, Hjerpe A: Reactivity of six antibodies in effusions of
Membranes. Silver Spring, Md, ARP Press, 2006. mesothelioma, adenocarcinoma and mesotheliosis stepwise
31. Choi YL, Song SY: Cytologic clue of so-called nodular logistic regression analysis. Cytopathology 2000;11(1):8-17.
histiocytic hyperplasia of the pleura. Diagn Cytopathol 55. Cury PM, Butcher DN, Corrin B, Nicholson AG: The use of
2001;24(4):256-259. histological and immunohistochemical markers to distinguish
32. Drury M, Anderson W, Heffner JE: Diagnostic value of pleural malignant mesothelioma and in situ mesothelioma
pleural fluid cytology in occult Boerhaave’s syndrome. Chest from reactive mesothelial hyperplasia and reactive pleural
1992;102:976-978. fibrosis. J Pathol 1999;189(2):251-257.
references 153

56. Aerts JG, Delahaye M, van der Kwast TH, et al.: The high 69. Zhang PJ, Livolsi VA, Brooks JJ: Malignant epithelioid vascular
post-test probability of a cytological examination renders tumors of the pleura: Report of a series and literature review.
further investigations to establish a diagnosis of epithelial Hum Pathol 2000;31:29-34.
malignant pleural mesothelioma redundant. Diagn Cytopathol 70. Jaffe ES, Harris NL, Stein M, Vardiman JW (eds): World Health
2006;34(8):523-527. Organization Classification of Tumours. Pathology and
57. Roberts F, Harper CM, Downie I, Burnett RA: Immunohisto­ Genetics of Tumours and Haematopoietic and Lymphoid
chemical analysis still has a limited role in the diagnosis of Tissues. Lyon, France, IARC Press, 2001.
malignant mesothelioma. A study of thirteen antibodies. Am J 71. Brimo F, Michel RP, Khetani K, Auger M: Primary effusion
Clin Pathol 2001;116(2):253-262. lymphoma a series of 4 cases and review of the literature with
58. Attanoos RL, Griffin A, Gibbs AR: The use of immunohisto- emphasis on cytomorphologic and immunocytochemical
chemistry in distinguishing reactive from neoplastic mesothe- differential diagnosis. Cancer 2007;111(4):224-233.
lium. A novel use for desmin and comparative evaluation with 72. Wakely PE Jr, Menezes G, Nuovo GJ: Primary effusion lymphoma:
epithelial membrane antigen, p53, platelet-derived growth cytopathologic diagnosis using in situ molecular genetic analy-
factor-receptor, P-glycoprotein and Bcl-2. Histopathology sis for human herpesvirus 8. Mod Pathol 2002;15(9):944-950.
2003;43(3):231-238. 73. Ansari MQ, Dawson DB, Nador R, et al.: Primary body
59. Xiao S, Renshaw A, Cibas ES, et al.: Novel fluorescence in situ cavity-based AIDS-related lymphomas. Am J Clin Pathol
hybridization approaches in solid tumors: Characterization 1996;105:221-229.
of frozen specimens, touch preparations, and cytological 74. Banks PM, Warnke RA: Primary effusion lymphoma. In Jaffe
preparations. Am J Pathol 1995;147:896-904. ES, Harris NL, Stein M, Vardiman JW (eds): World Health
60. Chhieng DC, Yee H, Schaefer D, et al.: Calretinin staining Organization Classification of Tumours Pathology and Genetics
pattern aids in the differentiation of mesothelioma from of Tumours of Haematopoietic and Lymphoid Tissues. Lyon,
adenocarcinoma in serous effusions. Cancer 2000;90:194-200. France, IARC Press, 2001, pp. 179-180.
61. Wieczorek TJ, Krane JF: Diagnostic utility of calretinin 75. Ohori NP, Whisnant RE, Nalesnik MA, Swerdlow SH: Primary
immunohistochemistry in cytologic cell block preparations. pleural effusion posttransplant lymphoproliferative disorder:
Cancer 2000;90:312-319. Distinction from secondary involvement and effusion
62. Hecht JL, Lee BH, Pinkus JL, Pinkus GS: The value of Wilms lymphoma. Diagn Cytopathol 2001;25(1):50-53.
tumor susceptibility gene 1 in cytologic preparations as a 76. Renshaw AA, Nappi D, Cibas ES: Cytology of metastatic
marker for malignant mesothelioma. Cancer 2002;96:105-109. adenocarcinoma of the prostate in pleural effusions. Diagn
63. Ordonez NG: Value of thyroid transcription factor-1, E-cad- Cytopathol 1996;15(2):103-107.
herin, BG8, WT1, and CD44S immunostaining in distinguishing 77. Parwani AV, Chan TY, Ali SZ: Significance of psammoma
epithelial pleural mesothelioma from pulmonary and non-pul- bodies in serous cavity fluid: A cytopathologic analysis. Cancer
monary adenocarcinoma. Am J Surg Pathol 2000;24:598-606. 2004;102(2):87-91.
64. Cibas ES, Corson JM, Pinkus GS: The distinction of 78. Smith-Purslow MJ, Kini SR, Naylor B: Cells of squamous cell
adenocarcinoma from malignant mesothelioma in cell carcinoma in pleural, peritoneal, and pericardial fluids: Origin
blocks of effusions: he role of routine mucin histochemistry and morphology. Acta Cytol 1989;33:245-253.
and immunohistochemical assessment of carcinoembryonic 79. Chhieng DC, Ko EC, Yee HT, et al.: Malignant pleural
antigen, keratin proteins, epithelial membrane antigen, effusions due to small-cell lung carcinoma: A cytologic
and milk fat globule-derived antigen. Hum Pathol and immunocytochemical study. Diagn Cytopathol 2001;
1987;18:67-74. 25(6):356-360.
65. Frisman DM, McCarthy WF, Schlieff P, et al.: Immunocyto­ 80. Walts AE: Malignant melanoma in effusions: A source of false-
chemistry in the differential diagnosis of effusions: use of logis- negative cytodiagnoses. Diagn Cytopathol 1986;2:150-153.
tic regression to select a panel of antibodies to distinguish 81. Das DK: Serous effusions in malignant lymphomas: a review.
adenocarcinomas from mesothelial proliferations. Mod Pathol Diagn Cytopathol 2006;34(5):335-347.
1993;6:179-184. 82. Walts AE, Marchevsky AM: Computerized interactive
66. Ordonez NG: What are the current best immunohistochemical morphometry: An expert system for the diagnosis of lymphoid-
markers for the diagnosis of epithelioid mesothelioma? rich effusions. Am J Clin Pathol 1989;92:765-772.
A review and update. Hum Pathol 2007;38(1):1-16. 83. Billingham ME, Rawlinson DG, Berry PF, Kempson RL:
67. Hecht JL, Pinkus JL, Weinstein LJ, Pinkus GS: The value of The cytodiagnosis of malignant lymphomas and Hodgkin’s
thyroid transcription factor-1 in cytologic preparations as a disease in cerebrospinal, pleural, and ascitic fluids. Acta Cytol
marker for metastatic adenocarcinoma of lung origin. Am J 1975;19:547-556.
Clin Pathol 2001;116:483-488. 84. Sasser RL, Yam YT, Li CY: Myeloma with involvement of the
68. MacDougall DB, Wang SE, Zidar BL: Mucin-positive epithelial serous cavities: Cytologic and immunochemical diagnosis and
mesothelioma. Arch Pathol Lab Med 1992;116:874-880. literature review. Acta Cytol 1990;34:479-485.

You might also like