You are on page 1of 111

Mostafa M.

Fraig
Editor

Diagnosis of
Small Lung Biopsy

An Integrated Approach

123
Diagnosis of Small Lung Biopsy
Mostafa M. Fraig
Editor

Diagnosis of Small
Lung Biopsy
An Integrated Approach
Editor
Mostafa M. Fraig, MD
William M. Christopherson Professor of Pathology
and Internal Medicine
Department of Pathology and Laboratory Medicine
School of Medicine, University of Louisville
Louisville, KY, USA

ISBN 978-1-4939-2574-2 ISBN 978-1-4939-2575-9 (eBook)


DOI 10.1007/978-1-4939-2575-9

Library of Congress Control Number: 2015936666

Springer New York Heidelberg Dordrecht London


© Springer Science+Business Media New York 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made.

Printed on acid-free paper

Springer Science+Business Media LLC New York is part of Springer Science+Business Media
(www.springer.com)
To my wife, Lamia, and our two lovely sons, Yousef and Karim,
who provided love and care that were the inspiration for this
endeavor, for them I am eternally indebted.
Preface

No doubt, there are several excellent pulmonary pathology textbooks with


comprehensive and encyclopedic coverage of the topic in the market.
However, we sensed a void when it comes to more pressing issue; when it
comes to small biopsies, what the general pathologist deal with in their daily
practice is more nuanced and different from what we can appreciate on a
large resection specimen. In the current era, when we as pathologists are
asked to do more with less, it becomes imperative that we adjust our perspec-
tive and sharpen our tools.
Most cases of lung lesions are biopsied with tiny biopsies from fine needle
aspiration, needle cores, transbronchial biopsy, and more recently cryobiop-
sies. In all these instances, the tissue is scant and the artifacts abound. Yet the
information expected to be gleaned from such biopsy is remarkable consider-
ing the size and the hindrances. There is a void in the library for a book to
provide a concise and synoptic approach to the interpretation of this type of
biopsies. This book is an attempt to provide this type of approach. It is to be
as beside the microscope quick reference for the general pathologist and
pathologist in training to get a quick understanding of basic pulmonary
pathology.
The authors are attempting to provide practical tips and tried methods for
the interpretation of these biopsies based on interaction with radiologists and
pulmonologists, medical oncologists, and other multidisciplinary team mem-
bers over the years. The reduction in the amount of tissue has to be compen-
sated for by the provision of more clinical and imaging information to
complete the picture. It is a difficult task in the busy shuffle of daily practice
to expect such information on regular basis. However, current billing practice
is requiring this information to be included on the requisition form. The intro-
duction and promotion of Electronic Health Records (HER) is providing an
easy access to such information to the average pathologist even in small com-
munity setting and have take to advantage of that.
Lung biopsies are expensive and difficult to obtain and every effort should
be made to maximize the benefit and information gained from them. This
includes talking to the physician taking care of the patient and discussing the
history and clinical suspicion.
The integrated approach is nothing new and every book tries to cover the
clinical and radiologic findings in the discussion of a given disease in the lung.

vii
viii Preface

In this book, we try to reiterate and emphasize the importance of those findings
in the proper interpretation of those small biopsies. Descriptive diagnoses can
be useless and even downright harmful when they are rendered in isolation and
without an understanding of the clinical context.
We hope this book can serve its intended purpose and be a modest contri-
bution to the enrichment of pulmonary pathology practice.

Louisville, KY, USA Mostafa M. Fraig, M.D.


Contents

1 Small Sample Lung Biopsy Techniques in the Diagnosis


of Airway and Parenchymal Lung Diseases ................................ 1
Rafael L. Perez
2 Principles of Imaging Lung Disease ............................................. 13
James G. Ravenel
3 Types of Biopsies ............................................................................ 21
Mostafa M. Fraig
4 Airspace-Occupying Diseases ....................................................... 29
Mostafa M. Fraig
5 Interstitial Lung Diseases in Small Lung Biopsies ...................... 39
Mostafa M. Fraig
6 Diagnosis of Granulomatous Disease
and Vasculitis in Small Lung Biopsies.......................................... 51
Sanjay Mukhopadhyay
7 Benign Tumors of the Lung in Small Lung Biopsies .................. 81
Mostafa M. Fraig
8 Malignant Tumors of the Lung in Small Lung Biopsies............. 91
Mostafa M. Fraig

Index ...................................................................................................... 101

ix
Contributors

Mostafa M. Fraig Department of Pathology and Laboratory Medicine,


School of Medicine, University of Louisville, Louisville, KY, USA
Sanjay Mukhopadhyay Department of Anatomic Pathology, Cleveland Clinic,
Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland,
OH, USA
Rafael L. Perez Department of Medicine, University of Louisville School
of Medicine, Louisville, KY, USA
James G. Ravenel Department of Radiology and Radiologic Services,
Medical University of South Carolina, Charleston, SC, USA

xi
Small Sample Lung Biopsy
Techniques in the Diagnosis 1
of Airway and Parenchymal
Lung Diseases

Rafael L. Perez

we define small lung biopsy samples as tissue


Introduction obtained by nonsurgical techniques that are 1 cm
or less in the longest axis.
Nonsurgical approaches to sample focal or diffuse, Small lung biopsy samples are obtained by
solid, or infiltrative pathology of the lung offer the various techniques. Flexible fiberoptic bronchos-
advantages of lower risk, comfort, and cost com- copy (FOB) with endobronchial and transbron-
pared to surgical biopsy techniques. However, chial biopsy with forceps is a standard procedure
sample size obtained by nonsurgical techniques performed by all pulmonologists, while endo-
can challenge pathological assessment. Sampling bronchial biopsy via rigid scope, cryobiopsy, and
error and artifacts of manipulation, in addition to navigational bronchoscopy biopsy require addi-
small size, are problematic. These problems may tional training and experience. Transthoracic core
be overcome to some extent by providing multiple biopsy is a radiological procedure. All of these
intact samples that are well directed by various procedures have in common that they can be per-
imaging techniques. Ultimately, a collaborative formed outside the operating room.
approach between the clinician, pathologist, and
radiologist will optimize diagnosis.
This chapter will focus on the nonsurgical Small Sample Lung Biopsy
techniques used to obtain small samples of pul- Techniques by Fiberoptic
monary tissue characterized by location of the Bronchoscopy
pathological tissue in the airways and lung paren-
chyma. How the pathological diagnosis informs In 1966, Dr. Shigeto Ikeda introduced the tech-
the clinician’s assessment and approach to the nique of flexible fiberoptics to access and visually
case in question will demonstrate the importance inspect the lungs [1]. The advantages of flexible
of an accurate diagnosis. Most small tissue sam- FOB as a nonsurgical procedure quickly brought it
ples obtained by nonsurgical approaches are in into widespread use. It could be performed in
the sub-centimeter size range, but can reach much less complicated settings than the operating
1 cm depending on the technique used. Therefore, room, and moderate sedation with topical anesthe-
sia could be used in most cases. Elective FOB is a
routine outpatient procedure where various
R.L. Perez, M.D. (*) applications to sample bronchial and parenchymal
Department of Medicine, University of Louisville
School of Medicine, 530 South Jackson Street,
tissues in stable non-acute individuals can be
Louisville, KY 40202, USA obtained. FOB is easily transportable to the oper-
e-mail: rafael.perez@louisville.edu ating room and to the acute care setting where it

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 1


DOI 10.1007/978-1-4939-2575-9_1, © Springer Science+Business Media New York 2015
2 R.L. Perez

has an important diagnostic and therapeutic role in


the intensive care units. Endobronchial Diseases Diagnosed
The bronchoscope has been refined over time by Small Sample Lung Biopsies
for a variety of applications [2–4]. Bronchoscopes
have working channels of 1.4–2.8 mm in diame- Sampling of the airways affords the lowest risk to
ter, the larger ones to allow the use of an array of benefit ratio involved in FOB. The abnormalities
interventional instruments. Preparation and per- are usually visible, so the diagnostic accuracy is
formance for FOB regardless of the type of pro- good. Complications, mostly bleeding, can be
cedure requires the same key considerations. directly controlled with topical vasoconstrictors
Foremost is patient safety. Many who come to or tamponade. Forceps sometimes accompanied
bronchoscopy already have underlying lung dis- by bronchial brushings and washings are used to
ease that affects ventilation and oxygenation. take multiple biopsy samples of the lesion. There
Benzodiazepines and opioids generally used for are no specific guidelines for the number of endo-
sedation and comfort may degrade both critically. bronchial biopsies that needs to be taken to maxi-
A clinical professional, usually a trained nurse, mize diagnostic yield. Four to five samples are
monitors ventilation, oximetry, blood pressure, common, but the aware bronchoscopist may
and other vital signs continuously during the pro- obtain greater numbers since the samples are
cedure. Reversal agents are on hand, and the small, and sufficient tissue for special stains or
equipment and drugs are within reach to adminis- biomarkers may be required. Table 1.1 lists the
ter cardiopulmonary resuscitation, if necessary. spectrum of diseases diagnosed by sampling of
A current history and physical is required before the airways.
the procedure with emphasis on allergies, con-
comitant medications, and coagulation status.
The history and physical speaks not only to Endobronchial Tumors
patient safety but also to the reason why the indi-
vidual is having the procedure and the anticipated Endobronchial biopsies in adult patients are
samples to be obtained. It is imperative that the mostly done for the diagnosis of bronchogenic
appropriate media and fixatives are available and malignancies. Patients with bronchogenic malig-
ready to receive the samples. nancies come to biopsy with symptoms that are
Most diagnostic tissue sampling is done with generally not specific, overlap with other airway
the standard instrument that typically returns tis- diseases, and involve other organ systems as the
sue samples measuring only a few millimeters. disease progresses [5]. Wheezing, cough, and dys-
Therefore, a high degree of specificity is para- pnea are the most common symptoms in tumors of
mount to obtain an accurate diagnosis. The oper- the airways and are shared with diseases that cause
ating characteristics of the FOB also vary
depending on the location of the abnormal tissue
Table 1.1 Endobronchial diseases diagnosed by small
to be sampled. The diagnostic accuracy of a sample lung biopsies
directly visualized endobronchial lesion is better
Endobronchial tumors
than a lesion located in the lung parenchyma. • Malignant
Parenchymal lesions may be sampled under flu- • Carcinoid tumors
oroscopic guidance, but the more distal and • Hamartomas
smaller the lesion, the greater the chance of sam- Infectious and other endobronchial diseases
pling error. The accuracy of obtaining a diagnos- • Mycoplasma and viral
tic sample from deep in the lung parenchyma has • Endobronchial tuberculosis
increased greatly in the past few years using • Allergic bronchopulmonary aspergillosis
navigational bronchoscopy as described in a • Bronchocentric granulomatosis
subsequent section. • Sarcoidosis
1 Small Sample Lung Biopsy Techniques in the Diagnosis of Airway… 3

airflow obstruction, most notably chronic obstruc- features that separate them from malignant lesions.
tive pulmonary disease and asthma. The classic The histological diagnosis leads to a second thera-
paraneoplastic symptoms associated with some peutic bronchoscopy with the removal of the tumor
bronchogenic cancers that lend some diagnostic that is curative in most cases.
specificity are not common. Hypertrophic osteoar-
thropathy, a periosteal inflammatory condition of
the long bones, and hypercalcemia can be found in Infectious and Other Endobronchial
squamous cell cancer of the lung. The Lambert– Abnormalities
Eaton myasthenic syndrome with weakness and
visual disturbance is seen in small cell lung cancer. Diseases of the airways extend well beyond the
Bronchorrhea is occasionally associated with ade- malignant and benign tissue transformations
nocarcinoma in situ. Hemoptysis is occasionally described above. These diseases include infectious
the presenting symptom for bronchogenic cancer and noninfectious disorders that present with a
and portends more extensive involvement of the broad constellation of symptoms that may extend
airway and poorer outcome [6]. Pneumonia, beyond the respiratory system. The history and
especially if it is recurrent in the same location, presentation of the individual is more complex
may indicate an obstructing lesion of the airway. with questions about immune status and lung
Bronchoscopy may be a challenge in such cases function abnormalities at the forefront. Fever and
because of the inflammation with swelling and metabolic abnormalities also become more promi-
purulence that make it difficult to biopsy the nent. The path to bronchoscopy usually takes
lesion. The clinician may elect to postpone the longer and only after clinical, laboratory, and
procedure until the pneumonia is treated and well radiological evaluation has not yielded an answer.
into resolution. Fever and weight loss will put infection at the
Like their malignant counterparts, benign top of the disease differential, so tissue sampling
endobronchial tumors present with similar symp- for both histological examination and culture and
toms of airway obstruction with wheezing, staining is prepared. On inspection, infections of
cough, dyspnea, and sometimes with hemoptysis the airways can take any form such as single or
or post-obstructive pneumonia. Carcinoid tumors multiple nodules, ulcerated epithelium, discolored
are derived from neuroendocrine tissues of the patches, or even only mild erythema and edema.
gastrointestinal (GI) and respiratory tracts. They Bronchial washings and brushings typically
are predominantly found in the GI tract, but up to accompany tissue biopsies in these cases. Multiple
20 % occur in the airways [7, 8]. Compared to samples for culture are taken first to avoid any pos-
tumors in the GI tract, pulmonary carcinoids are sibility of contamination of the bronchoscope
less likely to produce the carcinoid syndromes of channel or forceps with fixative. While a positive
flushing, diarrhea, and heart failure. They can be culture of a non-commensal or opportunistic
found from the most proximal to the most distal organism is sufficient to initiate specific therapy,
conducting airways where neuroendocrine rests demonstration of tissue invasion may be necessary
are found. On bronchoscopy, they have a very in some circumstances.
smooth, shiny, and sometimes very vascular sur- Diffuse infections of the airways can cause
face in contrast to malignant tumors that have them to become hyperresponsive and produce
irregular lobulated borders with varying degrees symptoms that are indistinguishable from asthma.
of necrosis. Though asthma is very prevalent worldwide,
Hamartomas are the most common benign lung individual cases sometimes present out of context
tumors, but only 10 % of those occur in the bronchi with respect to risk factors, a history of atopy,
[9]. They usually come to attention in patients who exacerbating exposures, or response to therapy.
present with post-obstructive symptoms or by The clinician may therefore elect to perform
chance in an abnormal chest film. On broncho- inspection and biopsy of the airways. Mycoplasma
scopic inspection, they have no distinguishing pneumoniae has been associated with and even
4 R.L. Perez

implicated as the cause of asthma in some case granulomatous lesion [16, 17]. About one third
series, the latter conclusion on the basis that the of individuals with BCG have asthma, and three
asthma was controlled after treating the infection fourths of them will have A. fumigatus hyphae
[10]. Rhinovirus, respiratory syncytial virus, and detected in their bronchial biopsies. Of the two
influenza viruses have a strong relationship with thirds who do not have asthma, hyphae are pres-
asthma exacerbations and may be relevant in ent in only about one third. Radiographically,
chronic uncontrolled asthma that may lead one to BCG may appear as a lung mass that will prompt
obtain bronchial tissues for examination [11]. the bronchoscopic examination. Steroid treat-
The key point on reviewing bronchial biopsies of ment is very effective, but the disease may recur
subjects with reactive airways is that, in addition so individuals must be monitored regularly.
to the anticipated inflammatory changes, aware- Sarcoidosis is a multisystemic granulomatous
ness and detection of concurrent infection will disease of unknown etiology with a variety of
aid the clinician with treatment. clinical presentations depending on the organ
Endobronchial tuberculosis is an uncommon involved [18]. Respiratory symptoms are nonspe-
manifestation of infection with Mycobacterium cific and include chest tightness, cough, and dys-
tuberculosis (MTB) [12, 13]. Cough, wheezing, pnea on exertion. A high proportion of patients
fever, and dyspnea are nonspecific symptoms, and with pulmonary sarcoidosis will have some evi-
it is not usually detectable on plain chest radiogra- dence of airflow obstruction suggesting endo-
phy. Moreover, sputum cultures for MTB may be bronchial involvement with this disease [19].
negative. On bronchoscopy, the lesions have no On bronchoscopy, the bronchial mucosa may
distinguishing features that would make the oper- appear normal to erythematous with “cobbleston-
ator suspect MTB. The lesions may appear as ing” that indicates a heavy granuloma burden in
endobronchial masses or ulcerations. They may the airways. The yield of random biopsies of the
obstruct airways or cause tracheal or bronchial bronchial mucosa depends on the degree of
stenosis. Expedient identification and medical involvement and in practice is a supplement to
treatment of endobronchial MTB are therefore transbronchial biopsies and transbronchial nee-
essential to prevent fixed stenosis and need for dle aspiration of enlarged hilar, paratracheal, or
surgical reconstruction. other mediastinal nodes.
Allergic bronchopulmonary aspergillosis
(ABPA) and bronchocentric granulomatosis
(BCG) are entities that overlap with asthma in Parenchymal Diseases Diagnosed
their clinical presentation [14, 15]. ABPA is by Small Sample Lung Biopsies
mostly associated with Aspergillus fumigatus,
but other fungi less commonly produce a similar New technologies have extended the reach and
syndrome. In addition to the symptoms and accuracy of small sample biopsies of solid and
examination findings of asthma, the presence of infiltrative diseases in the lung parenchyma.
mucus plugging or infiltrates may lead to bron- Table 1.2 lists parenchymal diseases categorized
choscopic examination and biopsy of the involved by radiological appearance. This categorization is
airways. The hallmark demonstration of fungal useful for differential diagnosis and the selection
hyphae in bronchial tissue, along with supportive of the biopsy technique and approach to diagnosis.
findings of elevated IgE and cutaneous reactivity Unlike endobronchial diseases, imaging using
or precipitins to A. fumigatus, significantly fluoroscopy and ultrasound assists in the localiza-
impacts therapy. In ABPA, systemic steroids are tion and biopsy of lesions in the lung parenchyma.
required in addition to bronchodilator therapy. In New tools for performing navigational bronchos-
recalcitrant cases, treatment with antifungals copy, linear and radial endobronchial ultrasonog-
may be necessary. raphy, and cryobiopsy have extended the reach and
BCG was first described in 1972 by Liebow as accuracy of bronchoscopic biopsy of even the
a focal bronchial and bronchiolar destructive deepest pulmonary abnormalities.
1 Small Sample Lung Biopsy Techniques in the Diagnosis of Airway… 5

Table 1.2 Parenchymal diseases diagnosed by small must be recovered for possible biomarker probing
sample lung biopsies
that may influence therapy.
Focal solid parenchymal lesions Nonmalignant lung masses can sometimes be
• Lung masses seen in chronic inflammatory and granulomatous
• Metastatic disease diseases produced by infection, as pulmonary
• Solitary pulmonary nodules involvement in autoimmune diseases, or through
Focal and diffuse infiltrative diseases
environmental exposures. Though mostly pre-
• Granulomatous diseases
senting as lung nodules, fungal infections may
– Sarcoidosis
sometimes present radiographically as lung
– Hypersensitivity pneumonitis
masses [24]. Histoplasmosis or blastomycosis
• Non-granulomatous diseases
– Idiopathic pulmonary fibrosis
may present as a mass-like consolidations in
– Nonspecific interstitial pneumonitis immunocompetent patients, while cryptococcosis
– Smoking-related interstitial lung diseases is found mainly in immunosuppressed individu-
– Organizing pneumonia als. Similar to malignant masses, the diagnostic
approach is by transbronchial biopsy supple-
mented by bronchial washings with saline for
culture.
Focal Solid Parenchymal Lesions Mass-like lung densities may also be seen in
autoimmune diseases like sarcoidosis, rheuma-
Standard flexible fiberoptic biopsy guided by toid arthritis, and granulomatosis with polyangi-
fluoroscopy is the mainstay procedure for locat- itis. When lung masses are detected in the context
ing and sampling solid parenchymal lesions. of an autoimmune disease, the individual is often
Fluoroscopy is also used for safety purposes on immunosuppressive therapy. Opportunistic
mainly to mitigate and assess for pneumothorax. infection must be considered, and appropriate
Lung masses that are larger and centrally located preparation to receive samples for Pneumocystis
are usually approached in this way. Lesions that jirovecii, other fungi, and acid-fast organisms
are smaller and located peripherally are more should be done. Often, bronchoalveolar lavage
accurately biopsied using navigational or (BAL) supplements biopsy by accessing deep
ultrasound-guided bronchoscopy as described respiratory surfaces not obtained by simple wash-
below [20, 21]. ings. BAL involves wedging the bronchoscope
Lung masses by definition are discrete lesions into a fourth- to sixth-generation bronchus to pro-
greater than 3 cm in diameter by radiography duce a tight seal. Then, sterile saline is instilled in
[22]. A single mass in an adult is considered to be 30–50 mL. Aliquots are recovered by gentle suc-
malignant until proven otherwise. Age, smoking tion. An acceptable BAL sample will demonstrate
history, and presence of chronic obstructive pul- a “foamy” meniscus in the suction receptacle
monary disease all increase the pretest probabil- indicating recovery of pulmonary surfactant from
ity of primary lung cancer [23]. Therefore, it is alveolar surfaces and return few to no ciliated
imperative that the bronchoscopic sample dem- epithelial cells indicating little contamination
onstrates malignancy to avoid a more invasive from the conducting airways.
biopsy approach. Radio- or chemotherapy is very Occupational exposures to silica, coal dust,
rarely undertaken without a true-positive histo- beryllium, and kaolin will produce one or more
logical evidence of malignancy. The bronchosco- discrete lesions that must be differentiated from
pist maximizes diagnostic yield using fluoroscopy malignancy. The occupational history must be pro-
to locate the mass and take multiple transbron- vided to the reviewing pathologist to prepare the
chial biopsies using a forceps “punched” into the appropriate microscopic assessment for the pres-
lung parenchyma. Angling the tip of the broncho- ence of particulates. Beryllium is indistinguish-
scope slightly with each pass is done to sample able from sarcoidosis histologically, but the lack of
different regions of the mass. Finally, ample tissue certain extrapulmonary manifestations found in
6 R.L. Perez

sarcoidosis and the exposure history will increase potential, determine to a large degree how the
the confidence that the noncaseating granulomas nodule is to be approached. Sub-centimeter
are the result of beryllium exposure [25]. nodules may be observed by serial radiology and
Metastatic malignancies to the lungs may applying various algorithms based on risk, smok-
present as solitary or multiple lesions of varying ing history, and lung function [29]. Nodules that
size. Colorectal cancers occasionally present as are one or more centimeters in diameter may be
solitary nodules. Identifying these lesions as origi- resected or sampled for diagnosis. Risk factors
nating from these organs is key since resection for malignancy, surgical risks, and patient desires
may improve survival [26]. In the setting of known determine the approach.
cancer, patients with solitary metastases may go The advent of electromagnetic guidance has
directly to surgery after risk assessment. However, improved the diagnostic reach and accuracy of
if diagnosis is elected, then navigational bronchos- bronchoscopy in the assessment of pulmonary
copy, described in more detail below, can accu- nodules by a technique called navigational bron-
rately sample even peripheral metastases 1 cm in choscopy. Navigational bronchoscopy is a com-
diameter or more. plicated procedure that matches the location of a
Lymphangitic metastases are typically subject’s lesion on computerized tomography to
addressed with standard transbronchial biopsy the anatomy of the subject who is enveloped in a
with fluoroscopic guidance. Five to six passes in magnetic field during the procedure [30]. A pro-
one selected lung lobe are done and yield an cess called “registration” matches the image and
almost 70 % chance of diagnosis [27]. Although magnetic field. Then, a probe with an extended
the risk of pneumothorax is low, the bronchosco- working channel is inserted through the broncho-
pist never performs transbronchial biopsy in both scope and guided to the lesion through a virtual
lungs in the same procedure. image on screen. Once the lesion is located, the
The approach to the diagnosis of solitary or extended channel is locked in place. A biopsy
multiple pulmonary nodules begins with a thor- forceps is inserted to sample the lesion under
ough interview of the patient emphasizing respi- fluoroscopic guidance. The diagnostic yield of
ratory exposures and risk assessment. Then, the navigational bronchoscopy alone is about 60 %,
decision to observe over time, perform biopsy, or and when combined with radial endobronchial
go directly to resection can be made. The advent ultrasound to confirm location, the yield
of low-dose computerized tomography (LDCT) approaches 90 % [31].
and comfort of practitioners to order this test
have produced an upsurge of studies demonstrat-
ing pulmonary nodules. Indeed, in the National Focal and Diffuse Infiltrative Diseases
Lung Screening Trial in the United States in
which over 53,000 individuals at risk for lung Infiltrative diseases of the lung prove challenging
cancer underwent LDCT, a reduction in lung when diagnosis is attempted using small sample
cancer mortality and discovery of earlier stage biopsy techniques. Routine transbronchial lung
disease were demonstrated [28]. Still, the vast biopsy may be attempted in settings where there
majority of nodules assessed were benign is clear expertise by both the clinical and patho-
through observation over time or by diagnostic logical personnel involved in making the diagno-
sampling or by resection. Biopsy and resection sis. In general, granulomatous lung diseases like
incur both cost and risks including mortality. sarcoidosis and hypersensitivity pneumonitis are
The challenge is the decision to wait and watch more likely to be diagnosed by transbronchial
biopsy or resect. lung biopsy [32] than the non-granulomatous
Solitary pulmonary nodules are by definition idiopathic interstitial pneumonias (IIPs) including
single nodular lesions of 3-cm diameter or less idiopathic pulmonary fibrosis [33, 34]. Surgical
surrounded by normal lung tissue [22]. Location lung biopsy is recommended for the latter despite
and size, along with risk factors for malignant the attendant risks of anesthesia and operation
1 Small Sample Lung Biopsy Techniques in the Diagnosis of Airway… 7

in patients who may already have significant pulmonary infiltrates in the sensitized individual.
pulmonary impairment. The symptoms resolve when the antigen is
Sarcoidosis is a granulomatous multisystemic removed and recur on exposure to the antigen.
disease with unknown etiology [18]. The most The predominant immune response in acute
commonly involved organ is the lung, and it is disease is the Arthus, or type III hypersensitivity
thought that the causative agent or agents gain response, involving immune complexes. The
access via the respiratory route. The respiratory more subtle subacute presentation with cough,
symptoms are nonspecific ranging from general- dyspnea, and fatigue displays centrilobular infil-
ized chest discomfort, or tightness, to cough and trates by computerized tomography. Subacute HP
wheezing. In many cases, the disease is suspected represents a transition to cell-mediated immu-
on the basis of accompanying symptoms such as nity generating the granulomatous response that
dry mouth and eyes (Sjogren’s syndrome), pain characterizes this disease. Continued exposure
in the large joints, or even unexplained weight to the antigen may result in chronic HP of which
loss. Acute disease may present with painful iritis the hallmark is the development of pulmonary
or uveitis and occasionally erythema nodosum of fibrosis. The removal of the individual from the
the lower extremities. The classic noncaseating exposure is paramount to prevent disease pro-
granulomas are not found in the joints or nodo- gression. Therapy with corticosteroids is effec-
sum lesions, but in the more chronic keloid-like tive, but should not supplant avoidance of the
lesions that can be found on any region of the causative antigen.
body. The diagnosis can be reliably made by skin Like sarcoidosis, the granulomatous phase of
biopsy alone in the context of the presenting ill- HP is readily obtainable by transbronchial biopsy.
ness and exclusion of mycobacterial or fungal The site of biopsy is guided by radiological appear-
infection or lymphatic malignancies. ance. The disease tends to be concentrated in the
As noted in a previous section, pulmonary sar- upper lobes, and this is where multiple passes with
coidosis often involves the airways and can be the biopsy forceps are taken. Although adenopathy
sampled by endobronchial biopsy. Parenchymal is not a prominent feature of HP, this disease may
disease usually presents with several components need to be differentiated from sarcoidosis in some
that include mediastinal and hilar adenopathy cases. In this situation, some clinicians elect to
with or without pulmonary infiltrates. perform BAL with cell differential and flow
Occasionally, sarcoidosis may present with mul- cytometry to assess the ratio of CD4 to CD8 lym-
tiple pulmonary nodules or as a lung mass. phocytes. Both diseases present with a lympho-
Finally, pulmonary impairment may be present cytic alveolitis, but sarcoidosis has an elevated
with no radiological evidence of disease in the CD4/CD8, usually greater than 3, while HP pres-
lungs. In all of these presentations, standard ents with an inverted ratio of less than 1.
transbronchial lung biopsy has a very good diag- While standard bronchoscopy with transbron-
nostic yield of up to 90 % when 4–5 transbron- chial biopsy is sufficient to diagnose diffuse gran-
chial biopsies are performed [32]. In cases where ulomatous diseases, the diagnostic accuracy drops
there is obvious adenopathy abutting the airways, significantly in diffuse non-granulomatous lung
endobronchial ultrasound-guided fine-needle diseases [33, 34]. A new bronchoscopic biopsy
aspiration is becoming the preferred way to make technique born out of endobronchial cryotherapy
the diagnosis because of its high yield and better may enhance the diagnostic accuracy of the trans-
safety profile compared to transbronchial lung bronchial approach to diagnosis in diffuse lung
biopsy [35, 36]. diseases. Cryobiopsy uses a super cold probe to
Hypersensitivity pneumonitis (HP), or extrin- obtain parenchymal lung tissue of up to 1 cm in
sic allergic alveolitis, is a granulomatous lung diameter [39]. The probe is inserted through the
disease caused by a host of environmental anti- bronchoscope into the region of interest. The
gens [37, 38]. The acute form presents with tip is cooled to −89 °C by rapidly expanding
fever, cough, dyspnea, malaise, and diffuse nitrous oxide gas. Within seconds, the probe and
8 R.L. Perez

bronchoscope are removed as a unit to recover UIP, possible UIP, and inconsistent UIP. Lung
the tissue for fixation. Bleeding does not appear to biopsy is recommended in the latter two designa-
be increased by cryobiopsy compared to standard tions, surgical risks permitting [42].
forceps biopsy, but the risk of pneumothorax may Nonspecific interstitial pneumonia, or NSIP,
be increased by this technique. Nevertheless, the presents with similar clinical symptoms and time
advantage of cryobiopsy is that it recovers tissue course as IPF but differs greatly in its pathogene-
that has three times the sectional area of transbron- sis and response to therapy [43]. There is much
chial biopsies and avoids the crush artifact com- overlap on HRCT findings between NSIP and
monly seen in the latter procedure. This technique IPF, but NSIP tends to spare the subpleural paren-
will likely find application in the focal and diffuse chyma, and honeycombing is uncommon [44].
parenchymal lung diseases described below. Ground-glass opacification is more common in
The IIPs, autoimmune-related interstitial lung NSIP, but just over one half of the cases may not
diseases, and the more recently recognized group have this feature. NSIP is found in a wide variety
of smoking-related interstitial lung diseases [40] of diseases that result from alterations of the
require ample tissue to make a confident diagno- immune response such as the connective tissue
sis. Sufficient tissue is usually obtained by surgi- diseases, infections including HIV, and drug
cal lung biopsy, typically by video-assisted reactions. However, the UIP pattern can be found
thoracoscopy, or VATS. Aside from the complex- in these diseases as well, most often in rheuma-
ity and risks of general anesthesia and surgery, toid arthritis-associated interstitial lung disease.
selection of biopsy sites by VATS or open lung A proportion of individuals have the idiopathic
biopsy is critical. The tissue is preferably sam- form of NSIP that occurs when no causative etiol-
pled in transitional areas of varying degrees of ogy can be discerned after a thorough search. In
disease activity to enhance pathological descrip- effect, there are no current clinical or radiological
tion and diagnostic accuracy. Surgical experience findings that can confidently separate IPF/UIP
and collaboration between the surgeon, clinician, from NSIP. Differentiating NSIP from IPF is criti-
and radiologist are needed to acquire optimal cal because NSIP has a variable, but significant,
tissue for sectioning. response to immunomodulating therapies [43].
Idiopathic pulmonary fibrosis, or IPF, is the Lung biopsy is necessary for patients who present
most common of the IIPs [41]. It has a highly with the idiopathic form of NSIP and may be con-
variable disease progression with random exacer- sidered when patients who have a known underly-
bations and is uniformly fatal. Dyspnea on exer- ing cause do not respond to therapy. In these
tion, with profound exercise hypoxemia, and cases, the underlying histopathology may have
sometimes violent cough paroxysms are the pro- prominent areas of UIP intermingled with the
gressive symptoms. There is no approved drug NSIP. Surgical lung biopsy is the accepted method
therapy in the United States as of this writing. to obtain sufficient tissue for examination.
The disease at one time required the pathological The role of transbronchial cryobiopsy is yet to be
finding of usual interstitial pneumonitis (UIP) on determined.
lung biopsy, but the findings of lower lobe pre- The relationship between smoking and cer-
dominant peripheral reticular infiltrates, traction tain diffuse lung diseases has been recognized
bronchiectasis, honeycombing, and lack of over the past 50 years. Virtually, all smokers
“ground-glass opacification” on high-resolution develop respiratory bronchiolitis (RB) that can
computed tomography (HRCT) are now accepted be detected histologically [45, 46]. In general,
for the diagnosis of IPF [42]. Lung biopsy is not RB is not symptomatic, and lung function mea-
required when other causes of diffuse lung sures are normal. A minority of individuals will
disease are excluded and these key findings are progress with dyspnea and cough as they develop
displayed on HRCT. A recent update to the clas- the diffuse interstitial phase termed RB-ILD.
sification of IPF defines HRCT findings in IPF as Pulmonary function testing will demonstrate a
1 Small Sample Lung Biopsy Techniques in the Diagnosis of Airway… 9

mixed restrictive and obstructive pattern with


decreased total lung capacity and relative Conclusion
increase in residual volume indicating air trap-
ping. Radiologically, a patchy ground-glass The development of pulmonary diagnostic proce-
appearance may be noted. Desquamative inter- dures founded on FOB has allowed access to vir-
stitial pneumonitis (DIP), more accurately an tually every region of the airways and lung
influx of macrophages filling alveolar spaces, parenchyma avoiding the complexity, risks, and
presents as a uniformly restrictive impairment costs of surgery. These procedures are transport-
and diffuse confluent ground-glass opacification able to settings outside the operating room into
on CT scanning. The bronchiolocentric localiza- specialized outpatient medical procedure units
tion of inflammatory cells of RB-ILD and alveo- and inpatient acute care wards including inten-
lar filling presentation of DIP can be found sive care units. The main drawback to these non-
together suggesting that they represent a spec- surgical approaches is the limitation on the size
trum of smoking-related inflammatory reaction of the biopsy samples that are typically in the
extending from respiratory bronchioles to the sub-centimeter size range. Selection of sampling
alveolar spaces. Treatment of the smoking- technique, selection of the biopsy site, and pro-
related ILD mandates tobacco cessation some- curement of multiple samples overcome this
times combined with corticosteroids. The disadvantage to some extent. Most importantly,
distribution of disease is such that surgical lung the multidisciplinary collaboration between the
biopsy is required to appreciate the extent and pathologist, radiologist, and clinician will yield
pattern of DIP and RB-ILD. the highest diagnostic “wisdom.”
Organizing pneumonia filling the alveolar
spaces and respiratory bronchioles has a variable
focal distribution that typically involves both References
lungs. The primary form termed cryptogenic
organizing pneumonia, or COP, is found most 1. Ikeda S, Yanai N, Ishikawa S. Flexible bronchofiber-
commonly in the sixth decade of life [47]. Unlike scope. Keio J Med. 1968;17(1):1–16. PubMed PMID:
most of the other IIPs, it has a very favorable 5674435. Epub 1968/03/01. eng.
2. Bolliger CT, Mathur PN, Beamis JF, Becker HD,
response to corticosteroid therapy. Secondary
Cavaliere S, Colt H, et al. ERS/ATS statement on
organizing pneumonia can be found as a repara- interventional pulmonology. European Respiratory
tive reaction to many forms of lung injury includ- Society/American Thoracic Society. Eur Respir J.
ing infection, autoimmune diseases, and 2002;19(2):356–73. PubMed PMID: 11866017. Epub
2002/02/28. eng.
malignancies [48]. Perhaps because of the
3. Rath GS, Schaff JT, Snider GL. Flexible fiberoptic
underlying diseases, the outcome and survival bronchoscopy. Techniques and review of 100 bron-
seem to be worse than in primary COP [49]. The choscopies. Chest. 1973;63(5):689–93. PubMed
presenting symptoms in primary and secondary PMID: 4703621. Epub 1973/05/01. eng.
4. Zavala DC. Diagnostic fiberoptic bronchoscopy: tech-
organizing pneumonia are not specific with
niques and results of biopsy in 600 patients. Chest.
cough and dyspnea on exertion being most com- 1975;68(1):12–9. PubMed PMID: 168036. Epub
mon. Fever is variable and there are no distin- 1975/07/01. eng.
guishing laboratory findings. The radiological 5. Cooley ME. Symptoms in adults with lung cancer: a
systematic research review. J Pain Symptom Manage.
presentation is protean ranging from dense areas
2000;19(2):137–53.
of parenchymal consolidation, patchy broncho- 6. Hirshberg B, Biran I, Glazer M, Kramer MR.
centric infiltrates, to discrete centrilobular densi- Hemoptysis: etiology, evaluation, and outcome in a
ties [50]. Accurate diagnosis is currently made tertiary referral hospital. Chest. 1997;112(2):440–4.
7. Oberg K, Castellano D. Current knowledge on diag-
mostly by surgical lung biopsy, but this approach
nosis and staging of neuroendocrine tumors. Cancer
may be supplanted as experience grows with Metastasis Rev. 2011;30 Suppl 1:3–7. PubMed
transbronchial cryobiopsy. PMID: 21311954. Epub 2011/02/12. eng.
10 R.L. Perez

8. Yao JC, Hassan M, Phan A, Dagohoy C, Leary C, 22. Austin JH, Muller NL, Friedman PJ, Hansell DM,
Mares JE, et al. One hundred years after “carcinoid”: Naidich DP, Remy-Jardin M, et al. Glossary of terms
epidemiology of and prognostic factors for neuroen- for CT of the lungs: recommendations of the
docrine tumors in 35,825 cases in the United States. Nomenclature Committee of the Fleischner Society.
J Clin Oncol. 2008;26(18):3063–72. PubMed PMID: Radiology. 1996;200(2):327–31. PubMed PMID:
18565894. Epub 2008/06/21. eng. 8685321. Epub 1996/08/01. eng.
9. Borczuk AC. Benign tumors and tumorlike conditions 23. de Groot P, Munden RF. Lung cancer epidemiology,
of the lung. Arch Pathol Lab Med. 2008;132(7):1133– risk factors, and prevention. Radiol Clin North Am.
48. PubMed PMID: 18605767. Epub 2008/07/09. eng. 2012;50(5):863–76. PubMed PMID: 22974775. Epub
10. Kraft M, Cassell GH, Henson JE, Watson H, 2012/09/15. eng.
Williamson J, Marmion BP, et al. Detection of 24. Gazzoni FF, Severo LC, Marchiori E, Irion KL,
Mycoplasma pneumoniae in the airways of adults Guimaraes MD, Godoy MC, et al. Fungal diseases
with chronic asthma. Am J Respir Crit Care Med. mimicking primary lung cancer: radiologic-pathologic
1998;158(3):998–1001. PubMed PMID: 9731038. correlation. Mycoses. 2013;57(4):197–208. PubMed
Epub 1998/09/10. eng. PMID: 24147761. Epub 2013/10/24. Eng.
11. Yamaya M. Virus infection-induced bronchial asthma 25. Ribeiro M, Fritscher LG, Al-Musaed AM, Balter MS,
exacerbation. Pulm Med. 2012;2012:834826. Hoffstein V, Mazer BD, et al. Search for chronic
PubMed PMID: 22966430. Pubmed Central PMCID: beryllium disease among sarcoidosis patients in
PMC3432542. Epub 2012/09/12. eng. Ontario, Canada. Lung. 2011;189(3):233–41.
12. Rikimaru T. Endobronchial tuberculosis. Expert Rev PubMed PMID: 21400234. Epub 2011/03/15. eng.
Anti Infect Ther. 2004;2(2):245–51. PubMed PMID: 26. Marin C, Robles R, Lopez Conesa A, Torres J, Flores
15482190. Epub 2004/10/16. eng. DP, Parrilla P. Outcome of strict patient selection for
13. Xue Q, Wang N, Xue X, Wang J. Endobronchial surgical treatment of hepatic and pulmonary metasta-
tuberculosis: an overview. Eur J Clin Microbiol Infect ses from colorectal cancer. Dis Colon Rectum.
Dis. 2011;30(9):1039–44. PubMed PMID: 21499709. 2013;56(1):43–50. PubMed PMID: 23222279. Epub
Epub 2011/04/19. eng. 2012/12/12. eng.
14. Sulavik SB. Bronchocentric granulomatosis and aller- 27. Descombes E, Gardiol D, Leuenberger
gic bronchopulmonary aspergillosis. Clin Chest Med. P. Transbronchial lung biopsy: an analysis of 530
1988;9(4):609–21. PubMed PMID: 3069292. Epub cases with reference to the number of samples.
1988/12/01. eng. Monaldi Arch Chest Dis. 1997;52(4):324–9. PubMed
15. Greenberger PA. Chapter 18: allergic bronchopulmo- PMID: 9401359. Epub 1997/08/01. eng.
nary aspergillosis. Allergy Asthma Proc. 2012;33 28. National Lung Screening Trial Research Team,
Suppl 1:S61–3. PubMed PMID: 22794691. Epub Aberle DR, Adams AM, Berg CD, Black WC, Clapp
2012/07/20. eng. JD, Fagerstrom RM, Gareen IF, Gatsonis C, Marcus
16. Liebow AA. The J. Burns Amberson lecture— PM, Sicks JD. Reduced lung-cancer mortality with
pulmonary angiitis and granulomatosis. Am Rev low-dose computed tomographic screening. N Engl J
Respir Dis. 1973;108(1):1–18. PubMed PMID: Med. 2011;365(5):395–409.
4577269. Epub 1973/07/01. eng. 29. Viggiano RW, Swensen SJ, Rosenow 3rd EC.
17. Clee MD, Lamb D, Clark RA. Bronchocentric granu- Evaluation and management of solitary and multiple
lomatosis: a review and thoughts on pathogenesis. Br pulmonary nodules. Clin Chest Med. 1992;13(1):83–
J Dis Chest. 1983;77(3):227–34. PubMed PMID: 95. 19920617 DCOM-19920617(0272-5231 (Print)).
6351887. Epub 1983/07/01. eng. eng.
18. Iannuzzi MC, Fontana JR. Sarcoidosis: clinical pre- 30. Leong S, Ju H, Marshall H, Bowman R, Yang I, Ree
sentation, immunopathogenesis, and therapeutics. AM, et al. Electromagnetic navigation bronchoscopy:
JAMA. 2011;305(4):391–9. PubMed PMID: a descriptive analysis. J Thorac Dis. 2012;4(2):173–
21266686. Epub 2011/01/27. eng. 85. PubMed PMID: 22833823. Pubmed Central
19. Harrison BD, Shaylor JM, Stokes TC, Wilkes PMCID: PMC3378214. Epub 2012/07/27. eng.
AR. Airflow limitation in sarcoidosis—a study of pul- 31. Eberhardt R, Anantham D, Ernst A, Feller-Kopman
monary function in 107 patients with newly diag- D, Herth F. Multimodality bronchoscopic diagnosis
nosed disease. Respir Med. 1991;85(1):59–64. of peripheral lung lesions: a randomized controlled
PubMed PMID: 2014359. Epub 1991/01/01. eng. trial. Am J Respir Crit Care Med. 2007;176(1):36–
20. Ernst A, Silvestri GA, Johnstone D. Interventional 41. 20070625 DCOM- 20071123(1535-4970
pulmonary procedures: Guidelines from the (Electronic)). eng.
American College of Chest Physicians. Chest. 32. Gilman MJ, Wang KP. Transbronchial lung biopsy in
2003;123(5):1693–717. PubMed PMID: 12740291. sarcoidosis. An approach to determine the optimal
Epub 2003/05/13. eng. number of biopsies. Am Rev Respir Dis.
21. Yarmus L, Feller-Kopman D. Bronchoscopes of the 1980;122(5):721–4. PubMed PMID: 7447156. Epub
twenty-first century. Clin Chest Med. 2010;31(1):19– 1980/11/01. eng.
27. Table of Contents. PubMed PMID: 20172429. 33. Shim HS, Park MS, Park IK. Histopathologic findings
Epub 2010/02/23. eng. of transbronchial biopsy in usual interstitial pneumonia.
1 Small Sample Lung Biopsy Techniques in the Diagnosis of Airway… 11

Pathol Int. 2010;60(5):373–7. PubMed PMID: based guidelines for diagnosis and management. Am
20518887. Epub 2010/06/04. eng. J Respir Crit Care Med. 2011;183(6):788–824.
34. Wall CP, Gaensler EA, Carrington CB, Hayes PubMed PMID: 21471066. Epub 2011/04/08. eng.
JA. Comparison of transbronchial and open biopsies 43. Park IN, Jegal Y, Kim DS, Do KH, Yoo B, Shim TS,
in chronic infiltrative lung diseases. Am Rev Respir et al. Clinical course and lung function change of idio-
Dis. 1981;123(3):280–5. PubMed PMID: 7224338. pathic nonspecific interstitial pneumonia. Eur Respir
Epub 1981/03/01. eng. J. 2009;33(1):68–76. PubMed PMID: 18829672.
35. Garwood S, Judson MA, Silvestri G, Hoda R, Fraig M, Epub 2008/10/03. eng.
Doelken P. Endobronchial ultrasound for the diagnosis 44. Elliot TL, Lynch DA, Newell Jr JD, Cool C, Tuder R,
of pulmonary sarcoidosis. Chest. 2007;132(4):1298– Markopoulou K, et al. High-resolution computed
304. PubMed PMID: 17890467. Epub 2007/09/25. eng. tomography features of nonspecific interstitial pneu-
36. von Bartheld MB, Dekkers OM, Szlubowski A, monia and usual interstitial pneumonia. J Comput
Eberhardt R, Herth FJ, in’t Veen JC, et al. Assist Tomogr. 2005;29(3):339–45. PubMed PMID:
Endosonography vs conventional bronchoscopy for 15891504. Epub 2005/05/14. eng.
the diagnosis of sarcoidosis: the GRANULOMA ran- 45. Devakonda A, Raoof S, Sung A, Travis WD, Naidich
domized clinical trial. JAMA. 2013;309(23):2457–64. D. Bronchiolar disorders: a clinical-radiological diag-
PubMed PMID: 23780458. Epub 2013/06/20. eng. nostic algorithm. Chest. 2010;137(4):938–51.
37. Hirschmann JV, Pipavath SN, Godwin JD. PubMed PMID: 20371529. Epub 2010/04/08. eng.
Hypersensitivity pneumonitis: a historical, clinical, 46. Marten K. [Smoking-related interstitial lung dis-
and radiologic review. Radiographics. 2009;29(7): eases]. Rofo. 2007;179(3):268–75. PubMed PMID:
1921–38. PubMed PMID: 19926754. Epub 17325994. Epub 2007/02/28. Interstitielle
2009/11/21. eng. Lungenerkrankungen bei Rauchern. ger.
38. Selman M, Pardo A, King Jr TE. Hypersensitivity 47. Myers JL, Colby TV. Pathologic manifestations of
pneumonitis: insights in diagnosis and pathobiology. bronchiolitis, constrictive bronchiolitis, cryptogenic
Am J Respir Crit Care Med. 2012;186(4):314–24. organizing pneumonia, and diffuse panbronchiolitis.
PubMed PMID: 22679012. Epub 2012/06/09. eng. Clin Chest Med. 1993;14(4):611–22.
39. Babiak A, Hetzel J, Krishna G, Fritz P, Moeller P, 48. Cordier JF. Organising pneumonia. Thorax.
Balli T, et al. Transbronchial cryobiopsy: a new tool 2000;55(4):318–28. PubMed PMID: 10722773.
for lung biopsies. Respiration. 2009;78(2):203–8. Pubmed Central PMCID: PMC1745738. Epub
PubMed PMID: 19246874. Epub 2009/02/28. eng. 2000/03/18. eng.
40. Caminati A, Harari S. Smoking-related interstitial 49. Drakopanagiotakis F, Paschalaki K, Abu-Hijleh M,
pneumonias and pulmonary Langerhans cell histiocy- Aswad B, Karagianidis N, Kastanakis E, et al.
tosis. Proc Am Thorac Soc. 2006;3(4):299–306. Cryptogenic and secondary organizing pneumonia:
PubMed PMID: 16738193. Epub 2006/06/02. eng. clinical presentation, radiographic findings, treatment
41. Raghu G, Nicholson AG, Lynch D. The classification, response, and prognosis. Chest. 2011;139(4):893–
natural history and radiological/histological appear- 900. PubMed PMID: 20724743. Epub 2010/08/21.
ance of idiopathic pulmonary fibrosis and the other eng.
idiopathic interstitial pneumonias. Eur Respir Rev. 50. Greenberg-Wolff I, Konen E, Ben Dov I, Simansky D,
2008;17(109):108–15. Perelman M, Rozenman J. Cryptogenic organizing
42. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, pneumonia: variety of radiologic findings. Isr Med
Brown KK, et al. An official ATS/ERS/JRS/ALAT Assoc J. 2005;7(9):568–70. PubMed PMID:
statement: idiopathic pulmonary fibrosis: evidence- 16190479. Epub 2005/09/30. eng.
Principles of Imaging Lung Disease
2
James G. Ravenel

Computed Tomography
Imaging Techniques
Virtually, all scanners in use utilize multi-slice
Chest Radiograph helical technology meaning the data is obtained
continuously throughout a single breath hold.
Chest radiography is best thought of as a screening As technology has advanced, the newest genera-
tool. Its strengths are ease and rapidity of acquisi- tion allows for the acquisition of isotropic datasets
tion and low radiation dose. Ideally, this is in under 10 s. The significance of this lies in the
obtained as a posterior-anterior (PA) and lateral ability to reconstruct thin sections in any plane to
projection. In the PA projection, the X-ray beam best display the relevant anatomy, assess the pul-
is behind the patient, and the radiographic cassette monary parenchyma, or obtain the volume of a
is against the anterior chest wall in order to mini- nodule. In these scanners, a volumetric acquisi-
mize magnification of anterior structures. The lat- tion replaces the need for axial inspiratory high-
eral projection is taken with the left chest adjacent resolution CT images. Thin-section (~1 mm) CT
to the cassette to minimize magnification of the is critical for the analysis of small pulmonary
heart. The chest X-ray can then be used to guide nodules (<10 mm), in the evaluation of growth,
further imaging work-up by providing an under- and the determination of part-solid and ground-
standing of whether abnormalities are primarily glass components.
confined to the gas-containing regions of the lung High-resolution CT, typically performed for
(airspace disease), the pulmonary interstitium the evaluation of interstitial lung disease, is some-
(the scaffolding of connective tissue that invests what of a misnomer. With previous CT technology,
pulmonary arteries, veins, lymphatics, and air- it was not possible to obtain thin sections through-
ways), the mediastinum, or pleura. Based on this, out the chest in a single breath hold. Therefore,
a selection of CT technique can be made that may individual thin sections (usually 1 mm) were
include the use of intravenous contrast, thin sec- obtained in inspiration at intervals of 10–20 mm in
tions, expiratory images, and/or multi-planar order to obtain a sampling across the lung paren-
reformatted images. chyma. These images are reconstructed in a man-
ner that accentuates the pulmonary interstitium.
In many institutions, the inspiratory supine images
J.G. Ravenel, M.D. (*) are now obtained as a volumetric acquisition. In
Department of Radiology and Radiologic Services,
additional, two adjuncts are typically performed:
Medical University of South Carolina, 96 Jonathan
Lucas Street, MSC 323, Charleston, SC 29425, USA expiratory images to assess for air trapping as a
e-mail: ravenejg@musc.edu manifestation of small airways disease and prone

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 13


DOI 10.1007/978-1-4939-2575-9_2, © Springer Science+Business Media New York 2015
14 J.G. Ravenel

inspiration to determine whether subtle attenuation


changes in the dependent portions of the lungs on Interstitium, Alveolar Walls,
supine images were due to gravitational fluid shifts and Airspaces
or mild interstitial disease. These images are typi-
cally performed at three levels: top of the aortic The pulmonary parenchyma can be compartmen-
arch, tracheal carina, and apex of the diaphragms. talized into various constituents that make up the
lung. While it can be difficult to absolutely delin-
eate these features with chest radiograph, thin-
18F-FDG PET/CT section CT (TSCT) can often distinguish these
components. It is important to recognize that even
Positron emission tomography (PET) is used to at “high” resolution, CT cannot image the alveolar
image various positron-emitting radionuclides, wall or normal intralobular septa, and thus abnor-
predominately 18F-fluorodeoxyglucose (18F-FDG) malities at this level cannot always be easily attrib-
with the PET images superimposed onto a low- uted to an alveolar filling or interstitial process [1].
dose attenuation correction CT (PET/CT). The following is a lexicon with definitions and
18F-FDG-like glucose is transported into the relevant images of the terminology associated with
cell; however, once phosphorylated, it becomes the pulmonary parenchyma [2]:
trapped. As such 18F-FDG acts as a surrogate for Airspace disease defines a process that fills the
glucose metabolism. Areas that have higher activ- alveolar spaces. The term itself does not define
ity than background are often termed “hypermeta- density and may range from hazy ground-glass
bolic” as raise the possibility than a given lesion is opacity to dense consolidation (Fig. 2.1). An air
a neoplasm. It is important to recognize that bronchogram is often present. It is otherwise a non-
(1) inflammatory lesion may also accumulate specific term that can be associated with infection,
high levels of 18F-FDG and (2) small neoplasms edema, hemorrhage, or cells/cellular debris.
and low-grade lung adenocarcinomas may not Air trapping can be seen on expiratory images
accumulate 18F-FDG above background. This as an accentuation of density between adjacent
means that CT morphology of lesions must be areas of the lungs. When attenuation is measured,
taken into consideration. areas of air trapping maintain their low attenuation

Fig. 2.1 Airspace disease due to bacterial pneumonia. (a) Axial CT image reveals two separate forms of airspace
Frontal chest radiograph reveals poorly defined lower lobe disease: (1) dense anterior segmental consolidation (C)
consolidation (C) with air bronchograms (arrow) and no and (2) bronchopneumonia pattern as evidenced posteri-
volume loss indicating an airspace-filling process. (b) orly by centrilobular nodules (arrows)
2 Principles of Imaging Lung Disease 15

Fig. 2.4 Bronchiectasis. Axial CT in patient with cystic


fibrosis reveals cystic bronchiectasis (C) in the right lung
and varicoid (V) bronchiectasis in the left lung
Fig. 2.2 Architectural distortion and honeycomb lung.
Axial TSCT reveals distortion of the inferior lingular seg-
ment (arrows) and extensive honeycomb change in the
right lower lobe (H)

Fig. 2.3 Lobar collapse. (a) Frontal radiograph reveals radiograph confirms collapse with anterior displacement
volume loss in left hemithorax with elevation of left dia- of the major fissure (arrowheads)
phragm due to left hilar neoplasm (arrow). (b) Lateral

values, whereas normal lung increases in density. Atelectasis is the loss of air and volume
This is typically a manifestation of small airway caused by the collapse of the airspaces (Fig. 2.3).
disease but can result from either luminal obstruc- It is therefore the opposite of airspace disease.
tion or concentric bronchial wall thickening [3]. Bronchiectasis is defined as irreversible
Architectural distortion refers to the displace- dilation of the airways usually measured as 1.5×
ment of normal lung structures with disruption of the size of the adjacent artery [4]. It can be cylin-
the usual lung parenchyma. It is generally associated drical (long and tubular), varicoid (variable lumen
with a fibrosing lung process (Fig. 2.2). size), or cystic (large gas-filled spaces) (Fig. 2.4).
16 J.G. Ravenel

Fig. 2.5 Subacute hypersensitivity pneumonitis. Axial Fig. 2.6 Lymphangitic carcinomatosis. Axial CT image
TSCT reveals both the heterogeneous generalized increase reveals smooth interlobular septal thickening (arrow-
in density of ground-glass opacity and ill-defined faint nod- heads) on the left compared with contralateral lung. Note
ular opacities centered in the secondary pulmonary lobule also small left pleural effusion (E)

The relatively small artery next to a dilated bron- Ground-glass opacity is used to describe a
chus has been referred to as the “signet ring sign.” hazy increase in lung attenuation that does not
Traction bronchiectasis is a unique subset of obscure the underlying lung architecture (see
bronchiectasis and is related to underlying paren- Fig. 2.5) [7]. It can be associated with airspace-
chymal fibrosis [5]. As opposed to bronchiectasis filling process or fibrosis that is below the resolu-
described above, the dilation is the result of exter- tion of CT. Because of this, the moniker of “active
nal retractile forces. When severe, it can be quite alveolitis” is discouraged, although this may rep-
difficult to segregate out from honeycomb lung. resent reversible cellular change in the setting of
Centrilobular nodules are hazy, ill-defined interstitial lung disease.
nodules that are centered around the central Honeycombing is a late radiographic finding
bronchus and artery in the secondary pulmonary of underlying fibrosis with stacked thick-walled
lobule (Fig. 2.5). These nodules do not extend out cystic structures (see Fig. 2.2). In the classic
to the interlobular septum and are classically basal and peripheral distribution, it helps to
associated with subacute hypersensitivity pneu- define a usual interstitial pneumonitis pattern and
monitis and respiratory bronchiolitis [1]. obviates the need for open lung biopsy [8]. It is
Consolidation refers to generalized increased important to recognize that honeycombing pres-
density within the lung parenchyma. As such it is ent within a pathologic specimen may not always
less specific than airspace disease or atelectasis be evident at TSCT.
and encompasses both of those possibilities. Infiltrate, as a term, should no longer be part
Crazy-paving pattern combines ground- of the imaging lexicon as it means too many dif-
glass opacity superimposed upon interlobular ferent things to too many people. It should be
septal thickening somewhat resembling paved understood as a nonspecific term in the vein of
cobblestones. Although classically associated opacity and may be used with appropriate
with alveolar proteinosis, it can occur as part of descriptors (air space, interstitial, etc.).
any disease process that involves both intersti- Interlobular septal thickening refers to any
tium and air spaces [6]. process that results in increased conspicuity of the
Emphysema, like its pathological counterpart, walls of the secondary pulmonary lobule [1].
is the irreversible destruction of alveolar walls. It This may alternatively be described as reticular or
can be manifest as predominately lucency of the reticulonodular opacities depending on morphol-
secondary pulmonary lobule (centrilobular ogy. Best seen perpendicular to the pleural surface,
emphysema) or more widespread destruction of smooth interlobular septal thickening (Fig. 2.6)
the lung parenchyma (panacinar emphysema). or reticular opacities are a classic manifestation
2 Principles of Imaging Lung Disease 17

of hydrostatic pulmonary edema. A beaded or


reticulonodular appearance can be seen with such
conditions as sarcoidosis and lymphangitic
carcinomatosis.
Miliary pattern is a form of interstitial disease
with small nodules (1–3 mm) confined to the inter-
stitium often in a random distribution without
septal thickening. Diagnoses classically associ-
ated with this pattern include disseminated infec-
tion, particularly tuberculosis and sarcoidosis.
Mosaic attenuation is a general term used to
describe differing lung densities. As such it can
be due to a mixture of ground-glass opacity and Fig. 2.7 Sarcoidosis. Axial CT reveals both subpleural/
normal areas of air trapping and normal lung or perilymphatic nodules (black arrowheads) and thickening
along bronchovascular bundles (white arrowheads). Note
areas of differential (mosaic) perfusion [9]. The also bilateral hilar adenopathy (A)
former two possibilities can be sorted out with
expiratory images as both ground-glass disease
and normal lung will both increase in density,
whereas air trapping will manifest as persistent
low density. Mosaic perfusion can be differenti-
ated by looking at the underlying vessels with
larger vessels going to areas of increased density
and small or diminutive vessels going to areas of
decreased density as in cases of chronic thrombo-
embolic pulmonary hypertension.
Opacity, like infiltrate, should denote a non-
specific increase in density and should be pre-
ceded by an appropriate descriptor such as air
space or interstitial.
Peribronchovascular nodule describes small Fig. 2.8 Active tuberculosis. Axial CT reveals branching
nodules (1–3 mm) that are discretely related to endobronchial nodules that appear like buds on a tree
the bronchial wall or peribronchial structures (arrows)
(Fig. 2.7). It therefore may be associated with
disease of the bronchus, adjacent vessels, or lym- seen in disease associated with distal mucoid
phatics. Nodules of this variety may also be impaction such as cystic fibrosis [10].
described as bronchocentric.
Perilymphatic nodule is quite similar in size
to peribronchovascular nodules but instead Nodules and Masses
resides in the interlobular septa (see Fig. 2.7).
This can be associated with interlobular septal By convention, a pulmonary nodule is defined as a
thickening and also may be described as reticulo- focal opacity surrounded by lung measuring less than
nodular opacities. 3 cm as defined by chest radiograph. A lesion
Tree-in-bud opacities are small nodules at greater than 3 cm in size is considered to be a mass.
the end of airways and are a manifestation of As a practical rule, the major task for the radiologist is
endobronchial spread of disease. Classically to distinguish between benign and malignant causes.
described with active tuberculosis (Fig. 2.8), they The presence of dense central calcification, the pres-
may be the result of any infectious process or be ence of macroscopic intralesional fat, and a lack of
18 J.G. Ravenel

Fig. 2.9 Pulmonary nodules. (a) Ground glass. (b) Part solid. (c) Solid

growth over 2 years are all strong predictors of benig-


nancy [11]. Morphologically, the lesions’ borders can
also help malignancy. Lesions with entirely smooth
borders are more likely to be benign, while lobulated
and spiculated nodules are more likely to be malig-
nant. CT is the primary means of nodule evaluation,
and the description of a nodule may lead to a refine-
ment of the differential diagnosis.
Ground-glass nodule is a nodule at TSCT
that is greater in density than the surrounding
lung but does not obscure the underlying struc-
ture of the interstitium (Fig. 2.9a). Some lesion
may contain internal bubble-like lucencies that
correspond to dilated alveolar spaces.
Part-solid nodule contains areas that are both Fig. 2.10 Invasive aspergillosis. Axial CT reveals solid
ground glass in density as well as components that nodular consolidation with faint surrounding ground-
are solid (Fig. 2.9b). Along with ground-glass nod- glass halo (arrowheads)
ules, these are lesions typically detected by CT as
incidental findings or at lung cancer screening and Halo sign refers to a ring of ground-glass
when malignant tends to be either premalignant, in opacity surrounding a centrally dense lesion with
situ, or low-grade invasive neoplasms [12]. or without cavitation. While classically associ-
Solid nodule is of sufficient density to obscure ated with invasive aspergillosis (the halo repre-
the underlying lung parenchyma (Fig. 2.9c). senting peripheral hemorrhage) (Fig. 2.10), it can
Cavitation refers to the process of internal be seen with any form of infection, vasculitis, or
necrosis leaving an internal area of gas within the malignancy [14].
lesion. The wall thickness around a cavity can Reversed halo sign (atoll sign) is the opposite
help predict malignancy as lesions with thick of the halo sign with ground-glass density cen-
irregular walls or external notches are more likely trally and a surrounding ring of solid density. It is
to be malignant [13]. Cavitation is distinct generally indicative of an inflammatory lesion,
from the bubble-like lucencies that can be seen in usually organizing pneumonia [15], but can also
low-grade adenocarcinomas described in ground- be seen with active infection and granulomatous
glass opacity above. inflammation.
2 Principles of Imaging Lung Disease 19

Mediastinum

The mediastinum contains the central core struc-


tures of the thorax and is invested by the parietal
pleura [16]. The mediastinum is continuous with
the pulmonary interstitium at the hila where the
central bronchi, pulmonary arteries, veins, and
lymphatics enter the lung. The mediastinum is
also in direct continuity with the soft tissues of
the neck and continuous with the sub-peritoneal
spaces of the abdomen at the esophageal, inferior
vena cava, and aortic hiatus. Although no direct
fascial planes exist to separate mediastinal com-
ponents, the mediastinum is classically divided
into three components, each suggesting a specific
group of lesions. It is important to recognize that Fig. 2.11 Mediastinal compartments. Lateral radiograph
all three sections are in continuity with each delineating approximate anatomic location of anterior
other, and therefore involvement of more than (A), middle (M), and posterior (P) mediastinum
one compartment may occur.
Anterior mediastinum extends from the pos- tissues. Lesions in this region are often tumors of
terior wall of the sternum to the ascending aorta neurogenic origin, extension of disk space infec-
superiorly and the anterior pericardium inferi- tion, and lesions involving vertebral bodies.
orly. Contents of this region include fat, thymus, Given the variability in position, lesions of the
and lymphoid tissue. Ectopic thyroid and para- descending aorta and esophagus may also local-
thyroid tissue as well as germ cell rests may also ize to the posterior mediastinum (Fig. 2.11).
reside here, and thyroid goiters may descend
anteriorly into this space. The differential diag-
nosis derives from these tissues and classically Pleura
includes lymphoma, thymic tumors, germ cell
tumors, and thyroid lesions. On chest radiographs, pleural effusions are often
Middle mediastinum extends from the ante- manifest as blunting of the lateral or posterior
rior mediastinum to the anterior margin of the costophrenic sulci resulting in a meniscoid
thoracic spine and as such contains the visceral appearance. Effusions of this type are usually
structures of the mediastinum (the esophagus and free to move throughout the pleural space. As
aorta can be variable as to whether they occupy pleural fluid becomes more organized, it may
what is classically considered to be middle or take on a lenticular appearance. In this setting,
posterior mediastinum). The primary abnormali- loculation is usually present, and the fluid does
ties tend to be those arising from the lymphatics not shift with changing position. As a rule of
such as mediastinal adenopathy from a primary thumb, transudative effusions tend to be free
lung malignancy or lymphoma. Lesions arising flowing, but exudative effusions can be either
from the aorta and esophagus as well as congeni- free flowing or loculated. Thus the content of
tal bronchopulmonary foregut duplication cysts pleural fluid cannot be based solely on imaging
may also occupy the middle mediastinum. appearance and should be based on pleural fluid
Posterior mediastinum is the region extend- analysis. At CT, the split pleura sign, enhance-
ing posteriorly from the middle mediastinum ment of both visceral and parietal pleura, is often
that encompasses predominately paraspinal soft used to suggest the possibility of an empyema.
20 J.G. Ravenel

It should be noted that chronic exudative effusions 6. Rossi SE, Erasmus JJ, Volpacchio M, Franquet T,
Castiglioni T, McAdams HP. “Crazy-paving” pattern
and complex parapneumonic effusions can also
at thin section CT of the lungs: radiologic-pathologic
present with a split pleura sign [17]. overview. Radiographics. 2003;23:1509–19.
Malignant pleural effusions are those with 7. Remy-Jardin M, Remy J, Giraud F, Wattinne L,
tumor cells in the pleural space and most often Gosselin B. Computed tomography (CT) assessment
of ground-glass opacity: semiology and significance.
exudate. Effusions in lung cancer patients may
J Thorac Imaging. 1993;8:249–64.
also result unrelated to the tumor itself (cardiac, 8. Lynch DA, David GJ, Safrin S, et al. High-resolution
hepatic, renal disease, etc.) or may be due to cen- computed tomography in idiopathic pulmonary fibro-
tral venous obstruction, central lymphatic obstruc- sis: diagnosis and prognosis. Am J Respir Crit Care
Med. 2005;172:488–93.
tion, or post-obstructive atelectasis/pneumonitis
9. Worthy SA, Muller NL, Hartman TE, Swensen SJ,
[18]. The latter categories are considered “parama- Padley SP, Hansell DM. Mosaic attenuation pattern
lignant.” Sampling of the fluid is considered on thin-section CT scans of the lung; differentiation
necessary to document a malignant effusion. CT among infiltrative lung, airway, and vascular diseases
as a cause. Radiology. 1997;205:465–70.
(and PET/CT) is not always diagnostic. Findings
10. Eisenhuber E. The tree-in-bud sign. Radiology.
suggesting a malignant effusion at CT include 2002;222:771–2.
parietal pleural thickness >1 cm, circumferential 11. Erasmus JJ, Connolly JE, McAdams HP, Roggli
thickening, nodules, and mediastinal pleural VL. Solitary pulmonary nodules: part I. Morphologic
evaluation for differentiation of benign and malignant
involvement. Increased metabolic activity may be
lesions. Radiographics. 2000;20:43–58.
seen associated with the pleura at PET/CT. 12. Naidich DP, Bankier AA, Macmahon H, et al.
Recommendations for the management of subsolid
pulmonary nodules detected at CT: a statement
from the Fleischner Society. Radiology. 2013;266:
References 304–17.
13. Honda O, Tsusubamoto M, Inoue A, et al. Pulmonary
1. Webb WR. Thin-section CT, of the secondary pulmo- cavitary nodules on computed tomography; differen-
nary lobule: anatomy and the image—the 2004 tiation of malignancy and benignancy. J Comput
Fleischner lecture. Radiology. 2006;239:322–38. Assist Tomogr. 2007;31:943–9.
2. Hansell DM, Bankier AA, MacMahon H, McLoud 14. Primack SL, Hartman TE, Lee KS, Muller NL.
TC, Muller NL, Remy J. Fleischner society: glossary Pulmonary nodules and the CT halo sign. Radiology.
of terms for thoracic imaging. Radiology. 2008;246: 1994;190:513–5.
697–722. 15. Kim SJ, Lee KS, Ryu YH, et al. Reversed halo sign on
3. Arakawa H, Webb WR, McCowin M, Katsou G, Lee high-resolution CT of cryptogenic organizing pneu-
KN, Seitz RF. Inhomogeneous lung attenuation at monia: diagnostic implications. Am J Roentgenol.
thin-section CT: diagnostic value of expiratory scans. 2003;180:1251–4.
Radiology. 1998;206:89–94. 16. Proto AV. Mediastinal anatomy: emphasis on conven-
4. Naidich DP, McCauley DI, Khouri NF, Stitik FP, tional images with anatomic and computed tomo-
Siegelman SS. Computed tomography of bronchiecta- graphic correlations. J Thorac Imaging. 1987;2:1–48.
sis. J Comput Assist Tomogr. 1982;6:437–44. 17. Aquino SL, Webb WR, Gushiken BJ. Pleural exudates
5. Desai SR, Wells AU, Rubens MB, DuBois RM, and transudates: diagnosis with contrast-enhanced CT.
Hansell DM. Traction bronchiectasis in cryptogenic Radiology. 1994;192:803–8.
fibrosing alveolitis: associated computed tomographic 18. Heffner JE, Klein JS. Recent advances in the diagno-
features and physiological significance. Eur Radiol. sis and management of malignant pleural effusions.
2003;13:1801–8. Mayo Clin Proc. 2008;83:235–50.
Types of Biopsies
3
Mostafa M. Fraig

depending on the case. Some pathologists like to


Types of Biopsies perform pentachrome stains like Movat stains,
reticulin, Masson trichrome, iron stains, and AFB
Wedge Biopsy and GMS stains on a routine basis. In our experi-
ence, these are not needed on all or on the major-
It is considered the gold standard for diagnosing ity of cases, and they can result in more work and
lung conditions where a relatively large area of the cost for the lab.
lung parenchyma is needed to scan for focal
changes or identify all the features required for
rendering a specific diagnosis. It is useful for diag- Transbronchial Biopsy
nosing interstitial lung disease and other diffuse
diseases like hypersensitivity pneumonia and dif- This is the most frequent type of lung biopsy to
ferentiating it from nonspecific pneumonia or come to the pathology laboratory and usually
other idiopathic interstitial pneumonias [1]. obtained by pulmonologists in the course of the
It is considered a complex surgical procedure bronchoscopy procedure along with other sampling
requiring splitting chest muscle and spreading the methods such as bronchial brushings and washings
ribs with disarticulation or resection near the and bronchoalveolar lavages. This affords the labo-
sternum. Patients are left with chest tubes and ratory the variety of specimens to perform other
stitches that could remain painful for weeks [1, 2]. ancillary studies like microbiologic cultures on
Handling these biopsies requires removing the the lavage fluid and spare some of the tissue for
stapled lines at the edge and inking the edge if histopathologic evaluation [3].
malignancy is suspected. The wedge is serially The tissue from this type of biopsy should be
sectioned starting from the cut surface going handled gently, and squeezing the tissue with for-
toward the free pleural edge. The specimen is ceps or leaving it out of formalin for a long time
usually entirely submitted for processing for max- to dry should be avoided. It is preferred to wrap it
imum use of the tissue. There should be a set of in lens paper and not sponges as the latter could
H&E-stained slides with other stains as needed introduce artifacts in the tissue. Several levels,
preferably three, should be performed with the
intervening sections kept on slides for further
M.M. Fraig, M.D. (*) stains or studies. This strategy saves most of the
Department of Pathology and Laboratory Medicine,
School of Medicine, University of Louisville,
tissue from being wasted during the process of
530 South Jackson Street, Louisville, KY 40202, USA facing the block and preserves the focal areas of
e-mail: m.fraig@louisville.edu interest for further evaluation.

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 21


DOI 10.1007/978-1-4939-2575-9_3, © Springer Science+Business Media New York 2015
22 M.M. Fraig

For adequacy purposes, the biopsy should Fine-Needle Aspiration Biopsy


contain at least one portion of the alveolated lung
if the disease process is mainly in that part of the Fine-needle aspiration biopsy is sometimes the
lung. Areas immediately next to the bronchial only option available to adequately biopsy a
wall are known to have many nonspecific findings given lesion. The transbronchial needle, known
like fibrosis or chronic inflammation to be infor- as Wang needle, has a larger gauge, 22, than the
mative about the dominant disease in the lung spring-loaded dissecting needle or needles used
parenchyma, and the pathologist should be care- in the endobronchial ultrasound (EBUS) modal-
ful not to overinterpret those areas. ity. For that reason, they are still useful in obtain-
ing a sizable specimen once the lesion or the
mass is identified by ultrasound.
Needle Core Biopsies The material obtained from fine-needle aspi-
ration is useful in providing smeared slides as
These are usually obtained during CT-guided well as cell blocks for immunohistochemical
biopsy of the lung in the evaluation of peripheral staining. It is also useful for other ancillary stud-
lung nodules that are less amenable to other ies such as microbiologic testing, flow cytometric
modalities for sampling. In the course of the pro- analysis, and molecular testing. On-site adequacy
cedure, the radiologist uses a coaxial needle to tar- assessment is important in the triage as well as
get the lesion and fix the lung so the lesion does providing real-time feedback to the operator per-
not move with respiration. This also helps to make forming the procedure.
several passes into the lesion possible without the
need to retarget the needle. The biopsy needle is
then inserted inside the coaxial needle to reach the Cryobiopsy
lesion of concern. These needles could vary from
fine aspiration one to spring-loaded dissecting Cryobiopsy is the latest type of biopsy that can
ones (Tru-Cut® needles). In the latter, the core is produce larger biopsies of alveolated lung through
rolled on a slide, and the cellularity is evaluated as the application of a flexible freezing probe to
well as the type of lesional tissue to avoid acellular the tissue and then extracting it along with the
material or normal lung tissue. After confirming surrounding tissue still attached to the probe.
the suitability of the targeting, subsequent passes The average size is 15 mm2 compared to 5 mm2
should be dedicated to obtaining tissue for H&E by the forceps-assisted biopsy [7]. The other
evaluation or other ancillary studies such as immu- advantage besides the increased size of the biopsy
nohistochemical stains, flow cytometric analysis, is the lack of crush artifacts frequently introduced
and molecular testing. The latter requires enough by the jaws of the forceps crushing the tissues.
viable cells to be informative, and 4–5 cores are For these reasons, cryobiopsy is becoming more
better for this type of testing as the test is complex suitable for diagnosing diffuse interstitial diseases
and still relatively expensive. Repeated touch that used to require wedge biopsy [1, 8, 9].
imprints on every core are not required, and it usu- An example of a cryobiopsy is shown in Fig. 3.1.
ally results in shedding most of the cells on the The disadvantage is that the bronchoscope needs
slides with few cells left in the core especially with to be withdrawn completely out of the lung with
well-differentiated adenocarcinoma where the the specimen as the specimen cannot pass through
cells are very discohesive and sparse [4–6]. the probe channel.
On sectioning these specimens, the protocol
mentioned in the transbronchial biopsy section
above should apply here too. The sections should Ancillary Studies on Biopsy Material
be kept on charged slides for the anticipated
immunohistochemical stains. For the very impor- Microbiology Studies: These could be performed
tant molecular testing, thicker sections could be on the fluid from the bronchoalveolar lavage
made for DNA or RNA extraction. collected in the course of bronchoscopy.
3 Types of Biopsies 23

Fig. 3.1 Low-magnification view of a transbronchial biopsy (left, a) and a cryobiopsy (right, b) for comparison. Notice
also the lost tissue from the center of the biopsy due to overfreezing

Alternatively the tissue that is removed with the newly diagnosed lung malignancies. At the cur-
staple lines from the wedge biopsy could be used rent time, only adenocarcinoma is the candidate
for this purpose if sterile techniques are followed. tumor for the testing for EGFR mutations and
Other possibility is to dedicate one pass or reflex testing for ALK mutations. However, there
more from the fine-needle aspiration procedure for is a new trend for genetic profiling of lung tumors
that purpose if adequacy assessment is provided, to detect any mutations that could be targeted by
and it is one of the invaluable reasons to provide the available drugs used in other tumors. For
the service. If there is a cell block, special stains example, a BRAF mutation in a lung tumor, while
could be performed on the material to rule out uncommon, may help in choosing a chemothera-
infections. peutic agent used in the treatment of melanoma
Flow Cytometry: Fine-needle aspiration with the same mutation to treat the lung tumor.
passes are used for this purpose especially from The minimum number of cells for adequate
lymph nodes in the chest when lymphoma is DNA extraction is often cited as 150 cells. Reports
entertained in the differential diagnosis. Needle of using the needle rinse for molecular have claimed
cores could be submitted in the suitable media, high percentage of recovery of tumor cells and
RPMI, for further testing. DNA for testing [10]. Different factors play a role in
Immunohistochemical Staining: This should the final adequacy as the presence of necrosis, nor-
be readily performed on sections from the tissue mal cell contamination, and noncellular debris or
biopsies regardless of the method of obtaining material contamination. Every effort should be
them. Cell block material is also invaluable in this made to provide adequate material for such testing
regard as it is better to validate the control tissue whenever possible as the repeat biopsies are time
with formalin-fixed cell block than with alcohol- consuming and expensive to arrange for.
fixed slides. An excellent cell block technique
involves letting the material clot first on a slide and
then scraping the clotted material in formalin. The Common Artifacts and Nonspecific
advantages are several. It is cheap requiring no Findings Identified in Small Lung
additional reagents and uses the patient’s blood as Biopsy
a medium. It provides colored material that is easy
to visualize in paraffin unlike agar, for instance, Several artifacts are known to be present in the
that is hard to distinguish from paraffin. lung and could represent diagnostic pitfalls in the
Molecular Testing: The emergence of tar- interpretation of these biopsies. Some result from
geted chemotherapy in recent years required the instrumentation handling and processing of
pathologist to anticipate this type of testing on the tissue (Fig. 3.2).
24 M.M. Fraig

Fig. 3.2 Nonspecific peribronchial fibrosis and chronic


inflammation are common, especially in older patients or Fig. 3.3 Pigmented macrophages with tiny anthracotic
smokers. The abrupt transition from fibrosed areas to nor- pigmentation are a frequent finding in smokers’ lungs and
mal lung parenchyma should suggest that this is a focal should not be confused with hemosiderin-laden macro-
phenomenon and has no bearing on the disease process phages from alveolar hemorrhage

Pseudo-atelectasis the characteristic chunky refractile material, which


is hemosiderin. In fact, iron stain could be prob-
Compression and squeezing the tissue between lematic as it is positive on pigmented macrophages
the forceps jaws causes this artifact. One way to from smokers and is reported as positive in those
differentiate this is to find sharp transition cases, which again lead to the wrong conclusion of
between the area of compression and the normal presence of intra-alveolar hemorrhage where in
surrounding well-alveolated lung parenchyma. reality it is not the case (Fig. 3.3). It takes only 24 h
On higher magnification, the slit-like air spaces for hemosiderin-laden macrophages to present in
can discerned within those areas of compression. the lung. If they are accompanied by clotted fibrin
This artifact makes it difficult to interpret those in the alveolar space, blood vessels should be scru-
areas affected by it. Gentle handling of the tissue tinized for evidence of vasculitis.
and keeping it moist should help eliminate most
of these artifacts. Careful interpretation is also
warranted by the pathologist. Intra-alveolar Macrophages

It is a frequent finding in many conditions to the


Fresh Blood in the Biopsy point that it is almost nonspecific on a small
biopsy if the background disease is not readily
Usually this is the result of bleeding from the recognized. Smokers and patients with pulmo-
biopsy procedure and does not indicate intra- nary edema or pneumonia or any type of fibrosis
alveolar hemorrhage. In our experience, most or condition that causes the lung to lose its elas-
descriptive diagnoses of small biopsies list this ticity are likely to result in numerous macro-
blood as one of the findings for lack of any specific phages in the alveoli.
diagnosis. This could be confusing to the clinicians
and could lead them along the wrong path investi-
gating the underlying etiology for the alveolar Pseudolipoid Artifact
hemorrhage. True intra-alveolar hemorrhage is rec-
ognized by the presence of numerous hemosiderin- Occasionally air bubbles in the alveolar spaces
laden macrophages that do not require iron stains could assume a rounded configuration that is
to be recognized as they are usually engorged with similar to fat globules noted in exogenous lipoid
3 Types of Biopsies 25

Fig. 3.4 Pseudolipoid artifact showing air bubbles with Fig. 3.5 Carcinoid tumorlets are small nests of cells with
globular shape similar to that of fat globules in lipoid neuroendocrine differentiation as noted by the “salt and
pneumonia pepper” chromatin and the amphophilic cytoplasm. They
are located directly around the endobronchial wall

pneumonia (Fig. 3.4). The difference is that in true Minute Meningothelial-Like


lipoid there are foamy macrophages surrounding Nodules
the fat globules. Other features of aspiration pneu-
monia could be seen like multinucleated giant Other incidental findings on large biopsies and
cells and vegetable matter. lobectomy specimens are the following: They
are usually located within the parenchyma
(to differentiate from carcinoid tumorlets).
Carcinoid Tumorlets Their morphology is akin to that of meningothe-
lial whorls. They have spindle cells with fre-
Nests of neuroendocrine cells growing around the quent pseudoinclusions, and they could be
bronchial walls could occur in patient with chronic positive for EMA and vimentin immunohisto-
obstruction. They are characterized as tumorlets as chemical stains (Fig. 3.6a, b). Although ultra-
long as the largest dimension is less than 0.5 cm. structural evidence was presented to show
Tumors beyond this size are considered as carci- similarity to meningothelial cells, which resulted
noid tumors, either typical or atypical type. These in a change of the name from chemodectoma to
are usually discovered incidentally in lobectomy or meningothelial-like nodules, on the genetic level,
wedge biopsy specimens obtained for other rea- they were not similar to meningioma; hence, their
sons. The morphology is usually bland with uni- origin is uncertain [13].
form cells and round or oval nuclei. The chromatin
is usually granular with “salt and pepper” pattern.
There is nesting of the cells with peripheral pali- Corpora Amylacea
sading of the cells (Fig. 3.5). The stroma could be
scant or could show sclerosis and even calcifica- This is another frequent finding in the lungs of old
tions. The cells are positive for neuroendocrine patients and those with massive pulmonary
markers such as chromogranin A and synaptophy- edema. They are seen in the alveolar spaces
sin [11]. The mitotic activity should be less than assuming the rounded deposits of concentrically
2/10 HPF, and the Ki 67 proliferative should be lamellate bodies with eosinophilic color. They
extremely low (less than 2 %). They can be mis- may represent degenerative by-products, and they
taken for metastatic carcinoma to the lungs [12]. have no clinical significance.
26 M.M. Fraig

Fig. 3.6 Low-magnification images of so-called chemodectoma or minute meningothelial-like nodules (left, a) and
higher magnification (right, b) showing the characteristic spindle cells with their pseudoinclusions

Fig. 3.7 Multilobated megakaryocyte with dark smudgy Fig. 3.8 Astroid body is a frequent finding in the setting
chromatin is entrapped in the alveolar lining and could be of sarcoidosis, but it is not pathognomonic of the disease
confused with viral cytopathic effect or radiation effect and could be found in other conditions

Mineralization and Ossification


Megakaryocytes of Bronchial Cartilage

The lung is a very vascular organ with massive Mineralization or calcification of the bronchial
circulation of blood going through it every min- cartilage is a frequent finding in older patients
ute. Megakaryocytes frequently get trapped in that has no clinical significance. They could also
the fine capillaries of the lung, and they some- have oncocytic change of the seromucinous
times appear in the alveolar lining (Fig. 3.7). glands in the wall of the bronchus. Ossification of
They could be mistaken for viral cytopathic the cartilage could also occur.
effect or the degenerative smudgy cells from che- Other insignificant or incidental findings
motherapy or radiation therapy. They are numer- include smooth muscle nodules, dystrophic calci-
ous in septic conditions and metastatic tumors. fication, and astroid bodies (Fig. 3.8).
3 Types of Biopsies 27

Practical Approach to Small Lung 2. Colt HG, Russack V, Shanks TG, Moser KM.
Comparison of wedge to forceps videothoracoscopic
Biopsy lung biopsy. Gross and histologic findings. Chest.
1995;107:546–50.
1. It is very important to be familiar with the 3. Schwartz LE, Aisner DL, Baloch ZW, Sterman D,
clinical history and/or the imaging studies of Vachani A, Gillespie C, Haas A, Litzky LA. The diag-
nostic efficacy of combining bronchoscopic tissue
the patients to maximize the diagnostic yield
biopsy and endobronchial ultrasound-guided trans-
from these biopsies. bronchial needle aspiration for the diagnosis of malig-
2. On scanning magnification, try to see if the nant lesions in the lung. Diagn Cytopathol. 2013;41:
process is mainly bronchocentric or parenchy- 929–35.
4. Logrono R, Wojtowycz MM, Wunderlich DW, Warner
matous in the alveolated areas.
TF, Kurtycz DF. Fine needle aspiration cytology and
3. Look for acute lung injury pattern with the core biopsy in the diagnosis of alveolar soft part sar-
characteristic fibroblastic plugs and swirls of coma presenting with lung metastases. A case report.
fibroblasts. If that is the case, look for hyaline Acta Cytol. 1999;43:464–70.
5. Priola AM, Priola SM. Computed tomography-guided
membrane, and then look for viral inclusions
needle biopsy of lung lesions: is fine needle aspiration
or acute pneumonia to discern the etiology of really more accurate than core needle biopsy? Acta
either bronchiolitis obliterans/organizing Radiol. 2013;54:1150–1.
pneumonia or diffuse alveolar damage. 6. Yamagami T, Iida S, Kato T, Tanaka O, Nishimura T.
Combining fine-needle aspiration and core biopsy under
4. Granulomas are the next item, and they could
CT fluoroscopy guidance: a better way to treat patients
be subtle and especially the poorly formed with lung nodules? Am J Roentgenol. 2003;180:811–5.
ones. The carrot-shaped nuclei with haphaz- 7. Babiak A, Hetzel J, Krishna G, Fritz P, Moeller P,
ard orientation in a thickened area should be a Balli T, Hetzel M. Transbronchial cryobiopsy: a new
tool for lung biopsies. Respiration. 2009;78:203–8.
tip-off. Follow the algorithm for our radio-
8. Casoni GL, Tomassetti S, Cavazza A, Colby TV,
logic correlation chapter as to what the type of Dubini A, Ryu JH, Carretta E, Tantalocco P, Piciucchi
lesion is. It is important to recognize the dif- S, Ravaglia C, Gurioli C, Romagnoli M, Gurioli C,
ference between airspace lesions and nodular Chilosi M, Poletti V. Transbronchial lung cryobiopsy
in the diagnosis of fibrotic interstitial lung diseases.
or diffuse processes.
PLoS One. 2014;9:e86716.
5. Fibrin in the alveoli should denote leaking 9. Pajares V, Puzo C, Castillo D, Lerma E, Montero MA,
vessels and prompt closer scrutiny of the Ramos-Barbon D, Amor-Carro O, Gil de Bernabe A,
blood vessels. Franquet T, Plaza V, Hetzel J, Sanchis J, Torrego A.
Diagnostic yield of transbronchial cryobiopsy in inter-
6. Sometimes it is what you can rule out from the
stitial lung disease: a randomized trial. Respirology.
biopsy rather than what you are able to diag- 2014;19:900–6.
nose that could help the pulmonologists to 10. Sakairi Y, Sato K, Itoga S, Saegusa F, Matsushita K,
narrow their differential diagnosis. Nakajima T, Yoshida S, Takiguchi Y, Nomura F,
Yoshino I. Transbronchial biopsy needle rinse solution
7. Do not be afraid to say “unremarkable endo-
used for comprehensive biomarker testing in patients
bronchial wall and alveolated lung” instead of with lung cancer. J Thorac Oncol. 2014;9:26–32.
listing all nonspecific findings that confuse 11. Aubry MC, Thomas Jr CF, Jett JR, Swensen SJ,
the clinician. Myers JL. Significance of multiple carcinoid tumors
and tumorlets in surgical lung specimens: analysis of
28 patients. Chest. 2007;131:1635–43.
12. Darvishian F, Ginsberg MS, Klimstra DS, Brogi E.
Carcinoid tumorlets simulate pulmonary metastases
References in women with breast cancer. Hum Pathol. 2006;37:
839–44.
1. Fruchter O, Fridel L, El Raouf BA, Abdel-Rahman N, 13. Torikata C, Mukai M. So-called minute chemodec-
Rosengarten D, Kramer MR. Histological diagnosis toma of the lung. An electron microscopic and immu-
of interstitial lung diseases by cryo-transbronchial nohistochemical study. Virchows Arch A Pathol Anat
biopsy. Respirology. 2014;19:683–8. Histopathol. 1990;417:113–8.
Airspace-Occupying Diseases
4
Mostafa M. Fraig

lining or with viral cytopathic effect showing


Acute Pneumonias ground-glass nuclei. On the other hand, influenza
virus and herpes simplex virus (HSV) tend to
Acute pneumonias could be divided into infec- cause areas of necrosis and some neutrophilic
tious, which include bacterial, fungal, and viral infiltrate similar to that of bacterial pneumonia.
ones, and noninfectious which include aspiration, The clue to the viral etiology would be based on
lipoid, and allergic ones. In most cases of bacterial the presence of viral cytopathic changes with
or viral pneumonia, a biopsy is not needed to make nuclear inclusions in case of the HSV and ground-
the diagnosis as other microbiological studies or glass nuclei in the influenza virus. These findings
empirical treatment is instated based on the clini- could be present in the background of diffuse
cal picture and imaging finding. Only in cases that alveolar damage (DAD), and recognizing the viral
do not respond to antibiotics or because of other etiology or other etiologies in general is important
compounding factor like in immunocompromised in the treatment of these cases. Cytomegalovirus
patients, a biopsy is obtained, and the infectious induces only chronic inflammation and is charac-
process is evident through the presence of numer- terized by the enlarged cells with basophilic
ous neutrophils with or without a mucopurulent nuclear inclusions. The virus usually infects alveo-
exudate as in Fig. 4.1a, b. lar lining cells and endothelial cells.
On a small biopsy, the presence of acute inflam-
mation and/or necrosis is a fortuitous finding.
It tends to be sporadic or focal judging from the Acute Lung Injury Pattern
autopsy material. Usually, by the time a patient is
scheduled for a biopsy, the most common finding is The clinical definition of acute lung injury has
the organizing pneumonia which will be discussed evolved over the years. In its latest iteration, it is
later under the acute lung injury pattern. defined as the ratio of the partial pressure of the
Viral pneumonia could present in different arterial oxygen relative to that pressure in inspired
ways. Adenovirus infection could be subtle and air. A ratio of <300 is considered indicative of
shows only as dark smudgy nuclei in the alveolar acute lung injury, and a ratio <200 confirms respi-
ratory failure due to acute respiratory distress
syndrome (ARDS) [1].
M.M. Fraig, M.D. (*) From the histopathologic point of view, the
Department of Pathology and Laboratory Medicine,
acute lung injury pattern is one that is predomi-
School of Medicine, University of Louisville,
530 South Jackson Street, Louisville, KY 40202, USA nantly of organizing fibrosis mainly by fibroblastic
e-mail: m.fraig@louisville.edu proliferation and a recent myxoid background.

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 29


DOI 10.1007/978-1-4939-2575-9_4, © Springer Science+Business Media New York 2015
30 M.M. Fraig

Fig 4.1 Low magnification view of the fibrinopurulent pattern shows the neutrophilic infiltrate in the alveoli as
exudate in the alveolar space with thickening of the inter- well as in the interstitium in (b)
alveolar septa in (a). A higher magnification of the same

The fibrosis is of the recent type without dense


collagen deposition or distortion of lung architec- Bronchiolitis Obliterans/Organizing
ture as would be highlighted by a reticulin stain. Pneumonia
The main two entities of this pattern are DAD
which is the majority of cases of ARDS. The It is by far the most common pattern seen on a lung
idiopathic variant of DAD is acute interstitial biopsy. Several reasons account for that; BOOP
pneumonia (AIP) or Hamman-Rich disease. The could be encountered as a reaction to the primary
other entity is bronchiolitis obliterans/organizing lung injury. This would include post-infectious
pneumonia (BOOP) with its idiopathic variant pneumonia, inhalational injury, post radiation,
called cryptogenic organizing pneumonia (COP) aspiration pneumonia, and in the setting of colla-
[2]. There is a substantial overlap in the histo- gen vascular disease. In addition, BOOP could be
morphologic features between BOOP and DAD. present around mass lesions in the lung such as
In both of these entities, there are commonly: tumors or granulomas. In the latter situation, if
1. Thickening of the interalveolar septa with there is familiarity with imaging finding and if
mild chronic inflammatory infiltrate and fibro- there is suspicion for a mass lesion, it is important
blastic proliferation. The septa could also to communicate to the clinician that the presence
demonstrate intercellular edema exaggerating of BOOP could be a nonspecific finding, which
the thickness of the septa and making it appear does not account for the mass lesion seen on imag-
as a reticular pattern. ing. The other scenario where BOOP can be pres-
2. Proliferation of type II pneumocytes lining the ent is as a minor component of other disease
alveolar spaces. entities such as hypersensitivity pneumonia or
3. Proliferation of the fibroblasts forming polyp- nonspecific interstitial pneumonia [4].
oid and fascicular bundles filling up the alveo-
lar spaces to be called “fibroblastic plugs.”
The latter could plug the alveolar air spaces or Histopathologic Features
the bronchial lumens.
The lung will turn into a consolidated mass BOOP usually appears as polypoid structures
with sporadic air spaces in predominantly fibro- bulging into the air spaces in the peribronchial
elastic mass comprised by the thick septa and areas with a short stalk embedded in the intersti-
fibroblastic plugs [3]. This in turn hinders gas dif- tium (Fig. 4.2a, b). Some fascicles of elongated
fusion and hence results in the shortness of breath fibroblasts could be seen extended in parallel to
and hypoxia when measuring blood gases. the interalveolar septa. Using collagen stains
4 Airspace-Occupying Diseases 31

Fig. 4.2 Low power magnification of a small biopsy septa. The air spaces are markedly reduced by the infil-
showing the pale areas with the myxoid fibroblastic plugs trate (a). The polypoid plug with its stalk is shown with
and the cellular infiltrate in the thickened interalveolar swirling bands of fibroblasts (b)

Fig. 4.3 Another example of a polypoid fibroblastic plug filling up the alveolar space (a), and another one is almost
obliterating the bronchial lumen (b)

such as Movatt stain, the highlighted light green Whereas, in most cases, the histomorphologic
color of recent organizing fibrosis pattern appears features do not provide the underlying etiology of
as a tree of cauliflower extending throughout BOOP, it is important to not ignore some of the
the lung parenchyma in the area of biopsy [5]. easily recognizable etiologies. Finding a frothy
The polypoid masses used to be called “Masson’s material in the alveolar space in an immunocom-
bodies.” The interstitial cellular infiltrate is usu- promised patient would suggest Pneumocystis
ally comprised by a mixture of lymphocytes, jiroveci pneumonia. In addition, looking for viral
plasma cells, macrophages, and neutrophils inclusions or smudgy nuclei characteristic of
(Fig. 4.3a, b). The presence of lymphoid aggre- adenovirus infection would elicit the causing
gates or lymphoid follicles with germinal centers agent in some cases.
should raise the possibility of a more chronic pro- Sometimes, it is difficult to rule out DAD
cess such as hypersensitivity pneumonia (HP) or based on a small biopsy, and because of the sig-
collagen vascular disease. The presence of poorly nificant overlap between BOOP and DAD, an
formed granulomas and eosinophils would make umbrella term such as “acute lung injury pattern”
the first more likely, and the history and serologic may be a useful first step in characterizing the
testing would confirm the latter. lesion in the lung. DAD usually has an interstitial
32 M.M. Fraig

pattern with less fibroblastic plugs and more The list of potential causative agents is long and
fascicles of fibroblasts and of course the hallmark variable. The list includes infection, toxic inhal-
of hyaline membrane. These could be sporadic ants, drugs, radiation and chemotherapy, trauma
and not included in the small transbronchial and shock, burns, and certain ingested material
biopsy, but the use of acute lung injury pattern like kerosene.
could cover both entities as they share common Pathologically, the disease goes through two
therapeutic and management pathways. stages:
Another term that is usually confused with
BOOP is obliterative bronchiolitis or constric- Exudative phase: usually in the first week of pre-
tive bronchiolitis. Many in the lung transplant sentation and is characterized by presence of
community refer to it as “bronchiolitis obliter- patchy areas of pulmonary edema and plasma pro-
ans” which is the opposite of BOOP clinically teins in the alveolar spaces. There are mild or focal
and histopathologically. Obliterative bronchiol- areas of thickening of the interalveolar septa.
itis most commonly occurs as a late complica-
tion of transplant rejection and is mainly a small Organizing phase: usually sets in the second
airway disease. The lung parenchyma is almost week and usually when the patient is stable
normal, and the main problem is the presence of enough to be biopsied. They are usually the find-
an annular band of fibrosis constricting the ings discussed above. In later stages the hyaline
lumen of the small airways [6]. Movatt stain is membrane would start to disappear leaving
helpful in highlighting the fibrosis. behind a small fragment of fibrin or hyaline
Idiopathic BOOP is usually a problem on membrane embedded within the interstitium.
imaging and clinically. As the name suggests, This is a result of the sticky surfaces of opposing
there is no prior incident to suggest the presence hyaline membrane-coated alveolar linings fusing
of a sole area of consolidation or ground-glass together and taking with them the hyaline mem-
opacity in the lung, and suspicion of malignancy brane in what would appear as the interstitium. In
is in the differential diagnosis, especially for ade- this stage the presence of organized fibrin thrombi
nocarcinoma in situ, which used to be called in small blood vessels could be used in lieu of the
bronchioloalveolar carcinoma. This particular hyaline membrane to diagnose DAD.
type was recognized as a separate clinicopatho-
logic entity and given the name COP. However,
from the histopathologic standpoint, it is similar Histopathologic Features
in every aspect to BOOP [7].
The biopsy from a patient with DAD shares many
similarities with those from BOOP. However,
Diffuse Alveolar Damage and Adult certain points need to be understood to recognize
Respiratory Distress Syndrome the difference between these two entities:
1. DAD is a combination of an epithelial injury
DAD is characterized by an insidious onset and a as well as endothelial injury. As such, the
rapid course of respiratory failure requiring endothelial injury will result in leakage from
mechanical ventilation in most cases. In clinical the small vessels and the formation of intra-
terminology, this is recognized as ARDS. vascular thrombi, and plasma proteins will be
However, there are several etiologies that could present in the alveolar spaces. When the
result in ARDS but fail to show the characteristic degeneration and necrosis by-products mix
findings of DAD in histopathologic examination. with the fibrin from the plasma proteins, they
For instance, acute silicosis could result in mas- form a slurry-like material that paints the
sive pulmonary edema that results in ARDS, but alveolar lining layer as what we recognize as
the pathology of course is not that of DAD. All the hyaline membrane (Fig. 4.4a, b). The lat-
cases of DAD are considered part of ARDS but ter makes gas diffusion more difficult than
not all cases of ARDS would result in DAD. even in BOOP. For this and other reasons,
4 Airspace-Occupying Diseases 33

Fig. 4.4 Organizing diffuse alveolar damage with the (a). The well-defined hyaline membrane is lining the alveo-
loose myxoid background and diminished air spaces simi- lar spaces with a continuous undulating pattern and the
lar to that in BOOP without the polypoid fibroblastic plugs thick interalveolar septa with intercellular edema (b)

Fig. 4.5 An intravascular thrombus blocking the lumen of medium-sized blood vessels (a). The area distal to these
blood vessels showed pulmonary infarction with subsequent squamous metaplasia and atypical mitotic figures (b)

patients with DAD usually require mechanical necrotic debris could lead to an erroneous
ventilation and have worse prognosis than diagnosis of squamous cell carcinoma
those with BOOP [8]. (Fig. 4.5a, b).
2. The presence of intravascular thrombi in 3. Most patients with DAD get biopsied by the
larger vessels could lead to areas of lung second week of presentation where the orga-
infarctions. These could ultimately result in nizing phase is the predominant phase.
the squamous metaplasia with atypia. These However, some patients may be biopsied
areas could include abnormal mitotic figures earlier. In the latter case, pulmonary edema
and areas of necrosis. It is very important to and an eosinophilic material representing the
be very cautious in interpreting fine needle leaked plasma proteins could be copiously
aspiration material or bronchoalveolar lavages present without the characteristic hyaline
(BALs) from patients suspected of having membrane or the areas of fibroblastic prolif-
DAD or who are on ventilators. The metaplas- eration. Some authors called this “acute fibrin-
tic squamous epithelial cells along with the ous and organizing pneumonia” (AFOP) [9].
34 M.M. Fraig

Fig. 4.6 Low magnification view of the slightly thick- (a). A closer view shows the lack of hyaline membrane
ened interalveolar septa, lined by type II pneumocytes and and fibroblastic proliferation seen in the later organizing
showing an eosinophilic material in the alveolar spaces phase (b)

Some investigators thought it belongs to a


form of BOOP; others thought it represents an Intra-alveolar Hemorrhage
early phase of DAD (Fig. 4.6a, b). In practice
the distinction is not important as the treat- As a general rule the diagnosis of hemorrhage in
ment would follow the patient’s performance the lungs requires the presence of hemosiderin
status and the picture would be of an acute macrophages. The mere presence of blood within
lung injury pattern. the biopsy is not enough evidence for intra-
4. The idiopathic form of DAD is also a distinct alveolar hemorrhage as blood from the biopsy
entity called “acute interstitial pneumonia” procedure is usually the culprit for such
(AIP) or Hamman-Rich disease after the two hemorrhage.
physicians who first described the disease Intra-alveolar hemorrhage in its primary form
occurring in relatively young patients and is a diffuse process involving all areas of the
resulting in an accelerated course of respiratory lung. The potential etiology is usually an autoim-
failure and a characteristic form of fibrosis on mune disease such as Goodpasture syndrome
autopsy. [11]. In this case a small portion of the biopsy
5. DAD could be superimposed on cases of usual should be snap frozen for immunofluorescence
interstitial pneumonia (UIP), and in that case it studies. Other common causes would include
is called “accelerated phase of UIP.” If the vasculitides such as capillaritis syndrome in
patient has been previously diagnosed with microscopic polyangiitis and Henoch-Schonlein
UIP or there is a radiologic evidence of UIP as purpura. Other primary diseases are associated
would be discussed later, the presence of a with toxic inhalants and drugs. An idiopathic
DAD picture along with the honeycomb form also exists.
changes in the lung would point to that diagno- In Goodpasture syndrome, there is an associ-
sis. If there are several biopsies from different ated renal lesion, and immunofluorescence with
lobes, one might show UIP without DAD [10]. linear deposits along the basement membrane is
6. A subset of patients with AIP may come with diagnostic. There is a marked accumulation of
a protracted history of respiratory symptoms hemosiderin-laden macrophages with their char-
that does require mechanical ventilation only acteristic chunky refractile material with an
to show histopathologic evidence of DAD on orange or brown discoloration. They can also be
the biopsy. deposited within the lung parenchyma. Iron stain
4 Airspace-Occupying Diseases 35

Fig. 4.7 Intra-alveolar hemorrhage with hemosiderin- lymphocytic infiltrate of this case of lymphatoid granulo-
laden macrophages containing chunky crystals brown to matosis with concomitant fibrin deposits in the alveoli.
yellow hemosiderin (a). In a case of vasculitis, there are Notice the lack of neutrophilic infiltrate (b)
leaking vessels due to inflammation of the wall by the

usually is not needed to highlight hemosiderin as


it is easy to identify on H&E stain (Fig. 4.7a, b). Pulmonary Alveolar Proteinosis
In cases of a vasculitis, a similar picture is
present with the additional finding of fibrin in the Pulmonary alveolar proteinosis (PAP) is a rare
alveolar spaces with the evidence of the vasculitis disease that could be idiopathic, secondary, or
also present in the blood vessels. congenital. In most cases the idiopathic or
In cases of idiopathic pulmonary hemosidero- acquired form affects middle-age adults in 90 %
sis, the diagnosis is by exclusion as in any idio- of the cases. The secondary form (5–10 % of
pathic case. The chronic bleeding may lead the cases) is mainly due to inhalational exposure to
marked deposits of hemosiderin to encrust the industrial material such as silica, cement dust,
medial wall of the blood vessels resulting in what aluminum, or titanium oxide. The rare congenital
is called “endogenous pneumoconiosis.” form affects children in the neonatal period.
In secondary alveolar hemorrhage, the causes There is a controversy as whether it is a true PAP
include infections especially necrotizing ones. condition; however, the prognosis is invariably
Systemic diseases that affect the coagulation poor. The etiology of the primary type is poorly
mechanisms such thrombocytopenia and coagu- understood. It is thought to be a mutation that
lation defects, uremia, and renal failure are the affects the function of GM-CSF, an important
most common ones. factor in surfactant homeostasis and an essential
Localized hemorrhage could be the result of a regulator in immune defense [12]. An increased
focal lesion or disease that causes bleeding in one level of GM-CSF autoantibodies was found in
or several small blood vessels. This includes vaso- patients with primary PAP. Most patients present
invasive fungi such as aspergillus or narcotizing with dyspnea and nonproductive cough. Less
tumors. The bronchial wall could have lesions common symptoms include low fever, weight
eroding through the wall and the accompanying loss, fatigue, and chest pain.
vessel. The significance of identifying these Imaging studies show areas of lung opacities
lesions is to justify the resection that could be per- sparing the apices and costophrenic angles on a
formed to stop the bleeding. Sometimes the chest X-ray. On CT scan the characteristic
pathologist is left with this task after the operation “crazy-paving” pattern is recognized. It is a com-
is performed, and if there is no lesion or an obvious bination of ground-glass opacities with reticular
culprit, the fear is that it could happen again. (septal) lines that involve any area of the lung but
36 M.M. Fraig

Fig. 4.8 General view of PAP showing the characteristic of the D-PAS-positive globules in PAP (b). Contrasting
amorphous material in the alveolar space with needle- view of pulmonary edema with a homogeneous material
shaped cholesterol clefts (a). A higher magnification view and several macrophages floating in the fluid (c)

tend to spare the costophrenic angles. Even The main differential diagnosis is with pulmo-
though the CT findings are highly suggestive, nary edema where the edema leaves little strands
they are still nonspecific, and a lung biopsy is of proteins or faint powdery material. Plasma
needed to confirm the diagnosis. proteins leaking in the alveolar space assume a
homogeneous eosinophilic material that shows
scalloping near the alveolar walls. The D-PAS-
Histomorphologic Features positive globules are also absent by special stains.
Another differential is with the frothy material
An eosinophilic amorphous material with choles- seen in Pneumocystis jiroveci. The latter is an
terol needle-shaped clefts is seen filling the alve- infection in immunocompromised patients and as
oli. The alveolar walls are not usually affected by such usually shows widening of the interalveolar
the process, but there could be a focal increase in septa, and the material is frothy rather than gran-
the wall thickness that is filled by an inflamma- ular with no cholesterol clefts or D-PAS-positive
tory infiltrate. Small eosinophilic globules that globules.
stain positive with periodic acid-Schiff stain with The treatment in these cases usually is by
diastase digestion are also present (Fig. 4.8a–c). repeated BAL. The prognosis is usually good and
The interlobular septa are also widened and show few patients might need to have a repeat BAL
abnormally dilated lymphatics. within few months.
4 Airspace-Occupying Diseases 37

Pneumoconiosis References

Most of pneumoconiosis results in an interstitial 1. Tsushima K, King LS, Aggarwal NR, De Gorordo A,
D’Alessio FR, Kubo K. Acute lung injury review.
disease. The old entity of giant cell interstitial
Intern Med. 2009;48:621–30.
pneumonia or GIP was considered part of the 2. Cordier JF. Organising pneumonia. Thorax. 2000;55:
idiopathic interstitial pneumonia till it was found 318–28.
to harbor many of the exogenous materials from 3. Epler GR. Bronchiolitis obliterans organizing pneu-
monia: definition and clinical features. Chest. 1992;
industrial exposure. However, some of these
102:2S–6S.
agents can manifest in the alveolar spaces as 4. Epler GR, Colby TV. The spectrum of bronchiolitis
numerous macrophages and giant cells contain- obliterans. Chest. 1983;83:161–2.
ing the offending agent. The determination of the 5. Capdevila E, Banus E, Domingo C, Ferrer A, Mata JM,
Marin A. [Bronchiolitis obliterans organizing pneumo-
agent requires careful clinical history and tissue
nia: the usefulness of the transbronchial biopsy as a
analysis of crystals and other materials in spe- diagnostic technic]. An Med Interna. 1994;11:449–51.
cialized laboratories. 6. Myers JL, Colby TV. Pathologic manifestations of
bronchiolitis, constrictive bronchiolitis, cryptogenic
organizing pneumonia, and diffuse panbronchiolitis.
Clin Chest Med. 1993;14:611–22.
Pulmonary Edema 7. Azzam ZS, Bentur L, Rubin AH, Ben-Izhak O, Alroy
G. Bronchiolitis obliterans organizing pneumonia.
Pulmonary edema is a common finding in hospi- Diagnosis by transbronchial biopsy. Chest. 1993;104:
1899–901.
talized patients who undergo intravenous infu-
8. Blennerhassett JB. Shock lung and diffuse alveolar
sion, and they end up with circulatory overload. damage pathological and pathogenetic considerations.
Patients with congestive heart failure, hypopro- Pathology. 1985;17:239–47.
teinemia, or cirrhosis are prone to have pulmo- 9. Beasley MB, Franks TJ, Galvin JR, Gochuico B,
Travis WD. Acute fibrinous and organizing pneumo-
nary edema. The edema fluid can be serous with
nia: a histological pattern of lung injury and possible
very few solutes or proteins, and this will appear variant of diffuse alveolar damage. Arch Pathol Lab
as thinly reticular mesh or crisscrossing strands Med. 2002;126:1064–70.
or powdery material in the alveolar spaces. 10. Kaarteenaho R, Kinnula VL. Diffuse alveolar damage:
a common phenomenon in progressive interstitial
If plasma proteins are admixed, they appear as a
lung disorders. Pulm Med. 2011;2011:531302.
homogenous eosinophilic material. Corpora 11. Boyce NW, Holdsworth SR. Pulmonary manifestations
amylacea, which are concentrically lamellated of the clinical syndrome of acute glomerulonephritis
eosinophilic bodies in the alveolar spaces, are and lung hemorrhage. Am J kidney Dis. 1986;8:31–6.
12. Frazier AA, Franks TJ, Cooke EO, Mohammed TL,
usually seen especially in older patients. They are
Pugatch RD, Galvin JR. From the archives of the
nonspecific findings in those patients and in AFIP: pulmonary alveolar proteinosis. Radiographics.
autopsy cases. 2008;28:883–99; quiz 915.
Interstitial Lung Diseases in Small
Lung Biopsies 5
Mostafa M. Fraig

diseases. BIP was also removed because it related


Interstitial Lung Diseases to the acute lung injury pattern which eventually
resolves and leaves no residual effects on the lung
Liebow and Carrington through their work in the (Table 5.2).
1970s were the first to classify and organize the The most recent attempt in classifying these
entity of interstitial lung disease [4]. Their land- entities has been through the combined effort of
mark work resulted in a classification schema that the American Thoracic Society and the European
recognized usual interstitial pneumonia (UIP) as a Respiratory Society with two schemas in almost
distinct entity and not just a common pathway to a decade [5–7].
end-stage lung disease. In addition they identified As more knowledge accumulated, the classifi-
desquamative interstitial pneumonia (DIP) as cation became more refined, and criteria for diag-
another entity even though the name was found to nosis and subset of patients were recognized as
be incorrect later on. They added lymphocytic and different in prognosis and response to treatment.
giant cell interstitial pneumonias (LIP and GIP, For example, patients with UIP associated with
respectively) to the category. These were found to CVDs were found to represent a subset that fares
represent cases of low-grade lymphoma in the better than the idiopathic UIP one [8]. The use of
case of the first and cases of pneumoconiosis in high-resolution CT and the experience of radiol-
the latter. The entity of bronchiolitis obliterans ogist in diagnosing UIP made the requirement for
interstitial pneumonia (BIP) was added as it surgical biopsy in many cases redundant.
represented to them an acute phase of the other
fibrosing conditions (Table 5.1).
Later, Katzenstein and Askin refined this clas- Clinical and Radiological Findings
sification and added other entities such as nonspe- of UIP
cific interstitial pneumonia (NSIP) and respiratory
bronchiolitis-interstitial lung disease (RB-ILD). It is important before looking at the biopsy from
LIP and GIP were removed from the classifica- interstitial lung disease to be familiar with the
tion as they were considered no longer idiopathic clinical and radiological findings. UIP usually
presents in the fifth and sixth decades of life with
an insidious onset of progressive dyspnea and
M.M. Fraig, M.D. (*) shortness of breath. Patients have bilateral crack-
Department of Pathology and Laboratory Medicine,
School of Medicine, University of Louisville,
les on auscultation with a characteristic “Velcro
530 South Jackson Street, Louisville, KY 40202, USA type.” The pulmonary function tests (PFTs) have
e-mail: m.fraig@louisville.edu a restrictive pattern.

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 39


DOI 10.1007/978-1-4939-2575-9_5, © Springer Science+Business Media New York 2015
40 M.M. Fraig

Table 5.1 Liebow classification of interstitial lung


disease, 1975
Usual interstitial pneumonia (UIP)
Bronchiolitis obliterans interstitial pneumonia (BIP)
Desquamative interstitial pneumonia (DIP)
Lymphoid interstitial pneumonia (LIP)
Giant cell interstitial pneumonia (GIP)

Table 5.2 Current classification by Katzenstein, 1995


Usual interstitial pneumonia (UIP)
Desquamative interstitial pneumonia (DIP)
Acute interstitial pneumonia (AIP)
Nonspecific interstitial pneumonia (NSIP)
Fig. 5.1 Patch fibrosis with broad areas with collagen
deposition alternating with areas with reticular fibrosis in
Radiologically, by high-resolution CT, there is UIP. In the center there is a fibroblastic focus
a bilateral fibrosis accentuated in the lower por-
tion of the lung and in the lung periphery with
occasional honeycomb pattern in those areas.
Ground-glass opacities are frequent findings in
all of interstitial lung disease. The picture could
be so characteristic that along with the clinical
findings, in half the cases the diagnosis could be
made without obtaining a biopsy.

Histopathologic Findings

Open lung biopsy used to be the gold standard for


diagnosing idiopathic interstitial disease. In this
biopsy several areas of the lung would be sam- Fig. 5.2 The fibroblastic focus is under higher magnifica-
pled preferably with a sample from each lobe tar- tion showing the streaming fibroblasts in a myxoid back-
geting involved areas guided by the CT scan ground. The focus is only slightly bulging from the
interstitium and is next to an area with dense fibrosis, in
image. In the affected areas there is a patchy contrast to the polypoid fibroblastic plugs seen in BOOP
process of fibrosis and mild to moderate chronic
inflammatory infiltrate. UIP could be called
“peripheral fibrosis of the lung.” The process is a feature that helps differentiating it from the
usually more pronounced in the periphery of the fibroblastic plugs of cryptogenic organizing
lung with honeycomb changes in those areas and pneumonia in which the age of fibrosis is uniform
also in the periphery of the lung lobules with the and plugs assume a polypoid configuration
center of the lobule less affected by fibrosis than (Fig. 5.2). The inflammation is never severe, but
the periphery. The fibrosis takes place by dense lymphoid aggregates could be present, and when
collagen deposition with a mild inflammation there are several of them, a possibility of UIP
within those areas (Fig. 5.1). Fibroblastic foci are associated with CVD should be raised to be con-
areas of recent fibrosis that show a recent type of firmed by serological testing. The latter group of
fibrosis with myxoid background and small patients usually has a better prognosis than the
fascicles of fibroblasts within the interstitium one with idiopathic UIP [8]. Features that indi-
juxtaposed immediately to areas of dense fibrosis, cate chronic irritation of the lung could be seen in
5 Interstitial Lung Diseases in Small Lung Biopsies 41

Table 5.3 Revised American Thoracic Society/European


Respiratory Society classification of idiopathic interstitial
pneumonias: multidisciplinary diagnoses [7]
Major idiopathic interstitial pneumonias
Idiopathic pulmonary fibrosis
Idiopathic nonspecific interstitial pneumonia
Respiratory bronchiolitis-interstitial lung disease
Desquamative interstitial pneumonia
Cryptogenic organizing pneumonia
Acute interstitial pneumonia
Rare idiopathic interstitial pneumonias
Idiopathic lymphoid interstitial pneumonia
Idiopathic pleuroparenchymal fibroelastosis
Unclassifiable idiopathic interstitial pneumonias
Fig. 5.3 An area of bronchial metaplasia with many alve-
olar spaces lined by columnar epithelium and inspissated
mucous filling bronchial lumens An important caveat to the diagnosis is the
location of the biopsy as the tips of the lobes are
notorious for harboring many of the features of
chronic irritation of the lung with a sudden transi-
tion to the completely normal lung away from the
very tip of the lobe. Caution should be exercised
and correlation with high-resolution CT should
be performed.
Recently, several reports of the possibility of
making the diagnosis of UIP from transbronchial
biopsy have surfaced [1]. However, the diagnos-
tic yield of such biopsies is still low (specificity
of 26–32 %) [3]. Another promising type of
biopsy is the cryobiopsy which offers better size
and less artifacts to make the diagnosis more reli-
able [3].
Fig. 5.4 An area with honeycomb changes under the
In the latest ATS/ERS schema for the diagno-
pleural surface which corresponds to areas of accentuated
fibrosis in the periphery of the lungs on high-resolution sis of UIP, the histopathologic findings were
CT scan images divided to definite, probable, possible, and not
UIP based on the presence or absence of certain
criteria summarized in Table 5.3.
the same setting. These would include type II
pneumocytes proliferation, bronchial metaplasia,
and smooth muscle cirrhosis (Fig. 5.3). Clinical Course
Honeycomb changes representative of those seen
on CT are also present in the periphery under the Many UIP patients succumb to the condition
pleura (Fig. 5.4). Foamy and pigmented macro- known as “accelerated phase of UIP” where the
phages could be seen in the alveolar spaces, but patients go quickly into respiratory failure requir-
their significance and contribution to the diagno- ing mechanical ventilation. Histologically, the
sis are limited. Rare granulomas or other inciden- findings are similar to organizing diffuse alveolar
tal findings could be seen, but their significance damage superimposed or alternating with areas
and detraction from the diagnosis should be of characteristic UIP pattern. Hyaline membrane
weighed against the other evidence of UIP. formation and fibroblastic proliferation along
42 M.M. Fraig

with edema and fibrin deposits are present along


with interstitial dense fibrosis and evidence of
chronic lung irritation.
Unfortunately, UIP presents a problem for
understanding the underlying pathogenesis and
also treatment. Immunosuppressive drugs failed
to make any significant difference in the lives of
these patients. Just recently, there are reports that
pirfenidone, an anti-fibrogenic drug, shows some
promising results in clinical trials [9]. Lung
transplant is currently the only available hope for
these patients.

Fig. 5.5 Uniform fibrosis and moderate interstitial inflam-


Nonspecific Interstitial Pneumonia mation with uniform age of the fibrosis are all criteria for the
diagnosis of nonspecific interstitial pneumonia (NSIP)
It used to be reserved for non-classifiable cases
that did not quite fit any of the idiopathic intersti-
tial pneumonia category. However, in a landmark
paper, Katzenstein et al. after analyzing a cohort
of these cases reached to the conclusion that they
represent a distinct group from UIP and other
entities, even though they share similar features
with UIP patients [10, 11].

Clinical and Radiological Findings


of NSIP

These patients are usually a decade younger than


those of UIP, and the male to female ratio is almost
equal. They usually present with an insidious onset Fig. 5.6 NSIP showing the same type of uniform fibrosis
and areas with markedly cellular infiltrate that lack the
of dyspnea and shortness of breath. They have the variegated pattern and fibroblastic foci seen in UIP
same pattern of restrictive changes on the PFTs.
Radiologically, there is a reticular pattern of fibro-
sis and ground-glass opacities on high-resolution Histopathologic Features
CT. The characteristic peripheral attenuation seen
in UIP is usually lacking, and honeycomb changes The main distinguishing feature of NSIP from
are not a standard feature. The picture is usually UIP, an important and crucial differential diagno-
more prominent in the lower lobes though, which sis, is the temporal and architectural uniformity of
helps in the differential diagnosis with hypersensi- the fibrosis. There is a thickening of interalveolar
tivity pneumonia [12]. septa by increased fibrosis and a chronic inflam-
There is a frequent association with CVD, matory infiltrate (Fig. 5.5). The amount of the lat-
especially rheumatoid arthritis and scleroderma. ter could range from marked to sparse. Based on
NSIP could be the presenting disease in the lung that, the NSIP patterns could be divided into cel-
of CVD. The prognosis of NSIP is much better lular NSIP on one extreme and fibrotic NSIP with
than that of UIP with an overall 5-year survival a possible third pattern of mixed NSIP when
between 75 and 90 % [11]. there are alternating patterns of both (Fig. 5.6).
5 Interstitial Lung Diseases in Small Lung Biopsies 43

later, and the presence of macrophages, whether


foamy or pigmented ones, should not detract
from the diagnosis.

Desquamative Interstitial
Pneumonia and Respiratory
Bronchiolitis-Interstitial Lung
Disease

In the original classification of Liebow, DIP was


thought of as a disease where the epithelial cells
slough off and fill the alveolar spaces and hence
Fig. 5.7 NSIP with cellular interstitial chronic inflamma- the name, DIP. However, later studies revealed
tion and uniform fibrosis these cells to be pigmented macrophages from
smoking. The close association with smoking
The fibrotic pattern imparts a worse prognosis history made this disease and others like it more
and has been lumped with cases of UIP in the smoking related rather than idiopathic pneumo-
past. The key difference from UIP is the lack of nia. Respiratory bronchiolitis (RB) is a disease
fibroblastic foci and the lack of variegated pattern with few symptoms that was reported in smokers
of fibrosis (Fig. 5.7). Microscopic foci of BOOP based on an autopsy study by Niewoehner et al.
could be encountered in the biopsy, and these [14]. These cases were reported to have a peri-
should not draw away from the diagnosis. The bronchial pattern of fibrosis with the accumula-
presence of granulomas or marked eosinophilia tion of pigmented macrophages in the same
should point to other granulomatous diseases areas, which is different from the diffuse pattern
instead of NSIP. seen on imaging and on biopsies of DIP. Patients
with RB did not have their PFTs measured, and
the presence of clinical evidence of interstitial
Clinical Course lung disease was not documented. Recognition of
the occurrence of restrictive pattern of changes
While many clinicians wondered about the clini- on PFTs led to the designation of this subset of
cal entity of NSIP from the standpoint of clinical patient as RB-ILD, a diagnosis that would require
presentation, the fact remains that it is mostly a the performance of PFTs and knowledge of the
treatable disease and has a good response to cor- restrictive pattern. It was thought then that the
ticosteroids, and it pays to distinguish it from two entities, DIP and RB-ILD, represented two
other idiopathic interstitial pneumonias, espe- ends of the spectrum. The presence or absence of
cially UIP. As to what NSIP really represent, sev- the peribronchial pattern of fibrosis became
eral speculations were put forward, including a almost irrelevant in making the diagnosis of
possibility of CVD presenting in the lung with RB-ILD. Some pathologists shied away from
these manifestations in this group of patients, making this diagnosis without knowing this
similar with what these diseases present in the information which in most cases is not provided
kidney. Another possibility is that some cases with the biopsy. However, there is more overlap
could represent cases of hypersensitivity pneu- in the clinical presentation, radiologic imaging,
monia where the insulting antigen has not been and histopathologic features that makes the dis-
identified. Another possibilities are cases of tinction between the two entities almost arbitrary.
slowly resolving BOOP or acute pneumonia and DIP with the classic marked fibrosis described in
lastly cases of inadequately sampled UIP [13]. the original reports is more akin to NSIP than to
The entity is expanding, as will be discussed RB-ILD [15]. The reason of this distinction is the
44 M.M. Fraig

effect of cessation of smoking and corticosteroid


therapy on one group that might not be enough to
alter the disease in the other one. Another obser-
vation is the presence of pigmented macrophages
in sheets whenever the lungs are fibrosed for any
other reason, like in cases of asbestos exposure or
bleomycin treatment, which could result in a sec-
ondary phenomenon of DIP-like reaction (see
below) in the setting of fibrosis and not the pri-
mary disease.

Clinical Presentation of RB-ILD


and Smoking-Related Diseases Fig. 5.8 RB-ILD showing clusters of lightly pigmented
macrophages in the alveolar spaces. There is air duct in
Dyspnea and shortness of breath with dry cough the center assuming a slightly curved line and an inter-
lobular septum on the right side
are the most common symptoms in these patients.
The history of smoking is very common in
90–100 % of cases. Males are more affected than
females and the mean age is a decade younger
that of UIP. As in all interstitial lung diseases,
there are restrictive changes on the PFTs.
Radiologically, there are diffuse ground-glass
opacities with fine reticular fibrosis in the lungs.
The lung images could be normal in some cases.
Usual emphysematous changes encountered in
smokers are not seen in these patients, so air
trapping and cyst formation are not features of
the disease.
Fig. 5.9 A loose cluster of pigmented macrophages is in
the alveolar space. Notice the mild fibrosis in the interal-
Histopathologic Findings veolar septa

The diagnosis of RB-ILD and DIP has been con- differentiated on routine stains from the
troversial as was outlined in the introduction. The hemosiderin-laden macrophages of alveolar
degree of fibrosis is an important parameter. In hemorrhage where the cytoplasm contains a
RB-ILD, mild fibrosis should be used as one chunky refractile material. In fact, iron stain can
important criterion of the diagnosis as cases with be confusing as it would be positive in both types
marked and broad fibrosis or interstitial scarring of macrophages with only the degree of positivity
are not going to improve unless these are spo- separating the two entities. The macrophages
radic and constitute as a small portion of the lung should be in every alveolar space.
(Fig. 5.8). The pigmented macrophages are usu- A newly described entity called “smoking-
ally dispersed in the alveolar spaces and only related interstitial fibrosis (SRIF)” that was
small clusters are noted. Small sheets of these described in patients with resected lung for lung
macrophages can be seen under the pleural sur- cancer is similar to RB-ILD except for a few dif-
face. The macrophages are usually tan or light ferences which include fibrotic interstitial scar-
brown and contain small dark powdery particles ring and the presence of emphysematous changes
in the cytoplasm (Fig. 5.9). They can be easily (Fig. 5.10) [16]. Since these patients presented
5 Interstitial Lung Diseases in Small Lung Biopsies 45

Fig. 5.11 A collection of abnormal histiocytes with convo-


luted nuclei and pale cytoplasm admixed with lymphocytes,
plasma cells, and eosinophils in the background. This is the
hallmark of Langerhans cell histiocytosis (LCH)
Fig. 5.10 An area of fibrosis and chronic inflammation in
the interstitium with clusters and single pigmented macro-
phages. Torn interalveolar septa with terminal clubbing focal obstruction of the lung due to the presence
consistent with emphysematous changes can be seen. This of mass lesion like a large granuloma or tumor
latter finding is not seen in RB-ILD or DIP. It is a feature
of smoking-related interstitial fibrosis (SRIF)
masses. Areas distal to these masses usually have
post-obstructive pneumonia with the accumulation
of foamy as well as pigmented macrophages.
with their lung cancer signs and symptoms, it is It could also occur whenever the structure of
hard to identify any distinguishing clinical pic- the lung is stiffening by fibrosis and the lung
ture to expect in patients with this condition. loses its elasticity.
Other investigators are calling the entity
“smoking-related interstitial lung disease
(SR-ILD)” or “combined pulmonary fibrosis and Acute Interstitial Pneumonia
emphysema syndrome” [17]. The presence of
nodules on radiologic imaging and in the biopsy Acute interstitial pneumonia (AIP) is similar in
should raise the possibility of Langerhans cell many aspects to DAD. It is also similar to the
histiocytosis (LCH), which presents in the same condition described by both Hamman and Rich.
population of patients but at a younger age and So the name Hamman and Rich disease is syn-
with spontaneous pneumothorax. On examining onymous with AIP. The only differentiating fac-
the histologic sections, there is a characteristic tor from DAD is that AIP is idiopathic and there
stellate shaped lesion with a collection pale areas is no known underlying etiology.
containing the abnormal histiocytes that stain
positive by CD1a immunostain (Fig. 5.11).
Clinical Presentation of AIP

DIP-Like Reaction Rapid respiratory failure unexplained by any pre-


cipitating factor is the most common presentation.
An important reason to correlate with the CT Patients usually require mechanical ventilation
findings is to avoid overcalling areas with excess and supportive measures. Imaging would show
pigmented macrophages, which are usually local- diffuse ground-glass opacities and consolidation
ized, as one of the smoking-related diseases. of the lungs. The patients could be of any age.
This phenomenon is usually associated with The prognosis is similar to that of DAD and is
46 M.M. Fraig

Fig. 5.12 Widening of the interalveolar septa with fibro- Fig. 5.13 Hyaline membrane is lining the alveolar space
blastic proliferation in the background of myxoid matrix in AIP and plump type II pneumocytes next to it
is a feature seen in acute interstitial pneumonia (AIP).
There is also type II pneumocyte proliferation indicating
an epithelial injury

variable from one series to another. The treat-


ment is by supportive measures and corticoste-
roid therapy. The mortality rate is between 40 and
70 % within a few months. The disease could
recur in those who survive the first episode.

Pathologic Findings
Fig. 5.14 Areas of squamous metaplasia in AIP. Caution
The findings are very similar to those from orga-
should be exercised in interpreting these cells in the bron-
nizing DAD. Typically, there is widening of the choalveolar lavage from these patients as an erroneous
interalveolar septa by recent fibroblastic prolifera- diagnosis of malignancy could be rendered
tion and moderate chronic inflammatory infiltrate
(Fig. 5.12). The hyaline membrane can be noted
coating the alveolar walls (Fig. 5.13). Remnants Hypersensitivity Pneumonia (HP)
of the hyaline membrane could be seen dispersed
within the interstitium. Intravascular thrombi HP is a very common differential diagnosis with
could be seen at different stages of organization the entities included in idiopathic ILD. There are
with areas of squamous metaplasia present wher- several clinical similarities as well as radiological
ever intermediate blood vessels are involved and pathologic overlapping features that warrant
(Fig. 5.14). In general, the picture is that of an a discussion here. The disease is also known as
acute lung injury and is different from that of UIP extrinsic allergic alveolitis and is immunologi-
or NSIP. There is no dense fibrosis and areas simi- cally mediated. The response is both humoral and
lar to BOOP are more dominant than those in cell mediated. Several antigens and agents present
NSIP. The presence of hyaline membrane and its a potential to invoke such reaction with differing
remnants is diagnostic. potencies. Many of these have been characterized
5 Interstitial Lung Diseases in Small Lung Biopsies 47

over the decades and recognized by their specific


names from the bird fancier’s lung, farmer’s lung,
humidifier’s lung, and several others.

Clinical Presentation

HP can present in an acute, subacute, or chronic


form depending on the amount and duration of
antigen exposure. In the acute form the patient
would suffer an acute onset of severe dyspnea,
high fever, and chills that will subside within 12 h.
On imaging there will be ground-glass opacities
and small nodules. As the relationship between Fig. 5.15 Low-magnification view of hypersensitivity
the exposure and the symptoms is clearly evident, pneumonia (HP) showing interstitial fibrosis and marked
these patients will be diagnosed clinically and cellular infiltrate with occasional lymphoid aggregates
rarely that a biopsy would be needed.
In the subacute and chronic forms, patients are in the mix. Epithelioid histiocytes could be present
exposed to smaller amounts of antigen over a in aggregates, and lymphoid follicles with germi-
more protracted period of time. The subacute con- nal centers are frequently present. Poorly formed
dition lasts for weeks to months where the chronic granulomas with multinucleated giant cells are
one is a prolonged process and takes years with present usually near or around the bronchial wall
more prominent changes resulting in an irrevers- (Fig. 5.15). Areas of BOOP are also present, and
ible damage to the lungs. Sometimes, there is no these create a mixed picture of acute and chronic
clear relationship to an antigen, and a lung biopsy lung injury that could be confused with UIP or
provides the first clue to the diagnosis. On high- accelerated phase of UIP. However, the marked
resolution CT imaging, there are ground-glass cellularity and absence of patchy fibrosis and
opacities and linear attenuation with traction fibroblastic foci should help rule that out. In addi-
bronchiectasis and reticular fibrosis indicating tion the presence of granulomas should rule out
honeycomb changes. There will be small nodules idiopathic interstitial lung disease altogether
corresponding to lymphoid aggregates scattered (Fig. 5.16a, b). Difficult cases can present as
in the mid to upper portion of the lungs [18]. This fibrosing HP where there are more pronounced
picture is very important in the differential diag- fibrosis and features of chronic irritation of the
nosis with both UIP and NSIP where the disease lung such as bronchial metaplasia and smooth
usually dominates in the lower lobes and there are muscle cirrhosis with less cellular infiltrate.
no nodules. However, a significant overlap could The presence of poorly formed granulomas and
occur, and because of the chronic nature of the frequent lymphoid follicle could help in the dif-
disease, HP is the most common disease in the ferential diagnosis [20].
differential diagnosis of UIP and NSIP [19].

Lymphoid Interstitial Pneumonia


Histopathologic Findings
This is a controversial entity as some authors
The changes follow the route of the antigen to consider these cases to belong to NSIP if they are
have a bronchocentric pattern. There are areas of not part of the low-grade lymphoproliferative
fibrosis with marked cellular infiltrate by lym- disorders. Others think some of these cases repre-
phocytes and plasma cells and some eosinophils sent a separate entity as in the case of patients
48 M.M. Fraig

Fig. 5.16 (a) A poorly formed granuloma from a case of from a bird fancier’s lung surrounded by a foreign-body
HP showing histiocytes and multinucleated giant cells, giant cell reaction
but no foreign material is seen. (b) A foreign material

with Sjogren’s syndrome. Diffuse lymphocytosis The more information is available, the better the
and lymphoid follicles with germinal centers in chance for a proper diagnosis. Teamwork is impor-
the background of areas with broad fibrosis and tant, and switching venues with scattering infor-
dense collagen deposition are the hallmarks of mation in different institutions and with different
the disease [21]. care providers should be discouraged.

Summary References
Idiopathic interstitial lung diseases are a group of 1. Berbescu EA, Katzenstein AL, Snow JL, Zisman
diseases with different outcomes and with signifi- DA. Transbronchial biopsy in usual interstitial pneu-
cant management differences. The integration of monia. Chest. 2006;129(5):1126–31.
2. Tomassetti S, Cavazza A, Colby TV, Ryu JH, Nanni
the clinical, radiological, and pathologic findings
O, Scarpi E, et al. Transbronchial biopsy is useful in
is crucial to making the diagnosis and avoiding predicting UIP pattern. Respir Res. 2012;13:96.
diagnostic pitfalls. The role of smaller biopsy 3. Casoni GL, Tomassetti S, Cavazza A, Colby TV,
especially cryobiopsy is evolving and might Dubini A, Ryu JH, et al. Transbronchial lung cryobi-
opsy in the diagnosis of fibrotic interstitial lung dis-
increase the number of biopsies as a tool not only
eases. PLoS One. 2014;9(2):e86716.
to establish the diagnosis but also to follow up on 4. Carrington CB, Gaensler EA, Coutu RE, Fitzgerald
the effect of treatment. Under those circumstances, MX, Gupta RG. Usual and desquamative interstitial
it is important to use all the information available pneumonia. Chest. 1976;69(2 Suppl):261–3.
5. Demedts M, Costabel U. ATS/ERS international mul-
to try to reach a definitive diagnosis. However, in
tidisciplinary consensus classification of the idio-
the absence of information, applying a strict set of pathic interstitial pneumonias. Eur Respir J. 2002;
criteria in making the diagnosis should be fol- 19(5):794–6.
lowed. There are difficult cases and even with all 6. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J,
Brown KK, et al. An official ATS/ERS/JRS/ALAT
available data, are still hard to categorize. In those
statement: idiopathic pulmonary fibrosis: evidence-
cases, a descriptive diagnosis is warranted, and an based guidelines for diagnosis and management. Am J
explanatory note as to what is present and what is Respir Crit Care Med. 2011;183(6):788–824.
absent and where the pathologist is standing from 7. Travis WD, Costabel U, Hansell DM, King Jr TE,
Lynch DA, Nicholson AG, et al. An official American
all the possible diagnoses could be helpful. In
Thoracic Society/European Respiratory Society
many cases it is an evolving process, and the dis- statement: update of the international multidisci-
ease might manifest itself better on follow-up. plinary classification of the idiopathic interstitial
5 Interstitial Lung Diseases in Small Lung Biopsies 49

pneumonias. Am J Respir Crit Care Med. 2013; 15. Liebow AA, Steer A, Billingsley JG. Desquamative
188(6):733–48. interstitial pneumonia. Am J Med. 1965;39:369–404.
8. Park JH, Kim DS, Park IN, Jang SJ, Kitaichi M, 16. Katzenstein AL, Mukhopadhyay S, Zanardi C, Dexter
Nicholson AG, et al. Prognosis of fibrotic interstitial E. Clinically occult interstitial fibrosis in smokers:
pneumonia: idiopathic versus collagen vascular classification and significance of a surprisingly com-
disease-related subtypes. Am J Respir Crit Care Med. mon finding in lobectomy specimens. Hum Pathol.
2007;175(7):705–11. 2010;41(3):316–25.
9. King Jr TE, Bradford WZ, Castro-Bernardini S, 17. Portillo K, Morera J. Combined pulmonary fibrosis
Fagan EA, Glaspole I, Glassberg MK, et al. A phase and emphysema syndrome: a new phenotype within
3 trial of pirfenidone in patients with idiopathic pul- the spectrum of smoking-related interstitial lung dis-
monary fibrosis. N Engl J Med. 2014;370(22): ease. Pulm Med. 2012;2012:867870.
2083–92. 18. Hirschmann JV, Pipavath SN, Godwin JD.
10. Katzenstein AL, Myers JL. Nonspecific interstitial Hypersensitivity pneumonitis: a historical, clinical,
pneumonia and the other idiopathic interstitial pneu- and radiologic review. Radiographics. 2009;29(7):
monias: classification and diagnostic criteria. Am J 1921–38.
Surg Pathol. 2000;24(1):1–3. 19. Akashi T, Takemura T, Ando N, Eishi Y, Kitagawa M,
11. Katzenstein AL, Fiorelli RF. Nonspecific interstitial Takizawa T, et al. Histopathologic analysis of sixteen
pneumonia/fibrosis. Histologic features and clinical autopsy cases of chronic hypersensitivity pneumonitis
significance. Am J Surg Pathol. 1994;18(2):136–47. and comparison with idiopathic pulmonary fibrosis/
12. Travis WD, Hunninghake G, King Jr TE, Lynch DA, usual interstitial pneumonia. Am J Clin Pathol.
Colby TV, Galvin JR, et al. Idiopathic nonspecific 2009;131(3):405–15.
interstitial pneumonia: report of an American 20. Trahan S, Hanak V, Ryu JH, Myers JL. Role of surgi-
Thoracic Society project. Am J Respir Crit Care Med. cal lung biopsy in separating chronic hypersensitivity
2008;177(12):1338–47. pneumonia from usual interstitial pneumonia/idio-
13. Katzenstein AL, Zisman DA, Litzky LA, Nguyen BT, pathic pulmonary fibrosis: analysis of 31 biopsies
Kotloff RM. Usual interstitial pneumonia: histologic from 15 patients. Chest. 2008;134(1):126–32.
study of biopsy and explant specimens. Am J Surg 21. Bolliger CT, Mathur PN, Beamis JF, Becker HD,
Pathol. 2002;26(12):1567–77. Cavaliere S, Colt H, et al. ERS/ATS statement on
14. Niewoehner DE, Kleinerman J, Rice DB. Pathologic interventional pulmonology. European Respiratory
changes in the peripheral airways of young cigarette Society/American Thoracic Society. Eur Respir J.
smokers. N Engl J Med. 1974;291(15):755–8. 2002;19(2):356–73.
Diagnosis of Granulomatous
Disease and Vasculitis in Small 6
Lung Biopsies

Sanjay Mukhopadhyay

Determine whether the granulomas are necrotizing


Introduction or non-necrotizing. Identification of necrosis is a
key step in narrowing the differential diagnosis
A granuloma is a compact (organized) aggregate since it influences the likelihood of finding an
of histiocytes [1]. Granulomas are a common organism. The majority of necrotizing granulomas
pathologic finding in transbronchial, endobron- of the lung are caused by mycobacterial or fungal
chial, and needle biopsies of the lung in routine infections, while sarcoidosis is the most common
practice [2]. Although sarcoidosis and infec- cause of non-necrotizing granulomas [1, 3]. Some
tions (mycobacterial or fungal) account for the entities, such as Cryptococcus infections and par-
majority of these lesions, several other entities ticulate matter aspiration, do not fit neatly within
enter the differential diagnosis [1, 3]. The aim of this paradigm, in that necrosis is present in some
this chapter is to discuss clinical, radiologic, cases but absent in others. These entities are dis-
and histologic findings that help to arrive at a cussed separately in “Causes of Granulomatous
specific diagnosis using small lung biopsies. Inflammation with Variable Necrosis” section.
A common problem faced by pathologists is that 1. Try to ascertain whether granulomas are
organisms cannot be demonstrated even on spe- located within the airspaces (alveolar spaces)
cial stains, and there are no pathognomonic fea- or interstitium (alveolar septa) and whether
tures of specific noninfectious entities. The they show a predilection for peribronchiolar
approach to such cases is discussed. Finally, or perivascular parenchyma. This further nar-
selected vasculitic diseases are reviewed since rows the differential diagnosis.
some of these overlap substantially with granu- 2. Look carefully for organisms on hematoxylin–
lomatous lung diseases. eosin (H&E)-stained slides before ordering
The diagnostic approach to granulomas in the special stains. Most organisms that cause
lung has been described in considerable detail, pulmonary granulomas are visible on H&E,
including an algorithmic approach [1, 4]. The key especially at high magnification. Notable
steps are as follows: exceptions are mycobacteria and Histoplasma
(in histoplasmomas) [5–7].
3. Obtain “special” stains for microorganisms. It is
S. Mukhopadhyay, M.D. (*) prudent to have a low threshold for ordering
Department of Anatomic Pathology, Cleveland stains, especially in the following situations:
Clinic, Robert J. Tomsich Pathology and Laboratory
• Necrotizing granulomas. Abundant necro-
Medicine Institute, 9500 Euclid Avenue/L25,
Cleveland, OH 44195, USA sis should prompt a rigorous search for
e-mail: mukhops@ccf.org organisms.

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 51


DOI 10.1007/978-1-4939-2575-9_6, © Springer Science+Business Media New York 2015
52 S. Mukhopadhyay

• Loose, vaguely granulomatous histiocytic fungal infections are a more common cause of
infiltrates. Such infiltrates may contain pulmonary granulomas, especially in endemic
numerous organisms. areas [3]. It is important for pathologists to
• The patient is immunocompromised. remember that tuberculosis is a clinical, radio-
• The clinician is particularly suspicious of logic, and/or microbiologic diagnosis, not a histo-
infection. logic one. The diagnosis may be confirmed by
Ziehl–Neelsen (commonly referred to as growth of M. tuberculosis in cultures of sputum,
AFB, which stands for acid-fast bacteria) and bronchial washings, or biopsied lung tissue. In the
Grocott methenamine silver (GMS) are the best appropriate clinical setting, a positive blood inter-
stains. Periodic acid–Schiff (PAS) is less sensi- feron-gamma release assay or a positive purified
tive than GMS, especially for Histoplasma, protein derivative (PPD) test may provide sup-
which is the only fungus that cannot be visual- portive evidence. In most cases, the role of the
ized without special stains. pathologist is to exclude alternative diagnoses,
4. Look for organisms only within the granulo- identify granulomatous inflammation, and detect
matous inflammatory infiltrate, using the cor- mycobacteria (AFB). Although differentiation of
responding H&E-stained slide for orientation. M. tuberculosis from non-tubercular mycobacte-
Organisms are usually found in the necrotic ria such as M. avium complex (MAC) is impor-
center of the granuloma, but may also be found tant for therapy, these organisms cannot be
in the granulomatous rim within histiocytes or distinguished histologically. Pathologists should
multinucleated giant cells [7–9]. Searching be prepared to explain to clinicians that mycobac-
randomly is unproductive. terial speciation is best performed by microbio-
5. Organisms may be visible at low magnifica- logic cultures. Cultures are often positive even
tion if they are large, darkly staining, or numer- when special stains on histologic specimens are
ous. However, mycobacteria and some fungi negative [3]. If, as is often the case, tissue was not
(e.g., Histoplasma) can be missed unless the submitted for cultures, or cultures are negative,
search includes painstaking examination at polymerase chain reaction (PCR) assays for M.
high magnification. A thorough search with a tuberculosis and non-tubercular mycobacteria can
40× objective is usually sufficient, but myco- be used on formalin-fixed paraffin-embedded tis-
bacteria can be so rare that a meticulous search sue. In practice, however, these assays are unavail-
in a large area of necrosis can take up to a half able for clinical use in most laboratories, and their
hour to find a single AFB. Constant adjustment yield is low in cases with small numbers of organ-
of fine focus is useful when hunting for AFB, isms. In a recent study, PCR for M. tuberculosis
and an oil immersion lens can also be helpful on formalin-fixed paraffin-embedded tissue did
in difficult cases. not detect even a single additional case of tuber-
6. Look for specific histologic features of nonin- culosis in pulmonary necrotizing granulomas in
fectious granulomatous diseases (described which histologic special stains and microbiologic
below) [1]. cultures were negative [10].

Histologic Findings in Small Biopsies


Causes of Necrotizing Necrotizing granulomatous inflammation is the
Granulomatous Inflammation hallmark of tuberculosis [11]. Although non-
necrotizing granulomas may occur alongside
Tuberculosis necrotizing granulomas, pure non-necrotizing
granulomatous inflammation is rare [1]. The
Clinical and Radiologic Findings granulomas of tuberculosis are often described
Worldwide, tuberculosis is the most common as “caseating,” an outdated term that refers to the
cause of necrotizing granulomas in the lung. This cheese-like macroscopic (gross) appearance of the
is not true in the United States, however, where necrosis. The necrosis of tuberculous granulomas
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 53

Fig. 6.1 Tuberculosis. Transbronchial lung biopsy from infiltrate is vaguely granulomatous. Compare with
an elderly individual with cough and weight loss. Cultures Fig. 6.2a. (b) Ziehl–Neelsen stain shows several acid-fast
grew M. tuberculosis. (a) This ill-defined histiocytic bacteria

is in fact quite variable histologically, ranging As stated above, M. tuberculosis and non-
from pink (eosinophilic, corresponding to the so- tubercular mycobacteria cannot be differentiated
called caseous necrosis) to “dirty” (basophilic histologically. Although some cases may show
due to necrotic nuclear debris) to frankly suppu- large numbers of organisms, AFB are often very
rative (neutrophil rich). Pink necrosis is the most few and difficult to find in immunocompetent
common, but this “caseation” is entirely nonspe- individuals. Screening with a 20× objective
cific since it can be seen in virtually any granulo- (or lower) may miss organisms if they are few in
matous mycobacterial or fungal infection [10]. number. Examination with a 40× or 100× (oil
Necrosis in tuberculosis is usually abundant. The immersion) objective lens for several minutes is
granulomatous infiltrate can be ill defined or required in most cases. Mycobacteria are most
vague, and necrosis may not be sampled, espe- commonly found within the necrotic center of
cially in small biopsies (Fig. 6.1a, b). There are necrotizing granulomas, although (unlike
no histologic features that differentiate tubercu- Histoplasma) they can also be found in the gran-
losis from other infectious granulomatous lung ulomatous rim. Each necrotic area in the slide
diseases. should be examined meticulously with constant
Detection of AFB is a key responsibility of the adjustment of fine focus so as not to miss organ-
pathologist. AFB are tiny rod-shaped structures isms that may be visible in only one plane. Areas
that stain red (in a blue background) on acid-fast other than the necrotic center and the granuloma-
stains such as Ziehl–Neelsen or Fite (Fig. 6.1a, b). tous rim virtually never contain organisms.
54 S. Mukhopadhyay

Non-tubercular Mycobacterial Histologic Findings in Small Biopsies


Infection The histologic findings of non-tubercular myco-
bacterial infections are highly variable. They
Clinical and Radiologic Findings include necrotizing granulomas identical to
Tuberculosis is such a well-recognized cause of tuberculosis, non-necrotizing granulomatous
pulmonary granulomas that few pathologists think inflammation associated with organizing pneu-
of non-tubercular mycobacteria (mycobacteria monia (“organizing granulomatous pneumonia”),
other than M. tuberculosis) as a cause of granulo- and other nonspecific findings such as bronchiec-
matous inflammation in the lungs. Clinicians and tasis and chronic bronchiolitis [10, 15, 16].
pathologists are most familiar with these organ- Sheets of foamy macrophages or ill-defined gran-
isms in the setting of acquired immunodeficiency ulomatous infiltrates—rather than well-formed
syndrome (AIDS), in which low CD4 counts pre- granulomas—are characteristic of infections in
dispose to uncontrolled proliferation of these immunocompromised hosts, most typically
organisms within macrophages. It is not widely patients with AIDS. The macrophages in such
recognized that infections with these organisms patients are usually packed with large numbers of
frequently involve immunocompetent patients AFB (Fig. 6.2a, b). The finding of granulomas
[12]. In fact, in the United States, non-tubercu- within airspaces (alveolar spaces and the lumens
lar mycobacteria are now more commonly iso- of small bronchioles) is a clue to non-tubercular
lated than M. tuberculosis, a trend that is mycobacterial infection (see “Hot Tub Lung”
reflected in lung specimens seen by pathologists section) [15]. AFB may be found in some cases
[3, 12]. For unclear reasons, there is a curious in immunocompetent individuals, but many are
predilection for the right middle lobe and lin- negative because of limited sampling or small
gula of middle-aged women (“middle lobe syn- numbers of organisms. In such cases, cultures
drome” or “Lady Windermere syndrome”) [13, may yield organisms even in the face of negative
14]. However, these infections can occur at any histologic special stains [3, 10]. Therefore, clini-
age and involve both genders. The most typical cians and pathologists must ensure that results of
radiographic picture is multifocal bronchiecta- cultures are negative before an active mycobacte-
sis with multiple small nodules, but cavities and rial infection is excluded—this can take up to
infiltrates are also common. The diagnosis is several weeks. Cultures are the only definitive
most often made by identifying non-tubercular means for differentiating tuberculosis from non-
mycobacteria in microbiologic cultures of respi- tubercular mycobacterial infection. For a discus-
ratory tract specimens. The most common organ- sion on examination of acid-fast stains and
ism is Mycobacterium avium–intracellulare mycobacterial speciation, see the preceding sec-
(MAI), currently known as MAC. Other species tion on tuberculosis.
such as M. kansasii, M. abscessus, M. fortuitum,
and M. chelonae are occasionally isolated. Since
non-tubercular mycobacteria are common in the Histoplasmosis
environment (especially in water), a common
clinical dilemma is whether the organisms are Clinical and Radiologic Findings
truly responsible for the patient’s symptoms or Histoplasmosis is the most common endemic fun-
are merely contaminants. There is no easy answer gal infection in the United States. It occurs in sev-
to this question, but clinical criteria to differenti- eral central, midwestern, and southern states along
ate true infections from saprophytic growth have the Ohio and Mississippi river valleys, and also in
been proposed [12]. As with tuberculosis, the role regions on both sides of the St. Lawrence Seaway,
of surgical pathologists is to exclude malignancy, which runs between Canada and the northeastern
identify granulomas, and find mycobacteria on United States. The infection is also endemic in
acid-fast stains. several countries in Central and South America.
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 55

Fig. 6.2 Non-tubercular mycobacterial infection in an vaguely granulomatous. Such lesions often contain greater
immunocompromised patient. Transbronchial lung numbers of organisms than well-formed granulomas. (b)
biopsy. Cultures grew M. avium. (a) Histiocytic infiltrate, Innumerable acid-fast bacteria on a Ziehl–Neelsen stain

The most common circumstance in which that stray from this norm may be considered
pathologists encounter Histoplasma is a biopsy of suspicious for malignancy. Thus, a solitary lung
a lung nodule to rule out malignancy (Fig. 6.3a, b). nodule in a smoker or a nodule that is spiculated,
The majority of such nodules are incidentally noncalcified, enlarging in size, or positron emis-
detected on radiographs performed for unrelated sion tomography (PET) positive is liable to be
reasons. The term “histoplasmoma” has been biopsied. Since each of these features has been
applied to such nodules when they are shown his- documented in pulmonary histoplasmomas, his-
tologically to be necrotizing granulomas contain- tology is the only modality that can definitively
ing Histoplasma organisms [6, 9, 17, 18]. Several determine the nature of such nodules. This type
studies have demonstrated that the organisms in of nodule is usually peripheral and is best sam-
the majority of these lesions are nonviable and pled with a needle biopsy [2, 18, 21]. Surgical
culture negative [3, 7, 10, 19]. The suffix “-oma” pathologists see histoplasmomas in institutions
is applied because these lesions can mimic many where core needle biopsies of lung nodules are
features of malignant lung nodules. Clinicians performed, whereas in centers where fine needle
and radiologists are trained to recognize small, aspiration is the preferred approach to peripheral
stable (nongrowing), calcified, non-spiculated lung nodules, this lesion falls into the domain of
nodules in nonsmokers as benign [20], but lesions the cytopathologist.
56 S. Mukhopadhyay

Fig. 6.3 Histoplasmoma. (a) Chest CT showing solitary (arrow) and granulomatous inflammation is at the bottom
lung nodule (arrow). (b) Core needle biopsy of the nodule left (arrow). No organisms are visible on H&E
shows a necrotizing granuloma. Necrosis is at the top

Rarely, transbronchial or needle biopsy of the Histologic Findings in Small Biopsies


lung may be performed in patients with a poten- Histologically, a histoplasmoma is a necrotizing
tially fatal, progressive form of histoplasmosis granuloma, the tissue reaction being indistin-
known as disseminated histoplasmosis [5]. Such guishable from the so-called caseating granu-
patients are usually immunocompromised and loma of tuberculosis (Fig. 6.3a, b) [6, 9]. The
develop disseminated disease that may second- lesion is characterized by a large necrotic center
arily involve the lungs, analogous to miliary surrounded by a relatively thin granulomatous
tuberculosis. In a few cases, clinical and radio- rim composed of a few layers of epithelioid his-
logic evidence of extrapulmonary involvement is tiocytes, occasionally accompanied by a few
absent at presentation, making the diagnosis of multinucleated giant cells. Since the majority of
disseminated disease challenging. Clinical clues the lesion consists of necrosis, the latter is often
suggestive of disseminated disease include the the most prominent finding in needle biopsies
immunosuppressed state of the patient com- [2, 18, 21]. The necrosis may be pink (“caseous”)
bined with the presence of fever, thrombocyto- or may contain nuclear debris (“dirty”) or ghosts
penia, elevated transaminases, multiple lung of necrotic alveoli (“infarct like”). Histoplasma
nodules, and/or bilateral reticulonodular lung yeasts are present within the necrosis, but are
infiltrates [5]. almost never visible on H&E, a diagnostically
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 57

Fig. 6.4 Histoplasma yeasts on GMS. (a) When organisms are numerous, narrow-based budding is likely to be present
(arrow). (b) Scarce organisms (arrows) may be missed, especially at low magnification

valuable feature that differentiates Histoplasma include the relatively few organisms, necrotic
from other fungal yeasts that cause pulmonary background, extracellular location, and non-
granulomas, all of which are visible on H&E viability of the organisms [5].
[1, 6, 7, 9]. GMS staining is required to demon- Histoplasma must also be differentiated from
strate Histoplasma organisms within the necro- tiny, lightly basophilic microcalcifications, which
sis, where they appear as small (3–5 μm), are often visible within necrosis on H&E, but are
uniform, oval, round, or tapered yeasts (Fig. 6.4a, b) nonuniform in shape and GMS negative. Dust
[1, 18]. Organisms can vary from numerous to particles, which are GMS positive, can mimic
rare [5]. The size, shape, and uniformity of the Histoplasma, but they are smaller and less uni-
yeasts, as well as their non-visibility within form and lack the typical oval or tapered shapes
necrosis on H&E, are the key diagnostic fea- of this organism. Conversely, if GMS-stained
tures, aiding to distinguish them from slides are examined only at low magnification,
Cryptococcus and Pneumocystis, which are the Histoplasma yeasts can be misinterpreted as dust
fungi most likely to cause diagnostic difficulties particles.
[1, 22]. Narrow-based budding is a helpful fea- The histologic features of the disseminated
ture, but is also seen in Cryptococcus, and may form of histoplasmosis, which can involve the lung,
not be present when organisms are few in num- are highly suggestive of aggressive, widespread
ber. Potential explanations for the non-visibility disease and serve as a histologic surrogate marker
of Histoplasma in histoplasmomas on H&E of immunosuppression. They include filling of
58 S. Mukhopadhyay

Fig. 6.5 Disseminated histoplasmosis in a transbronchial granulomas. (b) Morphologic details are best appreciated
lung biopsy. The patient was an immunocompromised under oil immersion. Histoplasma yeasts are round to oval
renal transplant recipient. (a) Numerous histiocytes are and ring like, with an eccentric purple dot or crescent.
present within the airspaces (short arrow) and alveolar This is the form of histoplasmosis that was described by
septa (long arrow). Organisms are readily visible within Samuel Darling in 1906
the cytoplasm of the histiocytes on H&E. There are no

airspaces and alveolar septa by numerous histio- New Mexico, Nevada, Utah, and western Texas,
cytes engorged by large numbers of yeasts that as well as in northern Mexico and other areas in
are easily visible on H&E (Fig. 6.5a, b) [5, 23]. Central and South America. Cases are occasion-
Granulomas are usually absent, although necro- ally encountered outside these areas in individu-
sis may be present. The visibility of the organ- als who visit or move to other states after being
isms within viable macrophages on H&E is in infected in an endemic area. In endemic areas,
sharp contrast to the morphology of the far more acute exposure causes a flu-like illness known as
common histoplasmomas described in the pre- acute coccidioidomycosis or valley fever (a dis-
ceding paragraphs. ease named for the San Joaquin Valley in
California). Healed and/or contained infections
can cause lung nodules or cavities that mimic
Coccidioidomycosis malignancy, analogous to histoplasmomas (see
“Clinical and Radiologic Findings” section of
Clinical and Radiologic Findings histoplasmosis). In some series of solitary pul-
Coccidioidomycosis is a fungal infection monary nodules in endemic areas, coccidioidal
endemic in the southwestern United States granulomas are more common than malignan-
including southern Arizona, southern California, cies. Only a minority of patients with nodular/
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 59

Fig. 6.6 Coccidioides in a necrotizing granuloma. Core organisms are visible on H&E, even at low magnification.
needle biopsy of a lung nodule. (a) The main finding in (b) Thick-walled Coccidioides spherules, filled with
this biopsy is necrosis. Granulomatous inflammation was endospores. The presence of these structures is
present elsewhere in the biopsy. Large round fungal diagnostic

cavitary disease are immunocompromised. The eosinophils are absent in some cases. Definitive
majority have negative results by serology. Thus, diagnosis rests on the identification of “spher-
a small lung biopsy can be a valuable nonopera- ules,” which are large (20–100 μm) round struc-
tive means of establishing a definitive diagnosis. tures with a thick cell wall (Fig. 6.6a, b) [18, 24].
Spherules may be filled with several small yeast-
Histologic Findings in Small Biopsies like “endospores” or may be ruptured, frag-
Coccidioidomycosis can be diagnosed on needle mented, collapsed, or empty [8]. It has been
biopsies [18], although the yield is considered to proposed that the morphologic diagnosis of coc-
be poor. Histology is often diagnostic despite cidioidomycosis should require at least one
negative cultures [3, 24]. In immunocompetent endospore-filled spherule [26]. Both spherules
individuals, coccidioidomycosis manifests as and endospores are visible on H&E, but spher-
necrotizing granulomas with pink (“caseating”) ules are easier to recognize because of their large
necrosis identical to tuberculomas or histoplas- size [9]. They may be found either within the
momas [9] or suppurative necrosis similar to necrotic center of the granuloma or within multi-
blastomycosis. Eosinophils can be numerous nucleated giant cells, even in small biopsies.
within the necrotic centers of the granulomas as Budding is absent. Hyphae may occasionally be
well as in the surrounding lung, and this can be a found. The main organism in the differential
valuable clue to the diagnosis [25]. However, diagnosis is Blastomyces, which can usually be
60 S. Mukhopadhyay

distinguished by the presence of broad-based broad-based bud, a key feature in the differential
budding and nuclear material. However, in some diagnosis with other morphologically similar
cases, Coccidioides and Blastomyces cannot be fungal yeasts, including Cryptococcus and
reliably differentiated on histologic grounds. Coccidioides [1].

Blastomycosis Aspergillosis

Clinical and Radiologic Findings Clinical and Radiologic Findings


Blastomycosis is an uncommon fungal infection Fungal hyphae such as Aspergillus usually do not
that usually involves immunocompetent individu- cause granulomatous inflammation in the lung.
als. In North America, most reported cases have However, a minority of pulmonary infections with
been from states bordering the Great Lakes, the these fungi are associated with a granulomatous
St. Lawrence Seaway, the Lower Mississippi response, which appears to be a marker of limited
valley, and the southeast United States, a geo- tissue invasion. The best-known clinical entity in
graphic distribution similar to histoplasmosis [27]. which granulomatous inflammation occurs is
Cough and fever are common symptoms. Patients chronic necrotizing pulmonary aspergillosis, also
may present with a flu-like illness, a bacterial known as semi-invasive aspergillosis [30, 31].
pneumonia-like picture, or acute respiratory dis- The condition was described as a chronic progres-
tress syndrome (ARDS). Chest imaging can sive infection characterized by persistent or enlarg-
show infiltrates, pneumonia-like consolidation, ing lung cavities in individuals without preexisting
nodules, or masses with or without cavitation. cavitary lung disease. Subsequent studies have
Extrapulmonary disease can occur, the major sites shown a predilection for men and individuals with
being the skin, genitourinary tract, and bone [27, 28]. emphysema or compromised immunity.
The sensitivity of serology is low. Cultures may
take 3–4 weeks to become positive. Histologic Findings in Small Biopsies
Most instances of this entity are characterized by
Histologic Findings in Small Biopsies necrotizing granulomatous inflammation similar
Blastomycosis can be diagnosed in small lung to tuberculosis or histoplasmosis [32]. The diag-
biopsies [21, 28]. Histology can be positive in nosis rests on demonstration of fungal hyphae
culture-negative cases [29]. Of the major fungal within the granulomas, usually in the cavitary
yeasts that infect the lung, Blastomyces forms the center. Aspergillus cannot be reliably differenti-
most distinctive granulomas. The granulomas are ated from other morphologically similar fungal
characteristically “suppurative,” i.e., they have hyphae without microbiologic confirmation.
neutrophil-rich centers resembling abscesses.
They are frequently mistaken for abscesses and
are referred to in the older literature as “pyogranu- Wegener’s Granulomatosis
lomatous.” The periphery of the granulomas is (Granulomatosis
composed of epithelioid histiocytes and variable with Polyangiitis/ GPA)
numbers of multinucleated giant cells. Organisms
can be found both in the suppurative center and Clinical and Radiologic Findings
the granulomatous rim [2, 21]. Blastomyces yeasts Wegener’s granulomatosis is a form of vasculitis
are large (8–15 μm) with a thick double-contoured that classically affects the lungs, kidneys, and
cell wall. A characteristic feature is the presence upper respiratory tract. It is the most common
of basophilic nuclear material. The organisms are type of vasculitis seen in lung biopsies. Patients
visible on H&E as well as GMS. Although GMS usually present with cough, hemoptysis, dyspnea,
highlights far more organisms than are apparent renal failure, sinusitis, and/or epistaxis.
on H&E, it also obscures the nuclear material. Radiographic findings include bilateral cavitary
Blastomyces characteristically forms a single nodules or infiltrates. Some patients present with
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 61

Fig. 6.7 Granulomatosis with polyangiitis (Wegener’s inflammatory infiltrate consisting of neutrophils and his-
granulomatosis). Core needle biopsy. (a) “Dirty” necrosis tiocytes (long arrow). Note that the left side of the vessel
is basophilic due to abundant nuclear debris. (b) is relatively spared (short arrow is in lumen). This type of
Necrotizing vasculitis. The wall of this medium-sized “eccentric vasculitis” is typical of Wegener’s
blood vessel is focally infiltrated and destroyed by an granulomatosis

clinical and radiologic features of diffuse alveolar granulomatosis in atypical settings and should not
hemorrhage. Most cases are positive for anti- be deterred by the absence of multisystem involve-
neutrophil cytoplasmic antibodies (ANCA). ment or a negative ANCA. Patients with localized
Positive immunofluorescence for cytoplasmic pulmonary necrotizing granulomas can subse-
ANCA (c-ANCA) with antibodies against pro- quently develop extrapulmonary vasculitis and
teinase 3 has high specificity for Wegener’s gran- become ANCA positive on follow-up [10].
ulomatosis. However, it is well documented that
Wegener’s granulomatosis can present with local- Histologic Findings in Small Biopsies
ized pulmonary involvement and that such cases The classic histologic findings of Wegener’s
are significantly more likely to be ANCA negative granulomatosis are necrotizing granulomatous
than those with multisystem disease. It is such inflammation and necrotizing vasculitis. This
cases that cause the greatest diagnostic difficul- combination is required for a definitive pathologic
ties. Radiologically, these cases may present with diagnosis of Wegener’s granulomatosis. Unfortu-
bilateral lung nodules. However, Wegener’s gran- nately, it is often difficult or impossible to demon-
ulomatosis presenting as a solitary lung nodule strate since the affected vessels may be completely
has also been described [33]. Biopsies are most destroyed. The necrosis of Wegener’s granuloma-
often performed when the clinical setting is tosis is often described as “dirty” due to its baso-
unusual. Pathologists must therefore be prepared philic hue, which is caused by nuclear debris
to recognize the histologic features of Wegener’s derived from necrotic neutrophils (Fig. 6.7a, b).
62 S. Mukhopadhyay

Suppurative (neutrophil rich) necrosis may also Churg–Strauss Syndrome


be present. “Geographic” necrosis, characterized (Eosinophilic Granulomatosis
by necrotic areas with irregular contours, cannot with Polyangiitis/EGPA)
be appreciated in a small biopsy. Tiny microab-
scess-like granulomas with suppurative centers Clinical and Radiologic Findings
may be a clue to the diagnosis. They consist of Churg–Strauss syndrome is a rare disease of
aggregates of a few multinucleated giant cells asthmatic individuals characterized by tissue and
surrounding a minute cluster of neutrophils [34]. blood eosinophilia, granulomatous inflammation,
Multinucleated giant cells with prominent hyper- and vasculitis. Patients typically present with
chromatic nuclei may accompany the necrotizing fever, weight loss, striking peripheral (blood)
granulomas. When demonstrable, necrotizing eosinophilia, and transient or migratory pulmo-
vasculitis manifests as a mixed neutrophilic–his- nary infiltrates. Imaging features include
tiocytic infiltrate within vessel walls, often ground-glass opacities, centrilobular nodules, and
accompanied by small, subtle foci of fibrinoid airspace consolidation [39]. Many patients have
necrosis. The infiltrate frequently involves only extrapulmonary manifestations such as cutaneous
one side of the affected vessel (“eccentric vascu- lesions or neuropathy. Perinuclear ANCA
litis”) (Fig. 6.7a, b). Karyorrhectic nuclear debris (p-ANCA) is positive in approximately two-thirds.
is often present. Findings that should not be mis- Many cases are diagnosed without biopsy or with
interpreted as necrotizing vasculitis include vas- a biopsy of a site other than the lung.
cular inflammation comprised entirely of
lymphocytes and plasma cells, granulomas in the Histologic Findings in Small Biopsies
vessel wall, and necrotic vessels within areas of The most consistent histologic feature of Churg–
parenchymal necrosis. In virtually all cases, there Strauss syndrome is the presence of increased
is a background of severe nonspecific acute and numbers of eosinophils within the affected tis-
chronic inflammation containing neutrophils, sues. The full spectrum of diagnostic findings,
lymphocytes, and plasma cells. Organizing pneu- which consists of a combination of necrotizing
monia (plugs of fibroblasts within airspaces) may vasculitis, granulomatous inflammation with
be present. The presence of hemosiderin-laden eosinophils, and eosinophilic pneumonia [4], is
macrophages in and around the inflammatory rarely seen, especially in transbronchial biopsies
process is another clue to the correct diagnosis. [40]. However, pathologists should recognize
Wegener’s granulomatosis occasionally pres- that any of these features—especially in combi-
ents with diffuse pulmonary hemorrhage, the nation with numerous eosinophils—raises the
histologic correlate of which is “capillaritis,” a possibility of Churg–Strauss syndrome in the
destructive vasculitis affecting alveolar septal cap- appropriate clinical context.
illaries [35–37]. It is characterized by a mixed neu-
trophilic–histiocytic inflammatory infiltrate that
destroys alveolar septa and is accompanied by Rheumatoid Nodule
intra-alveolar hemorrhage consisting of variable
combinations of fresh blood (red blood cells), Clinical and Radiologic Findings
fibrin, and hemosiderin-laden macrophages. The Rheumatoid nodules typically present as multiple
neutrophils invariably spill into the adjacent air- pulmonary nodules in patients with known
spaces, causing acute bronchopneumonia to enter rheumatoid arthritis [10]. Most patients are sero-
the differential diagnosis. positive at the time of diagnosis, and some have
Transbronchial lung biopsies may occasionally concomitant subcutaneous rheumatoid nodules.
show suggestive but nonspecific findings such as
acute inflammation mixed with granulomatous Histologic Findings in Small Biopsies
inflammation or granulomas with dirty necrosis It is impossible to make this diagnosis with cer-
[38]. In such cases, a definitive diagnosis cannot tainty on a small biopsy since it is partially a
be made by histology alone and requires correla- diagnosis of exclusion that rests on the finding of
tion with ANCA serologies. necrotizing granulomatous inflammation without
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 63

evidence of organisms or necrotizing vasculitis in most important one is to carefully reexamine the
patients with known rheumatoid arthritis. The histologic findings, especially the GMS-stained
histologic findings are not distinctive. The role of slides. Waiting for results of cultures may be pro-
pathology is to exclude alternative possibilities, ductive since some organisms, especially myco-
the most important being infection and Wegener’s bacteria, grow in cultures even though special
granulomatosis. stains may be negative. Finally, clinical informa-
tion, specifically radiologic findings, results of
ANCA tests, and a history of rheumatoid arthritis,
Rare Infectious Causes of Pulmonary can help in making a specific diagnosis in some
Granulomas cases. The best way to communicate these issues
to the clinician is in a comment, instead of
A handful of other organisms cause granuloma- encumbering the diagnostic line with a wordy
tous inflammation in the lungs, but are very description. The diagnosis can then simply be
unlikely to be seen in routine practice, especially “necrotizing granulomatous inflammation (see
in small lung biopsies. These include fungi such comment).”
as Sporothrix, which typically causes suppurative
granulomas in the skin but can also involve the
lungs [41], and Pneumocystis, which usually Causes of Non-necrotizing
causes a frothy intra-alveolar exudate but can Granulomatous Inflammation
rarely be associated with granulomatous inflam-
mation [22], and parasites such as Dirofilaria, Sarcoidosis
which causes granulomas with infarct-like necro-
sis due to embolization from the right side of the Clinical and Radiologic Findings
heart into pulmonary arteries [42]. The diagnosis of sarcoidosis depends on a com-
bination of clinical context and pathologic find-
ings [44]. Yet, details of clinical and radiologic
Necrotizing Granulomas of Unknown findings are rarely, if ever, provided to patholo-
Etiology gists at the time of the initial diagnosis. However,
pathologists with access to electronic medical
It is common for necrotizing granulomas to be records should be familiar with key findings that
found in lung specimens with no histologic evi- indicate whether the clinical context is appropri-
dence of a definite etiology, infectious or other- ate for sarcoidosis. Typical patients with sarcoid-
wise. This is especially true in small biopsies, osis are young individuals of either gender, with
which do not sample the entirety of the lesion. a peak in the 30s and 40s [45]. Cases have been
Diagnostic findings such as organisms or vasculi- well documented in the elderly but are infrequent
tis are therefore easily missed. The problem is [46]. The most common radiologic finding is
greater than mere sampling error. Studies have bilateral hilar or mediastinal adenopathy.
demonstrated that even when entire necrotizing Bilateral hilar adenopathy in an asymptomatic
granulomas are resected surgically, an etiology young person is considered highly suspicious for
cannot be found in a significant percentage of sarcoidosis [44]. In the lungs, radiologic findings
cases [7, 10]. There is substantial evidence to may be absent or may consist of bilateral infil-
support the notion that such lesions are “burnt- trates or nodules. Patients may be asymptomatic,
out” mycobacterial or fungal infections in which the radiologic findings having been discovered
organisms have been destroyed or removed by incidentally, or they may present with cough and
the granulomatous inflammatory response. dyspnea. Sarcoidosis is a multisystem disease
What can pathologists do if the etiology of a and may involve virtually any other organ.
granulomatous process is not apparent and spe- Patients can present with a wide variety of symp-
cial stains are negative? [4, 43] Of the several toms referable to involvement of these sites, such
steps that can reveal an etiology in such cases, the as erythema nodosum (skin) and uveitis (eye).
64 S. Mukhopadhyay

Fig. 6.8 Non-necrotizing granulomas consistent with normal lung away from the granulomas (arrowhead, bot-
sarcoidosis in a transbronchial lung biopsy. (a) Multiple tom right). (b) Same biopsy, high magnification. The
non-necrotizing granulomas are present within the inter- granulomas are well formed and surrounded by hyalinized
stitium (arrows). An important finding is the presence of fibrosis

Clinical findings that argue against sarcoidosis hilar lymph nodes [48]. The histologic hallmark
include unilateral infiltrates, a unilateral mass, of sarcoidosis is the non-necrotizing (“noncaseat-
bronchiectasis, predominant airspace opacities, ing”) granuloma [1, 20]. However, non-
lack of response to steroids, and absence of hilar necrotizing granulomas are also found in other
or mediastinal adenopathy. Needless to say, posi- lung diseases [47], making it important for
tive cultures for mycobacteria or fungi exclude pathologists to recognize histologic features that
the diagnosis. Unfortunately, culture results are are consistent with sarcoidosis and those that
seldom available at the time of biopsy [47]. exclude or argue against the diagnosis [1].
Therefore, the pathologic diagnosis, and often Pathologists are well served to remember that
the clinical diagnosis, is usually made before an one of their key responsibilities is the exclusion
infection can be definitively excluded. of alternative diagnoses, including infection,
malignancy, and other causes of granulomatous
Histologic Findings in Small Biopsies lung disease.
In western countries, sarcoidosis is by far the In transbronchial biopsies, the granulomas of
most common cause of granulomas in transbron- sarcoidosis are found in the bronchial mucosa or
chial biopsies [3]. Endobronchial ultrasound in the alveolar septa (interstitium) (Figs. 6.8a, b
(EBUS) with transbronchial needle aspiration is and 6.9a, b) [4, 47]. In classic cases, they are
increasingly used to demonstrate granulomas in plump, well formed, and surrounded by layers of
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 65

Fig. 6.9 Non-necrotizing granulomas consistent with granuloma contains an inclusion (arrow). There is no hya-
sarcoidosis. Transbronchial lung biopsy. (a) Non- linized fibrosis. Lung parenchyma away from the granu-
necrotizing granuloma within bronchiolar wall. An inclu- loma is normal. Although the findings are consistent with
sion is present within a giant cell (arrow). (b) sarcoidosis, they are less typical than the case illustrated
Non-necrotizing granuloma within the interstitium. The in Fig. 6.8. Also compare with hypersensitivity pneumo-
granuloma is small and ill defined. A giant cell in the nitis (Figs. 6.10, 6.11, and 6.12)

concentric hyalinized fibrosis (Fig. 6.8a, b). able diagnostic feature in surgical lung biopsies
Multiple small granulomas may fuse into larger but for obvious reasons cannot be appreciated in
nodules. Necrosis can occur but in most cases is transbronchial biopsies. Special stains for
minimal, tends to have a degenerated or fibrinoid microorganisms should be performed in all cases.
quality, and usually involves only a few granulo- Histologic features that argue against sarcoid-
mas [49]. Nonspecific inclusions of various osis—and suggest an alternative diagnosis—
kinds, including pink spider-like structures include extensive necrosis, suppurative or “dirty
(asteroid bodies), basophilic concentric calcifica- necrosis,” organizing pneumonia (fibroblast
tions (Schaumann bodies), and calcium oxalate plugs within the airspaces), and foreign particles
crystals, may be found within the granulomas (vegetable particles, talc, microcrystalline cellu-
(Fig. 6.9a, b) [1, 47, 49]. These are thought to be lose, or crospovidone) [4]. An important histo-
endogenous products of macrophage metabolism logic feature of sarcoidosis is that chronic
and are not specific for sarcoidosis. The classic inflammation, when present, is localized to the
lymphangitic (lymphatic) distribution of sarcoid area surrounding the granulomas (Fig. 6.9a, b).
granulomas—along the pleura, interlobular The lung away from the granulomas is normal.
septa, and bronchovascular bundles—is a valu- This feature helps to differentiate sarcoidosis
66 S. Mukhopadhyay

from hypersensitivity pneumonitis, in which offending antigen is a classic feature, but cases
interstitial chronic inflammation is found even in that come to biopsy usually do lack an obvious
lung parenchyma away from granulomas [1, 50, 51]. exposure. Perhaps the best-known form of hyper-
Needless to say, identification of mycobacterial sensitivity pneumonitis is “farmer’s lung,” a con-
or fungal organisms on H&E or special stains dition resulting from exposure of sensitized
excludes sarcoidosis. On the other hand, negative individuals to thermophilic actinomycetes such
special stains do not “confirm” a diagnosis of as Saccharopolyspora rectivirgula, a bacterium
sarcoidosis, since these can be negative in infec- found in moldy hay [51, 54, 55]. The most common
tions [47]. source of exposure identified in biopsy-proven
For practical purposes, the information in the cases is avian antigens derived from pet birds
preceding paragraphs must be distilled into a diag- such as parakeets (including budgerigars), cocka-
nosis on a pathology report. The most conservative tiels, finches, doves, canaries, macaws, pigeons,
approach on transbronchial biopsies is to simply and parrots (“bird fancier’s lung”) [50, 56–58].
state “non-necrotizing granulomas” in the diagnosis Other relatively common sources of exposure are
and put the “soft findings” in the comment, which household mold, hot tubs, humidifiers, and
can include a description of the granulomas, the feather duvets. The proportion of cases of well-
presence of typical or atypical findings, and the documented hypersensitivity pneumonitis in
extent to which the histologic findings are consis- which an inciting antigen cannot be found is
tent with sarcoidosis. When findings are minimal quite high, varying from 25 to 63 % [53, 57, 58].
and equivocal, it is appropriate to state this in the Radiologic findings are not specific, although
comment. On the other hand, histologically classic they may be suggestive [55]. The most common
granulomas are occasionally encountered in the findings are bilateral ground-glass infiltrates, tiny
appropriate clinical context. In such cases, the centrilobular nodules/opacities, and mosaic atten-
diagnosis could state “non-necrotizing granulo- uation, the last being a consequence of air trapping
mas consistent with sarcoidosis.” The final diagno- due to small airway (bronchiolar) involvement.
sis of sarcoidosis is a clinical one and ideally takes The findings are usually more prominent in the
into consideration clinical, radiologic, pathologic, upper lobes. However, one or more of these find-
and microbiologic findings. The difficulties of ings may be absent, and any lobe can be involved.
making a clinical diagnosis of an entity that lacks Long-standing cases can progress to fibrosis and
a gold standard and is largely a diagnosis of exclu- honeycomb change.
sion have been well described [44].
Histologic Findings in Small Biopsies
Hypersensitivity pneumonitis is occasionally
Hypersensitivity Pneumonitis encountered in transbronchial lung biopsies,
although less often than sarcoidosis. Although it
Clinical and Radiologic Findings is included in this chapter on granulomas, it is
Hypersensitivity pneumonitis (also known as important to remember that the hallmark of this
hypersensitivity pneumonia and referred to in the disease is diffuse interstitial chronic inflamma-
past as extrinsic allergic alveolitis) is a form of tion and that the predominant cell in the inflam-
interstitial lung disease caused by inhalational matory infiltrate is the lymphocyte [50, 51, 53,
exposure to organic antigens in susceptible and 54, 59]. Granulomatous inflammation is usually
sensitized individuals [52, 53]. The disease is subtle and is absent in a significant proportion of
often difficult to diagnose because there is no cases (30 % in most series) [51, 52]. Contrary to
universally accepted gold standard. The symp- what one might expect from the name of the con-
toms are similar to other interstitial lung diseases, dition, eosinophils are absent or at best occa-
the most common being cough with or without sional [51–54].
dyspnea [52, 54]. Most patients are nonsmokers. The main finding in small biopsies is a diffuse
Worsening of symptoms upon exposure to the chronic inflammatory infiltrate that expands the
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 67

Fig. 6.10 Hypersensitivity pneumonitis in a woman with half of the picture. The location of the giant cell adjacent
a pet bird (macaw). Transbronchial lung biopsy. (a) to a small artery suggests a peribronchiolar location. (b)
Diffuse interstitial chronic inflammation with slight Multinucleated giant cell at high magnification. Note the
accentuation around a respiratory bronchiole (arrow). A cholesterol crystal/cleft, a common inclusion in this
giant cell is present within the interstitium in the lower condition

alveolar septa (interstitium) (Figs. 6.10a, b and illustrated elsewhere [1]. Foamy macrophages
6.11a, b) [52, 59]. Granulomas, when present, are often present within the airspaces in hyper-
are interstitial, tiny, and poorly formed, often sensitivity pneumonitis. They are a nonspecific
consisting of no more than a few loosely aggre- manifestation of small airway involvement/
gated histiocytes or isolated multinucleated giant obstruction, but their presence can be a tip-off to
cells (Figs. 6.10a, b; 6.11a, b; and 6.12a, b) [60]. the diagnosis.
In many cases, the “granulomas” are actually Classic cases of chronic hypersensitivity
single multinucleated giant cells scattered within pneumonitis show a characteristic “histologic
the interstitial inflammatory infiltrate, containing triad,” which is important to recognize because it
a variety of nonspecific endogenous inclusions allows pathologists to suggest the diagnosis even
such as cholesterol crystals/clefts, Schaumann in the absence of clinical or radiologic informa-
bodies, or calcium oxalate crystals (Figs. 6.11a, b tion. Interestingly, the triad is defined differently
and 6.12a, b). Some of these inclusions are in different publications. Most authors define it
birefringent and may be misinterpreted as “for- as a combination of cellular bronchiolitis, inter-
eign material.” The differences between these stitial chronic inflammation, and scattered, small
structures and true foreign particles have been interstitial non-necrotizing granulomas [52, 53].
68 S. Mukhopadhyay

Fig. 6.11 Hypersensitivity pneumonitis in a transbron- long arrows). Note the tiny, poorly formed granuloma
chial lung biopsy. (a) Cellular chronic interstitial pneumo- (short arrow), shown at high magnification in (b). (b)
nia characterized by alveolar septal chronic inflammatory Poorly formed granuloma typical of hypersensitivity
infiltrate (arrows). The area at the top right is in the vicin- pneumonitis. Note the inclusion (Schaumann body)
ity of an alveolar duct (see scattered smooth muscle fibers, within a multinucleated giant cell

However, a slightly different triad consisting of pools [59, 61–63]. The clinical presentation is
interstitial chronic inflammation, granulomas, and similar to that of hypersensitivity pneumonitis.
organizing pneumonia has also been described. Women are more commonly affected than men.
This latter triad is present in 42 % of surgical The most common symptoms are dyspnea,
biopsies but only 9 % of transbronchial biopsies, cough, and fever. Imaging shows bilateral inter-
highlighting the difficulties in diagnosis intro- stitial infiltrates often described as nodular, retic-
duced by sampling error in small biopsies [56]. ulonodular, or ground glass, with centrilobular
nodules and air trapping [64, 65]. A history of hot
tub use is essential for the diagnosis.
Hot Tub Lung
Histologic Findings in Small Biopsies
Clinical and Radiologic Findings The most characteristic histologic finding is the
Hot tub lung is a hypersensitivity-like granulo- presence of well-formed granulomas within the
matous reaction to non-tubercular mycobacteria in lumens of alveolar ducts and respiratory bron-
aerosolized water derived from hot tubs and other chioles [1, 4, 66]. The granulomas are typically
sources such as showerheads and swimming non-necrotizing, but necrotizing granulomas
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 69

Fig. 6.12 (a, b) Inclusions in hypersensitivity pneumoni- are thought to be endogenous products of macrophage
tis. These concentric calcifications (Schaumann bodies) metabolism. They should not be mistaken for exogenous
are typically found within multinucleated giant cells and (foreign) particles

may also be seen. Interstitial granulomas and Talc Granulomatosis


interstitial chronic inflammation are often present.
The histologic features overlap considerably Clinical and Radiologic Findings
with hypersensitivity pneumonitis, but the granu- “Talc granulomatosis” is one of many terms used
lomas of hot tub lung are usually larger, better to describe a distinctive pulmonary perivascular
formed, and occur within the airspaces [1]. foreign-body granulomatous reaction seen in
Additionally, organizing pneumonia is more individuals who inject pills intended for oral con-
prominent in hot tub lung than in hypersensitivity sumption [67]. The practice differs from usual
pneumonitis. The presence of organizing pneu- forms of intravenous drug abuse in that it involves
monia and airspace granulomas helps to distin- injection of pills or tablets rather than better-
guish hot tub lung from sarcoidosis. The known drugs of abuse such as heroin or cocaine.
possibility of hot tub lung can be raised on the The pills are typically crushed, dissolved in an
basis of histologic findings, but definitive diagno- aqueous solution, and injected intravenously,
sis requires a history of hot tub use. AFB are hence the term “mainline granulomatosis” [68].
demonstrable in lung biopsies in some cases. The pills contain psychoactive substances such as
Cultures of respiratory specimens and/or the con- methadone, pentazocine, methylphenidate, and
taminated hot tub water may yield mycobacteria, tripelennamine, oral medications that produce a
mainly MAC [63–65]. greater and more immediate stimulant effect
70 S. Mukhopadhyay

Fig. 6.13 Talc granulomatosis. Transbronchial lung multinucleated giant cells surrounding pale-staining for-
biopsy. (a) Foreign-body granulomas within alveolar eign particles (arrows). Note that the foreign particles and
septa. Arrow indicates granuloma shown at high magnifi- giant cells are located adjacent to a capillary (arrowhead)
cation in (b). (b) Foreign-body granuloma containing

when injected [68–71]. The active ingredient in activity, the pathologist is often the first to sug-
the pills is soluble, but excipients (“fillers” or gest the diagnosis.
“binders”) such as talc, microcrystalline cellu-
lose, and crospovidone whose function is to pro- Histologic Findings in Small Biopsies
vide bulk and other physical properties to pills Although most cases have been reported in
are insoluble [72]. These insoluble excipients autopsies and surgical lung biopsies, the diagno-
travel via the venous circulation and the right side sis can be made on transbronchial lung biopsy.
of the heart to the pulmonary circulation, where The main finding is the presence of foreign-body
they are trapped within alveolar septal capillar- granulomas located adjacent to capillaries within
ies. Subsequently, they extrude out of the capil- alveolar septa. The granulomas can be numerous,
laries into the interstitium and elicit a foreign-body prominent, and obvious or few, tiny, and subtle.
giant cell reaction. The vascular occlusion can be They usually consist of multinucleated giant cells
associated with thrombosis, pulmonary hyperten- that surround and engulf foreign particles, which
sion, and right heart failure [69, 70]. Radiologic are usually found adjacent to capillaries rather
findings include bilateral micronodular or miliary than within their lumens (Fig. 6.13a, b). The most
lung nodules. Examination of the retina can be common foreign particles are microcrystalline
helpful in supporting the diagnosis in some cases. cellulose, talc, and crospovidone (Fig. 6.14a, b).
Since patients seldom admit to such illicit Microcrystalline cellulose and talc are pale and
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 71

Fig. 6.14 Foreign material in talc granulomatosis, material is crospovidone (arrowhead). (b) Movat penta-
same case as Fig. 6.13. (a) H&E. Foreign particles sur- chrome stain. Microcrystalline cellulose stains yellow
rounded by multinucleated giant cells. The pale material (arrow); crospovidone is green (arrowhead)
is microcrystalline cellulose (arrow); the purple coral-like

colorless on H&E but strongly birefringent under Chronic Beryllium Disease (Berylliosis)
polarized light. Crospovidone is purple or baso-
philic and has a distinctive coral-like shape on Clinical and Radiologic Findings
H&E, but is not birefringent [72]. Microcrystalline Chronic beryllium disease is often mentioned in
cellulose and crospovidone are often found standard textbooks as a mimic of sarcoidosis, but
together in the same biopsy. The Movat penta- is rarely seen in practice. The diagnosis is heavily
chrome stain can be helpful in distinguishing dependent upon a history of exposure to beryl-
between microcrystalline cellulose and talc [73]. lium. The diagnosis can be confirmed by a blood
Microcrystalline cellulose stains yellow on test known as the beryllium lymphocyte prolif-
Movat, while talc stains light blue. The main eration test (BeLPT). In a recent study, 34 of 121
entity in the differential diagnosis is aspiration patients with a diagnosis of sarcoidosis reported
pneumonia caused by aspiration of gastric con- a history of exposure to metals, including 17 who
tents that include pill fragments. In contrast to reported exposure to beryllium [75]. However,
talc granulomatosis, aspiration of pill fragments none of the 34 had a positive BeLPT, suggesting
is characterized by organizing pneumonia and that even a history of beryllium exposure does
peribronchiolar granulomas that may also contain not necessarily equate to a definitive diagnosis of
aspirated vegetable fragments [74]. chronic beryllium disease.
72 S. Mukhopadhyay

Histologic Findings in Small Biopsies taining embedded granulomas or multinucleated


Although the histologic features of chronic beryl- giant cells. Cryptococcus yeasts are scattered
lium disease are said to consist of non-necrotizing within the cytoplasm of the histiocytes and giant
granulomas mimicking sarcoidosis, the published cells (Fig. 6.15a, b). Some cases show areas of
literature describes findings that have more in “dirty” necrosis, whereas others manifest as nec-
common with hypersensitivity pneumonitis rotizing granulomas similar to tuberculosis or
[76, 77]. In practice, the diagnosis is seldom histoplasmosis. Organisms may be found both
entertained in the absence of an appropriate within the necrosis and in the granulomatous rim
exposure history. (Fig. 6.16a, b). Immunocompromised patients
may show identical findings, or they may show
large numbers of organisms within alveolar
Causes of Granulomatous septa, airspaces, and capillaries with minimal or
Inflammation with Variable no granulomatous inflammation [78, 79].
Necrosis Cryptococcus is a small (4–7 μm) round yeast
that is faintly visible on H&E within necrosis as
Cryptococcosis well as in the cytoplasm of histiocytes and multi-
nucleated giant cells [1, 4]. The cell wall is usu-
Clinical and Radiologic Findings ally light staining and difficult to see at low
In contrast to other fungal yeasts that cause magnification, but a clue to the presence of
pulmonary granulomas, Cryptococcus occurs Cryptococcus is a “bubbly” appearance within
worldwide without an endemic area. It can infect the cytoplasm of the histiocytes. Multiple organ-
the lung in immunocompromised as well as immu- isms are often present in clusters within the same
nocompetent individuals. The former circum- cell. On H&E, the engulfing cell characteristi-
stance is more familiar to clinicians and cally retracts from the ingested yeast, resulting in
pathologists, especially in the setting of AIDS the formation of a halo around individual organ-
[78]. However, infections in immunocompetent isms or clusters of organisms. The thin wall of the
individuals are also well documented and in most yeast stains pale gray, and the center is colorless.
cases present as lung nodules, which may be soli- Narrow-based budding may be seen, similar to
tary or multiple, solid or cavitary, and PET posi- Histoplasma. In contrast to Histoplasma, how-
tive. In this setting, a neoplasm is usually the main ever, the yeasts are round rather than oval or
clinical concern. Such nodules are usually sam- tapered, and there is significant variation in size.
pled by needle biopsies, but transbronchial biop- GMS stains the yeasts well and often reveals far
sies may also be used to sample accessible lesions. more organisms than are apparent on H&E. The
Cultures of infected tissue are often positive. size variation is also better appreciated on GMS,
and fragmented forms may be seen (Fig. 6.15b).
Histologic Findings in Small Biopsies Although mucicarmine is often touted as the
As with other fungal organisms, the tissue reac- stain of choice for this organism, in practice it is
tion to Cryptococcus depends on the immu- rarely positive in pulmonary cryptococcal granu-
nologic status of the host. Immunocompetent lomas (Fig. 6.17a, b). Since mucicarmine stains
individuals develop a granulomatous inflamma- the capsule of the organism, it has been proposed
tory response, which most commonly consists that the lack of staining with mucicarmine implies
of large numbers of multinucleated giant cells the lack of a capsule (“capsule-deficient
scattered in a background of histiocytes and lym- Cryptococcus”). Some have found the Fontana–
phocytes, the entire lesion forming an ill-defined Masson stain to be a helpful marker of
granulomatous mass rather than discrete granu- Cryptococcus in this situation [80–83]. In most
lomas. Organizing pneumonia, characterized by cases, however, the combination of H&E and
plugs of fibroblasts in a pale-staining matrix, GMS is sufficient for accurate identification of
may be a prominent finding, occasionally con- the organisms by histology. The main fungal
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 73

Fig. 6.15 Cryptococcus. Core needle biopsy of a lung (arrow). The appearance of Cryptococcus on H&E is dis-
nodule. (a) H&E. The bubbly appearance is a clue to the tinctive and virtually pathognomonic, even more so than
presence of the organisms. The yeasts are visible on H&E, its morphology on GMS. (b) GMS highlights more organ-
although they stain lightly and can easily be missed. A isms than were evident on H&E. Note the size variation
characteristic halo is seen around some of the yeasts compared to Histoplasma (Fig. 6.4a)

yeast in the differential diagnosis is Histoplasma, Aspiration of gastric acid in persons with seizures,
which can usually be distinguished by its non- stroke, or head trauma is also a well-recognized
visibility on H&E and the lack of significant size aspiration syndrome. Finally, clinicians are also
variation [1]. familiar with aspiration of relatively large foreign
objects into the tracheobronchial tree, usually in
infants and children.
Aspiration Pneumonia (Due In recent years, an under-recognized form of
to Aspiration of Particulate Material) aspiration pneumonia has been described, char-
acterized by dyspnea, fever, cough, or recurrent
Clinical and Radiologic Findings pneumonias caused by aspiration of particulate
Aspiration pneumonia is traditionally thought to material derived from gastric contents (mainly
be a bacterial bronchopneumonia with a predilec- food/vegetable particles but occasionally pill
tion for the right lower lobe, caused by aspiration fragments) [74]. A similar process has been
of oral flora into the lungs. Patients are considered described as diffuse bronchiolar disease due to
to be at risk for aspiration if they are debilitated chronic aspiration [84]. Chest imaging shows
and have obvious risk factors such as stroke, sei- infiltrates or nodules that may involve any lobe in
zures, head trauma, alcoholism, or major surgery. either lung. Risk factors for aspiration, present in
74 S. Mukhopadhyay

Fig. 6.16 Necrotizing granuloma containing Cryptococcus. (b) At high magnification, the organisms within the necrosis
Core needle biopsy of lung nodule. (a) Low magnification. appear as round yeasts with a light-staining wall (arrow).
The necrotic center of the lesion is at the top right (arrow), Fungal yeasts are also present within the granulomatous
and the granulomatous rim is at the bottom left (arrowhead). component (arrowhead)

approximately half of such patients, include peribronchiolar airspaces. In later stages, orga-
esophageal or gastric causes such as carcinoma, nizing pneumonia and foreign-body granulomas
prior surgery, achalasia, hiatal hernia, or gastric containing foreign material may be the only
outlet obstruction, neurologic causes such as sei- findings. Finally, remote aspiration may show
zures and multiple sclerosis, and the use of drugs only rare eosinophilic vegetable particles that
such as cocaine or narcotics. The diagnosis is are usually degenerated and difficult to identify.
often overlooked clinically. The particles are usually present in lung paren-
chyma adjacent to bronchioles. The tissue reac-
Histologic Findings in Small Biopsies tion at this stage may be limited to a few
The initial tissue response to aspirated food histiocytes surrounding the degenerated vegeta-
particles consists of acute bronchopneumonia, ble material. One or several of these findings
followed by a granulomatous response charac- may be identified in small lung biopsies, the
terized by suppurative and foreign-body granu- diagnostic finding being the presence of particu-
lomas, with prominent multinucleated giant late foreign matter (Fig. 6.18a, b). The morpho-
cells containing vegetable and food particles. logic features of the tissue reaction and the
This may be accompanied by organizing pneu- diagnostic vegetable particles in lung biopsies
monia, which consists of fibroblast plugs within and resections have been illustrated in several
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 75

Fig. 6.17 Mucicarmine and Fontana–Masson staining in case, mucicarmine is negative (arrows). Negative staining
a cryptococcal pulmonary necrotizing granuloma. Same should not deter pathologists from the diagnosis. (b)
biopsy as Fig. 6.16. (a) Mucicarmine stain. As is often the Fontana–Masson. The organisms stain positive (arrow)

publications [1, 4, 74, 85–89]. It is important to The hemorrhage may be massive and life
remember that—unlike foreign material derived threatening. Diffuse alveolar hemorrhage is usu-
from pill fragments—most vegetable particles ally characterized by extensive bilateral airspace
are not birefringent. consolidation [91]. The serologic hallmark of
Goodpasture syndrome is the anti-glomerular
basement membrane antibody.
Pulmonary Vasculitis
Histologic Findings in Small Biopsies
Goodpasture Syndrome The most consistent finding is intra-alveolar hem-
orrhage characterized by varying proportions of
Clinical and Radiologic Findings fresh blood (red blood cells), fibrin, and
Goodpasture syndrome is a vasculitis that is a hemosiderin-laden macrophages. Most cases show
classic example of a pulmonary–renal syndrome no other significant findings. There is no granulo-
[90]. It affects young individuals who present matous inflammation or parenchymal necrosis.
with hemoptysis due to diffuse alveolar hemor- Capillaritis has been reported, but it has been
rhage and acute renal failure caused by rapidly described as occasional and mild [92, 93].
progressive (crescentic) glomerulonephritis. Definitive pathologic diagnosis requires demon-
There is a predilection for young male smokers. stration of linear staining in pulmonary capillaries
76 S. Mukhopadhyay

Fig. 6.18 Particulate matter aspiration. Transbronchial (b) Particulate material within multinucleated giant cells
lung biopsy. (a) Foreign-body granuloma containing neu- (short arrows). The granulomas are embedded within a
trophils. This type of granuloma is a clue to the diagnosis. background of organizing pneumonia (long arrow)

for anti-glomerular basement membrane antibodies the lungs as diffuse alveolar hemorrhage and
by immunofluorescence. Immunofluorescence for capillaritis, but ANCA-related vasculitides are
anti-glomerular basement membrane antibodies far more common in this setting [36, 95].
can be performed on renal biopsies, but it has also
been shown to be a feasible technique in trans-
bronchial lung biopsies [94]. It is important for the References
physician who performs the biopsy to submit two
tissue samples, one in formalin and the other in a 1. Mukhopadhyay S, Gal AA. Granulomatous lung
disease: an approach to the differential diagnosis.
medium appropriate for immunofluorescence
Arch Pathol Lab Med. 2010;134:667–90.
(such as Michel’s). 2. Doxtader EE, Mukhopadhyay S, Katzenstein A-LA.
Core needle biopsy in benign lung lesions: pathologic
findings in 159 cases. Hum Pathol. 2010;41:1530–5.
3. Mukhopadhyay S, Farver CF, Vaszar LT, Dempsey
Other Causes of Pulmonary Vasculitis OJ, Popper HH, Mani H, et al. Causes of pulmonary
granulomas: a retrospective study of 500 cases from
Other causes of pulmonary vasculitis include seven countries. J Clin Pathol. 2012;65:51–7.
Wegener’s granulomatosis and Churg–Strauss 4. Mukhopadhyay S, Aubry M-C. Pulmonary granulomas:
differential diagnosis, histologic features and algorith-
syndrome, which have been discussed above in
mic approach. Diagn Histopathol. 2013;19:288–97.
the section on granulomatous diseases. Systemic 5. Mukhopadhyay S, Doxtader EE. Visibility of
lupus erythematosus (SLE) can rarely manifest in Histoplasma within histiocytes on hematoxylin and
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 77

eosin distinguishes disseminated histoplasmosis from 20. Yankelevitz DF, Henschke CI. Does 2-year stability
other forms of pulmonary histoplasmosis. Hum imply that pulmonary nodules are benign? AJR Am J
Pathol. 2013;44:2346–52. Roentgenol. 1997;168:325–8.
6. Puckett TF. Pulmonary histoplasmosis: a study of 21. Mukhopadhyay S. Utility of small biopsies for diag-
twenty-two cases with identification of H. capsulatum nosis of lung nodules: doing more with less. Mod
in resected lesions. Am Rev Tuberc. 1953;67: Pathol. 2012;25 Suppl 1:S43–57.
453–76. 22. Hartel PH, Shilo K, Klassen-Fischer M, Neafie RC,
7. Ulbright TM, Katzenstein AL. Solitary necrotizing Ozbudak IH, Galvin JR, et al. Granulomatous reaction
granulomas of the lung: differentiating features and to pneumocystis jirovecii: clinicopathologic review of
etiology. Am J Surg Pathol. 1980;4:13–28. 20 cases. Am J Surg Pathol. 2010;34:730–4.
8. Deppisch LM, Donowho EM. Pulmonary coccidioi- 23. Goodwin Jr RA, Shapiro JL, Thurman GH, Thurman
domycosis. Am J Clin Pathol. 1972;58:489–500. SS, Des Prez RM. Disseminated histoplasmosis: clini-
9. Zimmerman LE. Demonstration of Histoplasma and cal and pathologic correlations. Medicine (Baltimore).
Coccidioides in so-called tuberculomas of lung: pre- 1980;59:1–33.
liminary report on thirty-five cases. AMA Arch Intern 24. Forseth J, Rohwedder JJ, Levine BE, Saubolle
Med. 1954;94:690–9. MA. Experience with needle biopsy for coccidioidal
10. Mukhopadhyay S, Wilcox BE, Myers JL, Bryant SC, lung nodules. Arch Intern Med. 1986;146:319–20.
Buckwalter SP, Wengenack NL, et al. Pulmonary nec- 25. Lombard CM, Tazelaar HD, Krasne DL. Pulmonary
rotizing granulomas of unknown cause: clinical and eosinophilia in coccidioidal infections. Chest.
pathologic analysis of 131 patients with completely 1987;91:734–6.
resected nodules. Chest. 2013;144:813–24. 26. Schwarz J. The diagnosis of deep mycoses by mor-
11. Nayak NC, Sabharwal BD, Bhathena D, Mital GS, phologic methods. Hum Pathol. 1982;13:519–33.
Ramalingaswami V. The pulmonary tuberculous 27. Chapman SW, Dismukes WE, Proia LA, Bradsher
lesion in North India: a study in medico-legal autop- RW, Pappas PG, Threlkeld MG, et al. Clinical prac-
sies. I. Incidence, nature, and evolution. Am Rev tice guidelines for the management of blastomycosis:
Respir Dis. 1970;101:1–17. 2008 update by the Infectious Diseases Society of
12. Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro America. Clin Infect Dis. 2008;46:1801–12.
A, Daley C, Gordin F, et al. An official ATS/IDSA 28. Taxy JB. Blastomycosis: contributions of morphology
statement: diagnosis, treatment, and prevention of to diagnosis: a surgical pathology, cytopathology, and
nontuberculous mycobacterial diseases. Am J Respir autopsy pathology study. Am J Surg Pathol.
Crit Care Med. 2007;175:367–416. 2007;31:615–23.
13. Kwon KY, Myers JL, Swensen SJ, Colby TV. Middle 29. Patel AJ, Gattuso P, Reddy VB. Diagnosis of blasto-
lobe syndrome: a clinicopathological study of 21 mycosis in surgical pathology and cytopathology:
patients. Hum Pathol. 1995;26:302–7. correlation with microbiologic culture. Am J Surg
14. Dhillon SS, Watanakunakorn C. Lady Windermere Pathol. 2010;34:256–61.
syndrome: middle lobe bronchiectasis and 30. Binder RE, Faling LJ, Pugatch RD, Mahasaen C,
Mycobacterium avium complex infection due to vol- Snider GL. Chronic necrotizing pulmonary aspergil-
untary cough suppression. Clin Infect Dis. losis: a discrete clinical entity. Medicine (Baltimore).
2000;30:572–5. 1982;61:109–24.
15. Fujita J, Ohtsuki Y, Suemitsu I, Shigeto E, Yamadori I, 31. Gefter WB, Weingrad TR, Epstein DM, Ochs RH,
Obayashi Y, et al. Pathological and radiological Miller WT. “Semi-invasive” pulmonary aspergillosis:
changes in resected lung specimens in Mycobacterium a new look at the spectrum of aspergillus infections of
avium intracellulare complex disease. Eur Respir the lung. Radiology. 1981;140:313–21.
J. 1999;13:535–40. 32. Yousem SA. The histological spectrum of chronic
16. Marchevsky A, Damsker B, Gribetz A, Tepper S, necrotizing forms of pulmonary aspergillosis. Hum
Geller SA. The spectrum of pathology of nontubercu- Pathol. 1997;28:650–6.
lous mycobacterial infections in open-lung biopsy 33. Katzenstein AL, Locke WK. Solitary lung lesions in
specimens. Am J Clin Pathol. 1982;78:695–700. Wegener’s granulomatosis. Pathologic findings and
17. Goodwin Jr RA, Snell Jr JD. The enlarging histoplas- clinical significance in 25 cases. Am J Surg Pathol.
moma. Concept of a tumor-like phenomenon encom- 1995;19:545–52.
passing the tuberculoma and coccidioidoma. Am Rev 34. Mark EJ, Matsubara O, Tan-Liu NS, Fienberg R. The
Respir Dis. 1969;100:1–12. pulmonary biopsy in the early diagnosis of Wegener’s
18. Mukhopadhyay S. Role of histology in the diagnosis (pathergic) granulomatosis: a study based on 35
of infectious causes of granulomatous lung disease. open lung biopsies. Hum Pathol. 1988;19:
Curr Opin Pulm Med. 2011;17:189–96. 1065–71.
19. Weydert JA, Van Natta TL, DeYoung BR. Comparison 35. Myers JL, Katzenstein AL. Wegener’s granulomatosis
of fungal culture versus surgical pathology examina- presenting with massive pulmonary hemorrhage and
tion in the detection of Histoplasma in surgically capillaritis. Am J Surg Pathol. 1987;11:895–8.
excised pulmonary granulomas. Arch Pathol Lab 36. Travis WD, Colby TV, Lombard C, Carpenter HA.
Med. 2007;131:780–3. A clinicopathologic study of 34 cases of diffuse
78 S. Mukhopadhyay

pulmonary hemorrhage with lung biopsy confirma- 54. Emanuel DA, Wenzel FJ, Bowerman CI, Lawton
tion. Am J Surg Pathol. 1990;14:1112–25. BR. Farmer’s lung: clinical, pathologic and immuno-
37. Travis WD, Carpenter HA, Lie JT. Diffuse pulmonary logic study of twenty-four patients. Am J Med.
hemorrhage. An uncommon manifestation of 1964;37:392–401.
Wegener’s granulomatosis. Am J Surg Pathol. 1987; 55. Hirschmann JV, Pipavath SNJ, Godwin JD.
11:702–8. Hypersensitivity pneumonitis: a historical, clinical, and
38. Lombard CM, Duncan SR, Rizk NW, Colby TV. radiologic review. Radiographics. 2009;29:1921–38.
The diagnosis of Wegener’s granulomatosis from 56. Morell F, Roger A, Reyes L, Cruz MJ, Murio C,
transbronchial biopsy specimens. Hum Pathol. 1990; Muñoz X. Bird fancier’s lung: a series of 86 patients.
21:838–42. Medicine (Baltimore). 2008;87:110–30.
39. Silva CIS, Müller NL, Fujimoto K, Johkoh T, Ajzen 57. Hanak V, Golbin JM, Ryu JH. Causes and presenting
SA, Churg A. Churg–Strauss syndrome: high resolu- features in 85 consecutive patients with hypersensitiv-
tion CT and pathologic findings. J Thorac Imaging. ity pneumonitis. Mayo Clin Proc. 2007;82:812–6.
2005;20:74–80. 58. Fernández Pérez ER, Swigris JJ, Forssén AV, Tourin
40. Katzenstein AL. Diagnostic features and differential O, Solomon JJ, Huie TJ, et al. Identifying an inciting
diagnosis of Churg–Strauss syndrome in the lung. A antigen is associated with improved survival in
review. Am J Clin Pathol. 2000;114:767–72. patients with chronic hypersensitivity pneumonitis.
41. England DM, Hochholzer L. Primary pulmonary spo- Chest. 2013;144:1644–51.
rotrichosis. Report of eight cases with clinicopatho- 59. Mukhopadhyay S. Pathology of hypersensitivity
logic review. Am J Surg Pathol. 1985;9:193–204. pneumonitis. 2013. http://emedicine.medscape.com/
42. Flieder DB, Moran CA. Pulmonary dirofilariasis: a article/2078434-overview. Accessed 2 Jan 2014.
clinicopathologic study of 41 lesions in 39 patients. 60. Barrios RJ. Hypersensitivity pneumonitis: histopa-
Hum Pathol. 1999;30:251–6. thology. Arch Pathol Lab Med. 2008;132:199–203.
43. Aubry M-C. Necrotizing granulomatous inflamma- 61. Fjällbrant H, Akerstrom M, Svensson E, Andersson E.
tion: what does it mean if your special stains are nega- Hot tub lung: an occupational hazard. Eur Respir Rev.
tive? Mod Pathol. 2012;25 Suppl 1:S31–8. 2013;22:88–90.
44. Baughman RP, Culver DA, Judson MA. A concise 62. Sood A, Sreedhar R, Kulkarni P, Nawoor AR.
review of pulmonary sarcoidosis. Am J Respir Crit Hypersensitivity pneumonitis-like granulomatous
Care Med. 2011;183:573–81. lung disease with nontuberculous mycobacteria from
45. Baughman RP, Teirstein AS, Judson MA, Rossman exposure to hot water aerosols. Environ Health
MD, Yeager Jr H, Bresnitz EA, et al. Clinical charac- Perspect. 2007;115:262–6.
teristics of patients in a case control study of sarcoid- 63. Embil J, Warren P, Yakrus M, Stark R, Corne S,
osis. Am J Respir Crit Care Med. 2001;164:1885–9. Forrest D, et al. Pulmonary illness associated with
46. Chevalet P, Clément R, Rodat O, Moreau A, Brisseau exposure to Mycobacterium-avium complex in hot
J-M, Clarke J-P. Sarcoidosis diagnosed in elderly tub water. Hypersensitivity pneumonitis or infection?
subjects: retrospective study of 30 cases. Chest. Chest. 1997;111:813–6.
2004;126:1423–30. 64. Kahana LM, Kay JM, Yakrus MA, Waserman
47. Hsu RM, Connors Jr AF, Tomashefski Jr JF. Histologic, S. Mycobacterium avium complex infection in an
microbiologic, and clinical correlates of the diagnosis immunocompetent young adult related to hot tub
of sarcoidosis by transbronchial biopsy. Arch Pathol exposure. Chest. 1997;111:242–5.
Lab Med. 1996;120:364–8. 65. Cappelluti E, Fraire AE, Schaefer OP. A case of “hot
48. Garwood S, Judson MA, Silvestri G, Hoda R, Fraig tub lung” due to Mycobacterium avium complex in an
M, Doelken P. Endobronchial ultrasound for the diag- immunocompetent host. Arch Intern Med. 2003;
nosis of pulmonary sarcoidosis. Chest. 2007;132: 163:845–8.
1298–304. 66. Khoor A, Leslie KO, Tazelaar HD, Helmers RA,
49. Visscher D, Churg A, Katzenstein AL. Significance of Colby TV. Diffuse pulmonary disease caused by
crystalline inclusions in lung granulomas. Mod nontuberculous mycobacteria in immunocompetent
Pathol. 1988;1:415–9. people (hot tub lung). Am J Clin Pathol. 2001;115:
50. Kawanami O, Basset F, Barrios R, Lacronique JG, 755–62.
Ferrans VJ, Crystal RG. Hypersensitivity pneumoni- 67. Tomashefski Jr JF, Hirsch CS. The pulmonary vascu-
tis in man. Light- and electron-microscopic studies of lar lesions of intravenous drug abuse. Hum Pathol.
18 lung biopsies. Am J Pathol. 1983;110:275–89. 1980;11:133–45.
51. Reyes CN, Wenzel FJ, Lawton BR, Emanuel DA. The 68. Paré JA, Fraser RG, Hogg JC, Howlett JG, Murphy
pulmonary pathology of farmer’s lung disease. Chest. SB. Pulmonary “mainline” granulomatosis: talcosis
1982;81:142–6. of intravenous methadone abuse. Medicine
52. Myers JL. Hypersensitivity pneumonia: the role of (Baltimore). 1979;58:229–39.
lung biopsy in diagnosis and management. Mod 69. Lewman LV. Fatal pulmonary hypertension from
Pathol. 2012;25 Suppl 1:S58–67. intravenous injection of methylphenidate (Ritalin)
53. Coleman A, Colby TV. Histologic diagnosis of tablets. Hum Pathol. 1972;3:67–70.
extrinsic allergic alveolitis. Am J Surg Pathol. 70. Houck RJ, Bailey GL, Daroca Jr PJ, Brazda F, Johnson
1988;12:514–8. FB, Klein RC. Pentazocine abuse. Report of a case
6 Diagnosis of Granulomatous Disease and Vasculitis in Small Lung Biopsies 79

with pulmonary arterial cellulose granulomas and 82. Lazcano O, Speights Jr VO, Bilbao J, Becker J,
pulmonary hypertension. Chest. 1980;77:227–30. Diaz J. Combined Fontana-Masson-mucin staining of
71. Tomashefski Jr JF, Hirsch CS, Jolly PN. Cryptococcus neoformans. Arch Pathol Lab Med.
Microcrystalline cellulose pulmonary embolism and 1991;115:1145–9.
granulomatosis. A complication of illicit intravenous 83. Ro JY, Lee SS, Ayala AG. Advantage of Fontana-
injections of pentazocine tablets. Arch Pathol Lab Masson stain in capsule-deficient cryptococcal infec-
Med. 1981;105:89–93. tion. Arch Pathol Lab Med. 1987;111:53–7.
72. Ganesan S, Felo J, Saldana M, Kalasinsky VF, 84. Barnes TW, Vassallo R, Tazelaar HD, Hartman TE,
Lewin-Smith MR, Tomashefski Jr JF. Embolized cro- Ryu JH. Diffuse bronchiolar disease due to chronic
spovidone (poly[N-vinyl-2-pyrrolidone]) in the lungs occult aspiration. Mayo Clin Proc. 2006;81:172–6.
of intravenous drug users. Mod Pathol. 2003;16: 85. Lagstein A, Myers JL. Common diagnostic chal-
286–92. lenges in the pathology of nonneoplastic lung dis-
73. Sigdel S, Gemind JT, Tomashefski Jr JF. The Movat eases: a case-based review. Arch Pathol Lab Med.
pentachrome stain as a means of identifying micro- 2009;133:1782–92.
crystalline cellulose among other particulates found 86. Knoblich R. Pulmonary granulomatosis caused by
in lung tissue. Arch Pathol Lab Med. 2011;135: vegetable particles. So-called lentil pulse pneumonia.
249–54. Am Rev Respir Dis. 1969;99:380–9.
74. Mukhopadhyay S, Katzenstein A-LA. Pulmonary dis- 87. Crome L, Valentine JC. Pulmonary nodular granulo-
ease due to aspiration of food and other particulate matosis caused by inhaled vegetable particles. J Clin
matter: a clinicopathologic study of 59 cases diag- Pathol. 1962;15:21–5.
nosed on biopsy or resection specimens. Am J Surg 88. Moran TJ. Experimental food-aspiration pneumonia.
Pathol. 2007;31:752–9. AMA Arch Pathol. 1951;52:350–4.
75. Ribeiro M, Fritscher LG, Al-Musaed AM, Balter MS, 89. Bulmer SR, Lamb D, McCormack RJ, Walbaum PR.
Hoffstein V, Mazer BD, et al. Search for chronic Aetiology of unresolved pneumonia. Thorax. 1978;
beryllium disease among sarcoidosis patients in 33:307–14.
Ontario, Canada. Lung. 2011;189:233–41. 90. Ioachimescu OC, Stoller JK. Diffuse alveolar hemor-
76. Freiman DG, Hardy HL. Beryllium disease. The rela- rhage: diagnosing it and finding the cause. Cleve Clin
tion of pulmonary pathology to clinical course and J Med. 2008;75:258–65.
prognosis based on a study of 130 cases from the U.S. 91. Mayberry JP, Primack SL, Müller NL. Thoracic
beryllium case registry. Hum Pathol. 1970;1:25–44. manifestations of systemic autoimmune diseases:
77. Williams WJ. A histological study of the lungs in 52 radiographic and high-resolution CT findings.
cases of chronic beryllium disease. Br J Ind Med. Radiographics. 2000;20:1623–35.
1958;15:84–91. 92. Lombard CM, Colby TV, Elliott CG. Surgical pathol-
78. Gal AA, Koss MN, Hawkins J, Evans S, Einstein ogy of the lung in anti-basement membrane antibody-
H. The pathology of pulmonary cryptococcal infec- associated Goodpasture’s syndrome. Hum Pathol.
tions in the acquired immunodeficiency syndrome. 1989;20:445–51.
Arch Pathol Lab Med. 1986;110:502–7. 93. Colby TV, Fukuoka J, Ewaskow SP, Helmers R,
79. McDonnell JM, Hutchins GM. Pulmonary cryptococ- Leslie KO. Pathologic approach to pulmonary hemor-
cosis. Hum Pathol. 1985;16:121–8. rhage. Ann Diagn Pathol. 2001;5:309–19.
80. Bishop JA, Nelson AM, Merz WG, Askin FB, Riedel 94. Beechler CR, Enquist RW, Hunt KK, Ward GW,
S. Evaluation of the detection of melanin by the Knieser MR. Immunofluorescence of transbronchial
Fontana-Masson silver stain in tissue with a wide biopsies in Goodpasture’s syndrome. Am Rev Respir
range of organisms including Cryptococcus. Hum Dis. 1980;121:869–72.
Pathol. 2012;43:898–903. 95. Zamora MR, Warner ML, Tuder R, Schwarz
81. Bavishi AV, McGarry TM. A case of pulmonary cryp- MI. Diffuse alveolar hemorrhage and systemic lupus
tococcosis caused by capsule-deficient cryptococcus erythematosus. Clinical presentation, histology, sur-
neoformans in an immunocompetent patient. Respir vival, and outcome. Medicine (Baltimore). 1997;76:
Care. 2010;55:937–41. 192–202.
Benign Tumors of the Lung
in Small Lung Biopsies 7
Mostafa M. Fraig

ship to HPV exposure and potential for malignant


Benign Tumors of the Lung transformation, albeit to a much lower extent.
They can be exophytic or endophytic (inverted).
Endobronchial Wall-Related Tumors They can also be solitary or multifocal [1].
Clinically they present as pedunculated
This group of tumors arises in the trachea or one of masses with a papillary or smooth surface or a
the major bronchi. They tend to produce symptoms slight bulge in the endobronchial wall depending
of obstruction like wheezing, difficulty in breath- on the growth pattern.
ing, or post-obstructive pneumonia. As they are On microscopic examination, the surface has
easily accessible by bronchoscopic instrumenta- bland squamous epithelium with the eosinophilic
tion and subsequently amenable to forceps-assisted glassy cytoplasm and small dark nuclei of the
biopsy, good material could be obtained for histo- metaplastic squamous epithelium. In the case of
pathologic examination and immunohistochemical the papillary variant, delicate fibrovascular cores
staining if needed. Cytologic specimens such as are seen within the papillary fronds. On the other
brushings and washings are usually not productive hand, the inverted type has islands of squamous
when mucosal surface is intact, but they can cor- epithelium that seem to be embedded in the
roborate the benign nature of the lesion by exclud- underlying stroma. In the latter case, there is usu-
ing the presence of malignant cells. Nevertheless, ally an inflammatory infiltrate near the surface
lesional material should be obtained to confirm the with some degenerative vacuoles. Mitotic fig-
benign nature of the lesion; otherwise, the speci- ures, especially atypical ones, when they are
men is considered nondiagnostic. present above the basal layer along with the pres-
ence of cytologic atypia indicate the presence of
dysplasia [2]. It is important to mention this dys-
Squamous Papilloma plasia as in cases where the lesions are multifo-
cal, subsequent lesions will appear, and follow-up
Squamous papilloma of the lower respiratory tract screening for these lesions is warranted. These do
follows many of the characteristics of its counter- not represent recurrences but rather multifocal
part in the head and neck region as in its relation- disease or inadequately excised lesions. In rare
examples, invasive squamous cell carcinoma has
M.M. Fraig, M.D. (*) been reported [3]. One of the caveats is to be
Department of Pathology and Laboratory Medicine,
careful when the scar from a previously biopsied
School of Medicine, University of Louisville,
530 South Jackson Street, Louisville, KY 40202, USA lesion is interpreted as desmoplastic reaction and
e-mail: m.fraig@louisville.edu sequestered squamous epithelium is interpreted

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 81


DOI 10.1007/978-1-4939-2575-9_7, © Springer Science+Business Media New York 2015
82 M.M. Fraig

as an invasive component. The association with Schwannoma


HPV is well documented and koilocytic effect is
usually present in those cases with dysplasia. Schwannoma or neurilemmoma is a benign
This of course is different from flat lesions where peripheral nerve sheath tumor that could occur
carcinoma in situ arises in metaplastic squamous anywhere in the body where there are peripheral
epithelium secondary to smoking or other carci- nerves. It arises from the coating sheath of the
nogenic exposure. The cytologic atypia in the lat- peripheral nerves, hence the name. It can occur as
ter is significant and is prerequisite on cytologic a sporadic tumor or as part of Von Recklinghausen’s
and small biopsy specimens to diagnose disease neurofibromatosis, type I. In the latter
malignancy. instance, it can attain the plexiform type where
the tumor follows nerve twigs and appears as
infiltrative. Malignant transformation in the lung
Pleomorphic Adenoma is extremely rare [4].
Classically, the tumor is comprised of the two
Pleomorphic adenoma (PA) or benign mixed patterns, compact spindle cells (Antony A) alter-
tumor of the salivary gland can arise in the sero- nating with areas of less cellular loose and myxoid
mucinous glands that is present around the tra- areas (Antony B). Infiltration by eosinophils or mast
cheobronchial tree of the lung. In fact many of the cells can be noted. The blood vessels are thick and
salivary gland tumors occur in this area of sali- there are areas of collagen deposition with cells
vary gland analog [4]. The tumor can occur in palisading around recognized as Verocay bodies.
young adults as well as older population with a There are variants such as cellular one where the
slight predilection to females. The lesion usually tumor is mostly comprised of wavy spindle cells
presents as a bulging mass with smooth intact with no loose areas. Another is the “ancient
mucosal surface. On imaging, the lesion is usually schwannoma” where the cells show atypical mor-
centrally located or in close relationship to the phology with enlarged hyperchromatic nuclei as a
bronchial tree with a well-circumscribed outline. sign of degeneration which could be mistaken for
On histopathologic examination, there are the malignant transformation. Immunohistochemical
usual myoepithelial cells with plasmacytoid fea- stains for S100, EMA, GFAP, and Ki67 could con-
tures with interspersed myxoid or cartilaginous firm the neuronal nature of the lesion, and the latter
matrix. The chondroid matrix is less common in reveals the indolent nature of its behavior. Negative
the lung’s PA than in the salivary gland ones. In staining for cytokeratin could rule out sarcomatoid
cytologic material, the presence of magenta- carcinoma if such a differential is entertained.
colored fibrillary matrix with the plasmacytoid
“hyaline cells” is the very characteristic on modi-
fied Giemsa stains. Small biopsy material may Granular Cell Tumor
present a challenge with the differential diagno-
sis including adenoid cystic carcinoma, mucoepi- These tumors could occur anywhere in the body
dermoid carcinoma, and sarcomatoid carcinoma. and they are believed to be of schwannian origin.
Staining for myoepithelial marker such as cyto- As such they tend to follow nerve twigs creating
keratin and smooth muscle actin could help in islands of tumor cells on the cut surface that
those instances, and the lack of cytologic atypia could be mistaken for an infiltrative tumor.
or mitotic activity should eliminate the more Clinically and radiologically, they present in
aggressive malignant tumors like sarcomatoid the lumen of the tracheobronchial tree as peduncu-
carcinoma. Malignant transformation in those lated masses with smooth surface. They can attain
tumors is extremely rare, and there is a no histo- a large size up to 5.0 cm in maximum diameter.
morphologic feature to predict this type of bio- Under microscopic examination, the tumor is
logic behavior. comprised of uniform cells with abundant finely
7 Benign Tumors of the Lung in Small Lung Biopsies 83

Fig. 7.1 Granular cell tumor with polygonal cells, granu- Fig. 7.2 Low-magnification view of a small biopsy illus-
lar eosinophilic cytoplasm, and uniform small nuclei is trating the mixture of fat, connective tissue, and cartilage
usually located under the bronchial mucosa within the in a pulmonary hamartoma
endobronchial wall

granular eosinophilic cytoplasm and uniform cen- out malignancy, especially in cases with concur-
trally located nucleus (Fig. 7.1). The cytoplasm is rent malignancies. Whenever there is limited
believed to be full of lysosomes as proven by material to make the diagnosis, the use of adjunct
ultrastructural studies and also evidenced by information from clinical and radiologic findings
immunoreactivity to CD 68, a macrophage becomes paramount to avoid making the wrong
marker. The tumor shares some of the immuno- diagnosis in these cases.
histochemical profile with schwannoma and as
such is positive for S100, CD56, and CD57
immunostains. The polygonal cells with their Pulmonary Hamartoma
characteristic granular cytoplasm are relatively
easy to recognize on fine-needle aspiration mate- This is a benign neoplasm composed of a mixture
rial. Obtaining material for immunohistochemical of mesenchymal elements such as cartilage, fat,
stains to rule out neuroendocrine tumors such as connective tissue, and smooth muscle with
carcinoid is helpful in those situations. None of entrapped epithelium. They present as incidental
these tumors have ever been reported to transform finding on imaging studies in older patients in the
to malignant ones [5]. sixth decade. They are more common in males
than females. They can be identified as such on
imaging where they can be recognized as coin
Parenchymatous Tumors lesions with a “soap bubble” or “popcorn” appear-
ance and with occasional calcification. They can be
While most of these tumors occur within the in the endobronchial wall producing symptoms of
peripheral parenchyma of the lung, they are not obstruction and rarely hemoptysis due to the tumor
restricted to these areas. Most of them are silent impinging on the endobronchial wall vessels [6].
and only discovered incidentally during work-up Histologically, they are represented by lobules
for other diseases or screening for lung cancer. of cartilage and intervening fat and entrapped
Some of them, like hamartoma, have characteris- epithelium. In cytologic preparations, the
tic radiologic features to make the diagnosis very material is usually represented by fragments of
accurate; others require tissue sampling to rule cartilage with scant other elements (Fig. 7.2).
84 M.M. Fraig

The aspirator will usually report that the lesion is


hard and is well circumscribed. On modified
Giemsa stain, the cartilaginous material can be
appreciated with its magenta color.
On small biopsies, the presence of two mesen-
chymal elements helps in establishing the diag-
nosis. The presence of fat in close proximity to
the cartilage in small biopsy should be enough to
make the diagnosis. The lack of cytologic atypia
and pleomorphism, along with the radiologic
findings, should aid in establishing the benign
nature of the lesion.

Alveolar Adenoma

Alveolar adenoma (AA) is a well-circumscribed Fig. 7.3 Papillary carcinoma of the lung showing the
lesion within the lung parenchyma. They are com- delicate fibrovascular cores within the tumors and high-
prised of uniform cells of alveolar derivation. The grade nuclear features. Large pleomorphic nuclei with
size can be large (up to 6.0 cm), but the circum- prominent nucleoli are seen in a disordered pattern lining
the fronds in contrast to Fig. 7.4
scription and slow rate of growth if interval radio-
logic evaluation is applied should reveal the
benign nature of the lesion [7]. When the lesion is biopsies. It is important to pay close attention to
completely removed, the microscopic examina- the bland cytologic features, which contrasts with
tion reveals cystic spaces lined with what would those of true papillary carcinoma of the lung
appear as type II pneumocytes. On a small biopsy where the nuclei have much more malignant fea-
specimen, the differential diagnosis would include tures with vesicular nuclei having an irregular
adenocarcinoma in situ, adenocarcinoma with lep- nuclear membrane and very prominent nucleoli
idic pattern, and pneumocytoma (sclerosing hem- [9] (Fig. 7.3). Papillary carcinoma of the lung is
angioma). Immunoreactivity to TTF-1 and napsin one of the most aggressive tumors among the
A does not help in discerning the malignant or adenocarcinoma subtypes. The papillary frond
benign nature of the lesion [8]. Awareness of the usually has an appreciable fibrovascular core,
radiologic findings and bland natures of the cells is and there is no tertiary structure where the cells
important in working up these cases. Cystic spaces are piling up without a fibrovascular core
could be interpreted as emphysematous changes (Fig. 7.4a, b).
on a small-needle core biopsy. Again reviewing Immunohistochemical stains of this lesion
the radiologic findings is very helpful. would follow the alveolar histogenesis with posi-
tive staining with TTF-1, napsin A, and CK7.
There could be variable staining for CEA which
Papillary Adenoma may complicate the diagnosis of a benign entity
[10, 11].
This is another tumor with a well-circumscribed
outline and is an incidental finding in most cases.
Radiologic imaging reveals a mass within the Pneumocytoma “Sclerosing
lung that could be in the differential diagnosis Hemangioma”
with malignant tumors.
On microscopic examination, the presence of This is a tumor that was first described as an analog
papillary fronds is alarming for papillary to a skin lesion because of the presence of blood
carcinoma on cytologic material or needle core lakes and hemosiderin within the tumor [12].
7 Benign Tumors of the Lung in Small Lung Biopsies 85

Fig. 7.4 (a, b) The fibrovascular cores of papillary ade- higher magnification, the cells show apical snouts and fre-
noma are larger, and the lining cells have low-grade quent pseudoinclusions
nuclear features with a streaming pattern. In (b) under

Later on, the histogenetic origin was proved to be within the tumor or the area immediately next to
from alveolar cells. The name has been controver- it [14].
sial for decades, and recently Shimosato sug- The problem is usually encountered either on
gested the name pneumocytoma to correctly frozen section or small biopsies and fine-needle
represent the cell of origin of this tumor as an aspiration biopsies. In these examples the differ-
uncommitted cell of the alveolar lining [13]. The ential diagnosis with adenocarcinoma in situ can
tumor can occur at any age but it has a predilec- be problematic. Correlation with history of smok-
tion to women with male to female ratio of 1:5. It ing and imaging studies can avoid the overcall. It
can occur anywhere in the lung but usually it is in is also important to pay close attention to the
the periphery. It appears as a coin or oval lesion cytologic morphology. The nuclei are rounded
and it could be surrounded by a crescent or a rim and the N/C ratio is low to moderate. The pres-
of air around the lesion. It seldom shows any clin- ence of nuclear pseudoinclusions should not be
ical symptoms and is discovered incidentally. interpreted as a sign of malignancy because it is
On microscopic examination when the tumor frequently encountered even in benign reactive
is completely resected, the circumscription is type II pneumocytes.
appreciated and there are two types of cells: one The cells follow the same pattern of those
is similar to type II pneumocytes lining the alve- derived from alveolar lining, being positive for
olar spaces and the papillary fronds within the TTF-1 and napsin A, CK7, and EMA. The tumor
tumor (Fig. 7.5a, b). The others are a group of is negative for mesothelial markers WT-1, cal-
cuboidal cells representing the stroma or the retinin, and CK5/6. It is also negative for the neu-
interstitium within the lesion. Both types are roendocrine markers, synaptophysin,
polygonal or cuboidal in shape with uniform chromogranin, and CD56 [15].
nuclei and a fair amount of cytoplasm. Clusters Rarely, these tumors could metastasize to
of foamy histiocytes or calcifications could be draining lymph nodes. However, even when they
seen within the solid areas or within the papil- do, the course is indolent and the patient has been
lary fronds. Hemosiderin could be seen deposited reported not to die from the disease.
86 M.M. Fraig

Fig. 7.5 (a, b) Pneumocytoma or the so-called sclerosing noted in the spaces. Sclerotic areas are also present in (b),
hemangioma shows a combination of lining cells and and in all instances the cells are similar in morphology
solid areas between cystic spaces (a). Blood lakes are and histogenesis

Solitary Fibrous Tumor and rounded or globoid outline should alert the
pathologist to the benign nature and attempts to
Solitary fibrous tumors of the lung and pleura obtain material for immunohistochemical stains.
have been confused with mesothelioma because Immunostains show the tumor cells to be posi-
of the old name of “localized fibrous mesotheli- tive for CD34, CD99, and bcl-2. They are nega-
oma.” Later work proved these tumors to be of tive for pan cytokeratin, excluding sarcomatoid
multipotent fibrous derivation from the subpleu- carcinoma and mesothelioma. They are also nega-
ral fibrous tissue. They are considered of low tive for EMA, arguing against synovial sarcoma.
malignant potential due to the fact that they can They are negative for S100, desmin, and smooth
attain some features associated with malignancy muscle actin, ruling out neuronal as well as
like hypercellularity, increased mitotic activity smooth muscle tumors, which are in the differen-
(>4/10 HPF) with associated hemorrhage, and tial diagnosis. Rare tumors such as hemangioperi-
necrosis. In addition they can be locally aggres- cytoma, which solitary fibrous tumors have been
sive and recur after excision. claimed to be representative of, are usually posi-
Radiologically, they range from 1.0 to 36 cm tive for vascular tumor markers such as CD31. As
in their greatest dimension. They are usually with any spindle cell neoplasm, obtaining optimal
closely associated with the pleura or embedded material for immunohistochemistry is crucial for
within the lung parenchyma with a pedicle attach- proper characterization of these lesions.
ing it to the overlying pleura. Areas of calcifica-
tion or hemorrhage can be seen on imaging [16].
On small biopsies these tumors show as fibro- Clear Cell Tumor
blastic proliferation with bland nuclei and a “pat-
ternless pattern” of growth. The ropey collagen Clear cell tumor of the lung which was also
described in resected tumors is usually not appre- known as “sugar tumor” due to the presence of
ciated in small biopsies. The well-defined borders glycogen in the cytoplasm of these cells is a
7 Benign Tumors of the Lung in Small Lung Biopsies 87

member of the perivascular epithelioid cell neoplastic syndrome which can manifest with
tumors or PEComas. These include angiomyoli- fever, weight loss, anemia, and hyperglobulin-
poma, lymphangioleiomyomatosis (LAM), and emia. They can be clinically silent and reveal
rare tumors of the falciform ligament. They share themselves only during screening for other
the immunohistochemical profile of having a conditions.
melanocytic and occasionally smooth muscle Histologically, they are characterized by a
markers. They are positive for MART-1, HMB45, fibroblastic proliferation with vague fascicles of
and MITF-1 [17]. spindle cells traversing the mass with a loose
Clear cell tumors of the lung usually present myxoid background. The latter is considered the
as well-circumscribed lesions in the periphery of classic “tissue culture” pattern. Another one is
the lung. They occur in patients >40 years of age quite cellular and shows even sclerotic collage-
and are 1–5 cm in maximum diameter. nous stroma with sparse spindle cells. Cellular
Microscopically, they are characterized by pleomorphism and mitotic figures can be seen.
polygonal cells with clear to eosinophilic cyto- The outline of the tumor shows the interface with
plasm and small uniform nuclei. There are deli- the remainder of the lung to be irregular in
cate fibrovascular cores traversing among nests resected specimens [18].
of clear cells. Normal structures of the lung could In small biopsies the tumor tends to have vari-
be entrapped within the tumor. able areas of fibrous proliferation admixed with
On small biopsies the clear eosinophilic cyto- inflammatory cells. The latter include lympho-
plasm would invoke a wide differential diagnosis cytes, macrophages, neutrophils, and eosinophils.
including metastatic tumors like clear cell carci- As in any spindle cell neoplasm, immunohis-
noma from the kidney, clear cell melanoma, and tochemistry is very essential in characterizing
clear cell sarcoma. The tumor cells are positive these lesions.
for PAS stain without diastase digestion due to In the case of inflammatory myofibroblastic
the rich glycogen content. They are also positive tumors, they are positive for smooth muscle actin
for additional markers, CD117 and vimentin, and calponin. They are negative for desmin and
besides the melanocytic ones as mentioned CD34. In about 40 % of cases, they are positive
above. The negative reactivity to epithelial mark- for ALK antibodies, which were first identified as
ers such as EMA and cytokeratin excludes clear a marker for anaplastic lymphoma and for p80
cell carcinomas in general. Positive staining for protein [19, 20]. As in all spindle cell neoplasms
actin and cytoplasmic staining with Myo-D1 of the lung, sarcomatoid carcinoma is an impor-
should help in differentiating this tumor from tant differential diagnosis, and lack of reactivity
melanocytic tumors. to cytokeratin and other epithelial markers is
very helpful in this regard.

Inflammatory Myofibroblastic Tumor


Mucinous Cystadenoma
In the past these were described as inflammatory
pseudotumors as they were rich in inflammatory Mucinous cystadenoma of the lung is similar to
cells and what thought to be an organizing reac- other tumors arising in other organs in the GI
tive fibroblastic proliferation. Extensive studies tract and gynecologic locations. It is a cystic neo-
in the 1990s proved the presence of clonal expan- plasm characterized by proliferation of a layer of
sion and the presence of a neoplastic process. mucinous epithelium lining a cyst with pools of
These tumors present mostly at a young age in viscid mucin. They present in the periphery of the
half the cases, but they can be seen in any age lung or next to one of the terminal bronchi. The
group and can produce symptoms if they extend fibrous shell is characteristic and is helpful on
in the airways. They can be associated with para- frozen section diagnosis.
88 M.M. Fraig

Radiologic imaging shows these as well- in adults: a clinicopathologic and in situ hybridization
study of 14 cases combined with 27 cases in the litera-
circumscribed lesions in the periphery with a cys-
ture. Am J Surg Pathol. 1998;22(11):1328–42.
tic center. 3. Cook JR, Hill DA, Humphrey PA, Pfeifer JD,
These are tumors which can have an aggres- El-Mofty SK. Squamous cell carcinoma arising in
sive behavior. The latter fact explains why they recurrent respiratory papillomatosis with pulmonary
involvement: emerging common pattern of clinical
are considered borderline tumors [21].
features and human papillomavirus serotype associa-
Small biopsy from these tumors can be difficult tion. Mod Pathol. 2000;13(8):914–8.
to sort out without the clinical and radiologic cor- 4. Moran CA, Suster S, Askin FB, Koss MN. Benign
relation as most malignant mucinous lesions of the and malignant salivary gland-type mixed tumors of
the lung. Clinicopathologic and immunohistochemi-
lung could have bland mucinous epithelium.
cal study of eight cases. Cancer. 1994;73(10):
2481–90.
5. Thomas de Montpreville V, Dulmet EM. Granular cell
Other Tumors tumours of the lower respiratory tract. Histopathology.
1995;27(3):257–62.
6. van den Bosch JM, Wagenaar SS, Corrin B, Elbers
Some other tumors can exist in the lung, but they JR, Knaepen PJ, Westermann CJ. Mesenchymoma of
are either too small for biopsy and they are only the lung (so called hamartoma): a review of 154
encountered in resected specimens. Minute parenchymal and endobronchial cases. Thorax.
1987;42(10):790–3.
meningothelial-like nodules are an example of
7. Burke LM, Rush WI, Khoor A, Mackay B, Oliveira P,
those small lesions. Mature cystic teratoma, Whitsett JA, et al. Alveolar adenoma: a histochemi-
chondroma, and heterotopic intrapulmonary thy- cal, immunohistochemical, and ultrastructural analy-
moma can rarely present in the lung. In these sis of 17 cases. Hum Pathol. 1999;30(2):158–67.
8. Cavazza A, Paci M, De Marco L, Leporati G, Sartori
examples where the morphology is not typical
G, Bigiani N, et al. Alveolar adenoma of the lung: a
and characteristic, a generic diagnosis of a neo- clinicopathologic, immunohistochemical, and
plastic process and additional tissue could lead molecular study of an unusual case. Int J Surg Pathol.
eventually to the proper characterization of the 2004;12(2):155–9.
9. Silver SA, Askin FB. True papillary carcinoma of the
lesion. The important rule in all of these lesions is
lung: a distinct clinicopathologic entity. Am J Surg
to avoid using malignant terms to describe them Pathol. 1997;21(1):43–51.
unless one is absolutely sure of the diagnosis. 10. Fantone JC, Geisinger KR, Appelman HD. Papillary
The lung is a vital organ that is needed for the adenoma of the lung with lamellar and electron dense
granules. An ultrastructural study. Cancer.
optimal quality of life, especially in old age. Loss
1982;50(12):2839–44.
of function from any lobes of the lung can com- 11. Fukuda T, Ohnishi Y, Kanai I, Emura I, Watanabe T,
promise this quality of life. Removing a lobe of Kitazawa M, et al. Papillary adenoma of the lung.
the lung when there is a sizable irreversible dis- Histological and ultrastructural findings in two cases.
Acta Pathol Jpn. 1992;42(1):56–61.
ease, tumor or when the lobe nonfunctioning can
12. Liebow AA, Hubbell DS. Sclerosing hemangioma
be justified on it own merits but for a small benign (histiocytoma, xanthoma) of the lung. Cancer.
nodule, it can be quite tragic. Using the clinical 1956;9(1):53–75.
and radiologic imaging data is crucial to avoid 13. Shimosato Y. Lung tumors of uncertain histogenesis.
Semin Diagn Pathol. 1995;12(2):185–92.
this type of mistake.
14. Yousem SA, Wick MR, Singh G, Katyal SL, Manivel
JC, Mills SE, et al. So-called sclerosing hemangiomas
of lung. An immunohistochemical study supporting a
respiratory epithelial origin. Am J Surg Pathol.
References 1988;12(8):582–90.
15. Devouassoux-Shisheboran M, Hayashi T, Linnoila
1. Al-Saleem T, Peale AR, Norris CM. Multiple papil- RI, Koss MN, Travis WD. A clinicopathologic study
lomatosis of the lower respiratory tract. Clinical and of 100 cases of pulmonary sclerosing hemangioma
pathologic study of eleven cases. Cancer. with immunohistochemical studies: TTF-1 is
1968;22(6):1173–84. expressed in both round and surface cells, suggesting
2. Flieder DB, Koss MN, Nicholson A, Sesterhenn IA, an origin from primitive respiratory epithelium. Am J
Petras RE, Travis WD. Solitary pulmonary papillomas Surg Pathol. 2000;24(7):906–16.
7 Benign Tumors of the Lung in Small Lung Biopsies 89

16. Goodlad JR, Fletcher CD. Solitary fibrous tumour 19. Coffin CM, Patel A, Perkins S, Elenitoba-Johnson
arising at unusual sites: analysis of a series. KS, Perlman E, Griffin CA. ALK1 and p80 expres-
Histopathology. 1991;19(6):515–22. sion and chromosomal rearrangements involving
17. Gaffey MJ, Mills SE, Askin FB, Ross GW, Sale GE, 2p23 in inflammatory myofibroblastic tumor. Mod
Kulander BG, et al. Clear cell tumor of the lung. A Pathol. 2001;14(6):569–76.
clinicopathologic, immunohistochemical, and ultra- 20. Chan JK, Cheuk W, Shimizu M. Anaplastic lym-
structural study of eight cases. Am J Surg Pathol. phoma kinase expression in inflammatory pseudotu-
1990;14(3):248–59. mors. Am J Surg Pathol. 2001;25(6):761–8.
18. Coffin CM, Hornick JL, Fletcher CD. Inflammatory 21. Gao ZH, Urbanski SJ. The spectrum of pulmonary
myofibroblastic tumor: comparison of clinicopatho- mucinous cystic neoplasia: a clinicopathologic and
logic, histologic, and immunohistochemical features immunohistochemical study of ten cases and review
including ALK expression in atypical and aggressive of literature. Am J Clin Pathol. 2005;124(1):
cases. Am J Surg Pathol. 2007;31(4):509–20. 62–70.
Malignant Tumors of the Lung
in Small Lung Biopsies 8
Mostafa M. Fraig

small carcinoma with specific clinical and molecular


Background implications. Combined or mixed differentiation
could also occur.
The first classification of lung tumors was pub- The majority of tumors in the lung are carcino-
lished in 1967 by the International Association for mas (90–95 %), with the remainder 5 % represent-
the Study of Lung Cancer under the auspices ing bronchial carcinoid and 2–5 % representing
of the World Health Organization. It was updated mesenchymal or other miscellaneous tumors.
for the first time in 1981. The third edition of the
same classification was published in 1997. The
last published WHO classification was in 2004. Adenocarcinoma
The WHO adopts a policy of making any clas-
sification based on methods and criteria that are This is a malignant epithelial tumor with glandular
easy to apply and reproduce in any setting any- formation or mucin production. It is the most
where in the world. For histopathologic diagnosis, common type of lung cancer, especially in women
hematoxylin and eosin (H&E)-stained histologic and nonsmokers. The glandular differentiation
sections are the standard type of morphology that could take the forms of acinar, papillary, solid, or
is followed for classification. The role of ancillary micropapillary formations.
studies such as immunohistochemistry (IHC) or On imaging, the tumor could present as a
molecular markers should be to confirm not to speculated nodule, ground-glass opacities or
make the diagnosis. Differentiation along the epi- pneumonia-like picture, or multiple nodules with
thelial versus mesenchymal types of tissue would central lucency (cheerios pattern).
serve as a basis to differentiate most carcinomas Grossly the tumors are soft white-tan with
from sarcomas. Within the epithelial category, car- close proximity to the pleural surface or with
cinomas would be distinguished according to their pleural puckering when the pleura is involved by
further differentiation along glandular or squa- the tumor or the tissue reaction around it. Areas of
mous lineage. Undifferentiated carcinoma and necrosis could be seen as well as carbon pigments
neuroendocrine differentiation are reserved for if the patient has been a smoker.
Sampling these lesions became very versatile in
recent years with the advent of electromagnetic
M.M. Fraig, M.D. (*) navigational biopsy where a probe could be
Department of Pathology and Laboratory Medicine,
School of Medicine, University of Louisville,
directed to the periphery of the lung using a
530 South Jackson Street, Louisville, KY 40202, USA bronchoscopic approach, and a sizable biopsy
e-mail: m.fraig@louisville.edu can be obtained for diagnosis and molecular testing

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 91


DOI 10.1007/978-1-4939-2575-9_8, © Springer Science+Business Media New York 2015
92 M.M. Fraig

Fig. 8.1 Adenocarcinoma in situ at the interface with Fig. 8.2 Invasive adenocarcinoma of the lung on a small
normal lung shows an abrupt transition, unlike reactive biopsy showing acini of tumor with lumen formation infil-
type II pneumocytes where they blend gradually with nor- trating into the lung parenchyma. Blue mucin can be seen
mal type I. The cytologic atypia is higher than that seen in easily into lumens
reactive epithelium, and there is no evidence of invasion
or host response (lymphocytic infiltrate)

if needed. In addition, fiduciary markers can be


inserted for future radiotherapy if indicated.
CT-guided fine needle aspiration and core biopsy
have achieved high rate of diagnostic yield and
gained popularity in community settings.
Microscopically the tumor could present with a
“lepidic pattern,” an expression used to describe
birds sitting on a fence, where the tumor cells line
the alveolar spaces without invading or invoking
much of host response (Fig. 8.1). This type of ade-
nocarcinoma of the non-mucinous type is consid-
ered an adenocarcinoma in situ (AIS), as the
prognosis of these tumors is considered 100 %
survival at 5 years on complete resection. Once the
tumor invades, and as long as the focus of invasion Fig. 8.3 Papillary carcinoma of the lung is comprised of
is less than 5 mm, the prognosis is still close to papillary fronds with delicate fibrovascular cores and loss
100 % survival at 5 years. In the latter case, the of polarity of the nuclei. High-grade nuclear features are
tumor is called adenocarcinoma with minimal the norm. Sometimes the feature is partially present in
another pattern
invasion (AMI) [1]. The distinction between the
last two entities on a small biopsy can be extremely
difficult in the absence of architectural changes to and those with ground-glass attenuation suggestive
suggest invasion and the ability to estimate the size of lepidic pattern [2]. Any invasion beyond 5 mm
of invasion. However, radiological image has makes the tumor an invasive adenocarcinoma.
achieved a high level of accuracy in predicting The subtyping of this tumor is based on the pre-
the size of invasion in these tumors based on the dominant pattern, acinar (Fig. 8.2), solid with mucin
difference between areas of high optical density production, papillary (Fig. 8.3), and micropapillary,
8 Malignant Tumors of the Lung in Small Lung Biopsies 93

and these carry with them an increasing risk of


worse prognosis, respectively.
Diagnosis on small biopsies of well-
differentiated adenocarcinoma is fraught with
several difficulties; distinguishing this tumor
from reactive type II pneumocytes, especially in
areas around fibrosis and chronic inflammation,
is one that is underappreciated in lung pathology.
Looking for cilia and cytologic features is very
important before making the diagnosis of cancer.
The history of smoking, recent weight loss, and
the radiologic findings are some of the helpful
findings to support the diagnosis of malignancy
in those cases.
A tumor formerly known as mucinous bron-
chioloalveolar carcinoma is now called muci-
nous adenocarcinoma as these tumors have a Fig. 8.4 Mucinous adenocarcinoma in a small biopsy is
different presentation and immunohistochemical difficult to recognize because of the overlap with bron-
and molecular profile than AIS. These tumors chial epithelium exhibiting goblet cell hyperplasia.
could present as a multifocal disease, pneumonia- Lack of cilia and the network of mucinous epithelium
along with the tufting seen here are strong evidence to
like pattern, or as a solitary irregular area of con- the nature of the lesion, especially in the setting of large
solidation on imaging studies. They are mass in the lung
characterized by a copious amount of secreted
mucin which patients would cough out. The cells
usually have enlarged nuclei with irregular
nuclear contours but are basally located with an
abundant amount of columnar cytoplasm
(Fig. 8.4). They differ from non-mucinous type
in their reactivity to CK20 and variable staining
with CK7 and TTF-1; the latter two are consis-
tently positive in AIS. Mucinous adenocarci-
noma has high level of Kras mutations as it is
also associated with history of smoking [3]. For
those reasons, the biology and prognosis of
mucinous adenocarcinoma are thought to repre-
sent a different entity from AIS even though they
were previously lumped together under the term
bronchioloalveolar carcinoma.
Mucinous adenocarcinoma should always be
differentiated from metastatic counterparts from
other organs such as breast colloid carcinoma, pan- Fig. 8.5 Minute fragment of mucinous adenocarcinoma
creas, and colon in addition to gynecologic tumors in a needle core biopsy of the lung showing tufting of the
with similar morphology. Reactivity to such mucinous epithelium and lack of cilia
markers as CDX2 is helpful in this regard, but the
clinical correlation with lesions or history of carci- irregularity on cytologic material are helpful
noma in these organs is always helpful [4, 5]. features to make such diagnosis (Fig. 8.5).
Small biopsies from mucinous adenocarci- Mucinous carcinoma in situ is extremely rare and
noma can be difficult to diagnose. However, the most of mucinous carcinomas of the lung are
presence of tufting and nuclear membrane invasive by the time they are being diagnosed.
94 M.M. Fraig

The majority of patients with adenocarcinoma


are smokers, but the occurrence of AIS is com- Squamous Cell Carcinoma
mon in nonsmokers. The lesion starts as a small
precancerous lesion known as atypical adenoma- This is the second most common carcinoma in
tous hyperplasia (AAH) where atypical cells with the lung. It is characterized by squamous differ-
early molecular genetic aberration similar to those entiation with keratinization and formation of
in AIS appear in the lung. Their size is usually less intercellular bridges corresponding to desmo-
than 5 mm in diameter and could be encountered somes on the ultrastructural level.
in resected lungs for any other reason, and they Over 90 % of squamous cell carcinomas occur
are considered as “field defect.” in smokers. They are usually preceded by squa-
By immunohistochemistry the majority of mous metaplasia and dysplasia of the bronchial
adenocarcinomas react positively to thyroid lining epithelium before progressing to squa-
transcription factor (TTF-1) in about 80 % of mous cell carcinoma in situ and finally into inva-
cases. Another marker is napsin A, which stains sive squamous cell carcinoma. The tumor is
surfactant producing cells as it also stains other usually centrally located; however, peripherally
tumors from the kidney, thyroid, and others [6]. located tumors occur in a minority of cases.
These two markers are very useful in differenti- On imaging, the central location of tumor and
ating poorly differentiated adenocarcinoma from proximity to relatively large bronchi and bronchi-
poorly differentiated squamous cell carcinoma, oles are associated with obstruction and occlu-
along with other markers for squamous cell dif- sion with the resultant collapse or atelectasis of
ferentiation as p63 and/or CK5/6. Recently, lung segments distal to the tumor. These tumors
cocktails of these markers using different chro- could also extend to the hilar or mediastinal
mogens have been used in performing the stains lymph nodes appearing as masses in those areas.
on limited material. TTF-1 and napsin A could Squamous cell carcinoma is the most common
be combined using alkaline phosphatase (brown tumor to cavitate resulting in a thick-walled cav-
color and nuclear pattern) combined with horse- ity with areas of central lucency. When they
radish or victor red (red color and cytoplasmic). occur in the superior sulcus of the lung, they are
Both could stain the same cells concurrently. called Pancoast tumor. They could erode into
The same could be done with p40 (nuclear) and the posterior ribs and could cause Horner’s
CK5/6 (cytoplasmic) [7]. Mucin stain could also syndrome.
be used as a cheap and quick method in identify- Grossly, the tumor is white or gray with black
ing intracellular mucin secretion and as a proxy carbon pigments throughout. They may show
for glandular differentiation. necrotic center with stellate-shaped periphery.
On the molecular level, adenocarcinomas There may be central necrosis or polypoid growth
express higher frequency of Kras mutation, espe- pattern, especially when the tumor extends into
cially in smokers (30 %) as compared to non- the bronchial lumen.
smokers (5 %). Microscopically, the tumor shows keratin for-
The revolutionary discovery of epithelial growth mation, the amount of which is proportionate to
factor receptor (EGFR) mutation in patients with the degree of differentiation; more differentiated
adenocarcinoma (especially women nonsmokers tumors have more keratinization. The cells have
from Asian descent) and the introduction of tyro- large dark nuclei and a moderate amount of waxy
sine kinase inhibitor chemotherapy made it imper- eosinophilic cytoplasm. Sometimes the cells
ative to identify patients with adenocarcinoma and have a smaller amount of cytoplasm with dark
to test these patients for the mutation. Other muta- and amphophilic color and peripheral palisading
tions such as EML 4-ALK mutation which is similar to that of basal cell carcinoma of the skin,
encountered less frequently than EGFR ones which invoked the name basaloid variant of
opened the door for more molecular testing and squamous cell carcinoma (Fig. 8.6). When the cells
targeted therapy to these patients [8]. show cytoplasmic clearing, this would indicate
8 Malignant Tumors of the Lung in Small Lung Biopsies 95

on the presence or absence of neuroendocrine


differentiation.
Immunohistochemistry is very helpful in
differentiating poorly differentiated squamous cell
carcinoma from other types of carcinomas.
Squamous cell carcinoma is usually positive for
pancytokeratin, high molecular weight cytokeratin,
and CEA. Two specific markers that are frequently
used in practice for squamous differentiation are
p63 and CK5/6. A more specific clone of p63
came into use recently and is known as p40 [7].
On the molecular level, squamous cell carci-
noma harbors EGFR, in about 84 % of cases.
Expression of Her-2/neu is more frequent in ade-
nocarcinoma but rare in squamous cell carcinoma
as is the case with Kras activation.
Fig. 8.6 Basaloid squamous cell carcinoma with periph-
eral palisading of the basal layer, which has darker nuclei
and high N/C ratio. These cells can look similar to small Small Cell Carcinoma
cell carcinoma on cytologic material
It is a malignant epithelial tumor characterized
by small cells (two times the size of a resting
lymphocyte) with scant cytoplasm, ill-defined
borders, and granular chromatin and absent or
inconspicuous nucleolus. Extensive necrosis is
usually present and mitotic activity is high.
The cells exhibit nuclear molding where the
nuclei are set together as a cobblestone pattern.
The tumor shows a central location as in squa-
mous cell carcinoma with early spread to hilar or
mediastinal lymph nodes. The tumor spreads
early to distant locations in the liver, adrenal
glands, bone marrow, and possibly brain. It is fre-
quently associated with superior vena cava
obstruction and paraneoplastic syndrome.
On imaging, the tumor is usually associated
with lung obstruction, atelectasis, and collapse of
Fig. 8.7 Squamous cell carcinoma with clear cell pattern. lung segments. Early spread to regional lymph
There is an island of squamous differentiation in the nodes could manifest as hilar or mediastinal
middle masses. Since these tumors are metabolically
active, they are strongly positive on FDG-PET
clear cell change and the name clear cell variant scanning.
is used (Fig. 8.7). When the cells still get smaller Grossly the tumors are white-tan, soft, and
but with distinct borders, prominent nucleoli, and friable. Extensive areas of necrosis could be noted
intercellular bridges, small cell variant is rendered within the tumor. A minority of tumors (about
in the diagnosis. This needs to be distinguished 5 %) could present as peripheral coin lesions.
from small cell carcinoma or combined small cell Microscopically the tumor presents as sheetlike
carcinoma and squamous cell carcinoma based growth with small nuclei and very scant amounts
96 M.M. Fraig

Fig. 8.8 Small cell carcinoma with extensive necrosis Fig. 8.9 Small cell carcinoma in a small biopsy lacking
and small blood vessels in the middle demonstrating the areas of necrosis but showing the same nuclear features
Azzopardi effect (smearing of blood vessels with released as in Fig. 8.8
DNA). The cells show remarkable nuclear molding and
very high N/C ratio
express one or more of the neuroendocrine
markers. Small cell carcinoma is also positive for
of cytoplasm around them. The chromatin is TTF-1 in up to 90 % of cases.
finely granular with absent or inconspicuous Small cell carcinoma should be differentiated
nucleoli. The mitotic activity is very high and from other neuroendocrine tumors as well as
extensive areas of necrosis could be seen. The small round blue cell tumors. The neuroendo-
smearing of loose DNA material around the walls crine category includes large cell neuroendocrine
of blood vessels is known as Azzopardi effect carcinoma, atypical carcinoid, and typical carci-
(Fig. 8.8). The combination of small cell carci- noid. In cases of carcinoid tumors, the mitotic
noma with other types of non-small cell carcino- activity is much lower (less than 10/2 mm2) with
mas could be encountered. For this diagnosis to lack of areas of necrosis and the presence of
be made, there should be at least 10 % of the organoid pattern. Large cell neuroendocrine car-
other components along with the small cell cinoma usually shows prominent nucleoli and
tumor. On small biopsy, the areas of necrosis and more abundant cytoplasm than that of small cell
marked mitotic activity might not be represented. carcinoma; otherwise the areas of necrosis and
However the cytologic features are very charac- the immunohistochemical profile would be simi-
teristic in that there is nuclear molding and high lar [10]. Small round blue cell tumors such as
N/C ratio and lack of nucleoli in spite of the high primitive neuroectodermal tumors (PNET) are
grade of the tumor (Fig. 8.9). mitotically active than small cell carcinomas, and
By immunohistochemistry, small cell carci- they mark with CD99 and not for cytokeratin or
noma is positive for neuroendocrine markers TTF-1. Merkel cell carcinoma when it is meta-
such as CD56, chromogranin A, and synaptophy- static to the lung can be difficult to distinguish
sin in a majority of cases. Less than 10 % of all from small cell carcinoma on morphology alone.
small cell carcinomas are negative for all neuro- Positivity for CK20 and lack of TTF-1 positivity
endocrine markers. This possibility makes the are helpful in distinguishing these two tumors
diagnosis a morphologic one [9]. On the other from each other.
hand, other non-small cell carcinomas such as On the molecular level, small cell carcinoma
adenocarcinoma and large cell carcinoma could is usually associated with a higher rate of p53
8 Malignant Tumors of the Lung in Small Lung Biopsies 97

mutation than other non-small cell carcinomas, Specific subtype of large cell carcinoma is
as well as amplification of MYC and methylation large cell neuroendocrine carcinoma which is
of caspase-8, a key antiapoptotic gene. characterized by cells growing in organoid nest-
ing, trabecular or rosette-like and palisading pat-
terns. The cells have an amphophilic cytoplasm
Large Cell Carcinoma and the nuclei have prominent nucleoli as opposed
to small cell carcinoma. Areas of tumor necrosis
This is an undifferentiated carcinoma that lacks and high mitotic count are also characteristic fea-
either squamous or glandular differentiation on tures of this tumor. The tumor cells react posi-
light microscopic evaluation. It has been used as tively to neuroendocrine markers such as
a diagnosis by exclusion or a wastebasket group. chromogranin A, synaptophysin, and CD56.
In the era of targeted chemotherapy, this group of
carcinoma is expected to decrease significantly in
number as more testing is being performed to Carcinoid Tumors
classify this group to either a squamous or adeno-
carcinoma category [11]. Carcinoid tumors are neuroendocrine tumors of
These tumors usually present anywhere in the low malignant potential. They arise around air-
lung and share their consistency and color with ways as small tumorlets which are nests of neuro-
other lung cancers. endocrine cells measuring less than 5.0 mm in
Microscopically, the cells are large (larger than diameter. Those are usually discovered in resection
two resting lymphocytes) and they grow in sheets specimens performed for other reasons.
with no specific configuration to suggest either Carcinoid tumors are those measuring more
squamous or glandular differentiation. The nuclei than 5 mm in diameter. They are usually well cir-
are large and vesicular with prominent nucleoli. cumscribed in close proximity to major airways.
Mitotic activity is usually high and areas of tumor They may bulge into the bronchial lumen causing
necrosis could be seen (Fig. 8.10). partial obstruction of those airways. They remain
under an intact smooth mucosa and less likely to
produce erosion or necrosis. In these locations,
they are amenable to biopsy and a forceps-
assisted biopsy can provide a generous biopsy for
morphologic evaluation and IHC.
Under the microscope, they have polygonal
cells with fine granular or amphophilic cyto-
plasm. The nuclei are rounded with absent or
inconspicuous nucleoli (Fig. 8.11). The mitotic
activity is less than 2/10 HPF. There is usually an
organoid pattern of some sort. The cells can form
nests, trabecula, or festoons with peripheral pali-
sading of the cells. On cytologic preparations,
the cells are discohesive and many of them
would come as single cells, while others may
show rosettes and pseudoglandular pattern. The
background is usually clean with no evidence of
Fig. 8.10 Large cell carcinoma is by definition a poorly dif- necrosis or apoptosis. Peripheral carcinoid
ferentiated carcinoma with no morphologic evidence of either tumors are notorious for showing a spindle cell
squamous or glandular differentiation; the cells have larger
nuclei and lower N/C ratio that in small cell carcinoma.
pattern that could be confused with mesenchy-
Attempts should be made to categorize these tumors as either mal tumors as well as sarcomatoid carcinoma.
adenocarcinoma or squamous cell carcinoma using IHC The nesting pattern and the granular type of
98 M.M. Fraig

Fig. 8.11 Carcinoid tumor can assume several patterns. Fig. 8.13 Large cell neuroendocrine carcinoma is show-
In this example, the cells show cytoplasmic clearing but ing a slight nuclear molding but more abundant cytoplasm
maintain their uniformity and nesting or “organoid” pattern, and prominent nucleoli than seen in small cell carcinoma.
when try to form a specific pattern like nests, trabecula, or Frequent mitotic figure and areas of necrosis are usually
even pseudoglandular patterns present

docrine markers such as synaptophysin, chromo-


granin A, CD 56, NSE, and leu 7 (CD57).
The strong diffuse positivity for chromogranin
A is in contrast to the weak or focal pattern in
small cell carcinoma. The proliferative index as
illustrated by the Ki67 labeling is low.
Atypical carcinoid is characterized by the
presence of single cell necrosis and partial loss of
the organoid pattern. The mitotic activity is
between 2 and 10/10 HPF. They present with
more aggressive behavior and higher potential
for metastasis. It is important to recognize the
difference between these entities on a small
biopsy and routinely perform Ki67 immunostain-
ing on all neuroendocrine tumors [12].
At the other end of the spectrum is large cell
Fig. 8.12 Spindle cell carcinoid with nesting and nuclei neuroendocrine carcinoma with extensive areas
having the characteristic “salt and pepper” pattern. This is of necrosis and large cells forming rosettes. The
different from smooth muscle tumors, which usually runs cells have more cytoplasm than is seen in small
in fascicles and continuous bundles
cell carcinoma and the nucleoli are more evident
and an amphophilic cytoplasm characteristic of
chromatin “salt and pepper pattern” are clues to neuroendocrine tumors [13] (Fig. 8.13). It is
the nature of the tumors (Fig. 8.12). important to remember that large cell carcinoma
IHC helps in elucidating the nature of these is biologically different and is treated differently
tumors. They are typically positive for neuroen- from small cell carcinoma, notwithstanding the
8 Malignant Tumors of the Lung in Small Lung Biopsies 99

overlap on morphologic and immunophenotypic


References
features. One of the important differential diag-
noses is basaloid squamous cell carcinoma, espe- 1. Travis WD, Brambilla E, Noguchi M, Nicholson AG,
cially on cytologic preparation and small biopsy. Geisinger KR, Yatabe Y, et al. International
Immunostains for squamous epithelial markers Association for the Study of Lung Cancer/American
such p40, p62, and CK5/6 should help in ruling Thoracic Society/European Respiratory Society inter-
national multidisciplinary classification of lung ade-
out this possibility. nocarcinoma. J Thorac Oncol. 2011;6(2):244–85.
2. Austin JH, Garg K, Aberle D, Yankelevitz D,
Kuriyama K, Lee HJ, et al. Radiologic implications of
the 2011 classification of adenocarcinoma of the lung.
Sarcomatoid Carcinoma Radiology. 2013;266(1):62–71.
3. Ichinokawa H, Ishii G, Nagai K, Kawase A, Yoshida
Sarcomatoid carcinoma of the lung is comprised J, Nishimura M, et al. Distinct clinicopathologic char-
of areas of clear epithelial differentiation along acteristics of lung mucinous adenocarcinoma with
KRAS mutation. Hum Pathol. 2013;44(12):2636–42.
squamous, adenocarcinoma, or merely poorly dif- 4. Saad RS, Cho P, Silverman JF, Liu Y. Usefulness of
ferentiated carcinoma admixed with either spin- Cdx2 in separating mucinous bronchioloalveolar ade-
dle cell carcinoma or giant cell carcinoma. Each nocarcinoma of the lung from metastatic mucinous
of the latter components should account for at colorectal adenocarcinoma. Am J Clin Pathol. 2004;
122(3):421–7.
least 10 % of the tumor, which is difficult to 5. Travis WD, Rekhtman N. Pathological diagnosis and
appreciate on a small biopsy. Immunostains for classification of lung cancer in small biopsies and
epithelial markers such as pancytokeratin or EMA cytology: strategic management of tissue for molecu-
are important to make the distinction from carci- lar testing. Semin Respir Crit Care Med. 2011;
32(1):22–31.
nosarcoma where the mesenchymal elements fail 6. Bishop JA, Sharma R, Illei PB. Napsin A and thyroid
to react with such markers. transcription factor-1 expression in carcinomas of the
lung, breast, pancreas, colon, kidney, thyroid, and
malignant mesothelioma. Hum Pathol. 2010;41(1):
20–5.
Metastatic Tumors 7. Bishop JA, Teruya-Feldstein J, Westra WH, Pelosi G,
Travis WD, Rekhtman N. p40 (DeltaNp63) is superior
As the lung is a vast vascular organ and a large to p63 for the diagnosis of pulmonary squamous cell
carcinoma. Mod Pathol. 2012;25(3):405–15.
pool of blood goes through the lung with every
8. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H,
heartbeat, it has an ability to trap circulating Gabriel S, et al. EGFR mutations in lung cancer: cor-
tumor cells and develop metastasis more fre- relation with clinical response to gefitinib therapy.
quently than other organs. The most common Science. 2004;304(5676):1497–500.
9. Zakowski MF. Pathology of small cell carcinoma of
tumors are those of melanoma, breast, and
the lung. Semin Oncol. 2003;30(1):3–8.
ovarian cancer. The colon, pancreas, liver, and 10. Jiang SX, Kameya T, Shoji M, Dobashi Y, Shinada J,
gynecologic organs are all capable of metasta- Yoshimura H. Large cell neuroendocrine carcinoma of
sizing early to the lung when they possess the the lung: a histologic and immunohistochemical study
of 22 cases. Am J Surg Pathol. 1998;22(5):526–37.
aggressive grade and critical volume to do so. 11. Sholl LM. Large-cell carcinoma of the lung: a diag-
Checking the history for any previous cancer nostic category redefined by immunohistochemistry
or any concurrent lesion helps in quickly sort- and genomics. Curr Opin Pulm Med. 2014;20(4):
ing out the question of metastatic vs. primary 324–31.
12. Travis WD, Rush W, Flieder DB, Falk R, Fleming MV,
lung tumors. The presence of multiple nodules Gal AA, et al. Survival analysis of 200 pulmonary neu-
in the lung would favor a metastatic tumor over roendocrine tumors with clarification of criteria for
a primary. Characteristic patterns like renal atypical carcinoid and its separation from typical carci-
cell carcinoma of the clear cell type or mela- noid. Am J Surg Pathol. 1998;22(8):934–44.
13. Watanabe R, Ito I, Kenmotsu H, Endo M, Yamamoto
noma with plasmacytoid nuclei and nuclear N, Ohde Y, et al. Large cell neuroendocrine carcinoma
pseudoinclusions should prompt investigation of the lung: is it possible to diagnose from biopsy
and performance of IHC. specimens? Jpn J Clin Oncol. 2013;43(3):294–304.
Index

A peribronchial fibrosis and chronic inflammation,


Acquired immunodeficiency syndrome (AIDS), 54, 72 23, 24
Acute fibrinous and organizing pneumonia (AFOP), 33 practical approach, 27
Acute interstitial pneumonia (AIP) pseudo-atelectasis, 24
clinical presentation, 45–46 pseudolipoid, 24–25
idiopathic form, DAD, 34 Aspergillosis
pathologic findings, 46 clinical and radiologic findings, 60
Acute lung injury pattern, 29–30 histologic findings, 60
Acute pneumonias, 29, 30 Aspiration pneumonia
Acute respiratory distress syndrome (ARDS), BOOP, 30
29, 30, 32, 60 clinical and radiologic findings, 73–74
Adenocarcinoma histologic findings, 74–76
atypical adenomatous hyperplasia (AAH), 94 Atelectasis, 15, 16, 94
abrupt transition, 92 Atypical adenomatous hyperplasia (AAH), 94
diagnosis, 93 Azzopardi effect, 96
EGFR, 94
electromagnetic navigational biopsy, 91
imaging, 91 B
immunohistochemistry, 94 Basaloid squamous cell carcinoma, 94, 95
invasive, 92 Benign tumors
lepidic pattern, 92 alveolar adenoma (AA), 84
mucinous, 93 clear cell tumor, 86–87
pleural surface/puckering, 91 endobronchial wall-related tumors, 81
types, 91 granular cell tumor, 82–83
Adenocarcinoma minimal invasion (AMI), 92 inflammatory myofibroblastic tumor, 87
AFOP. See Acute fibrinous and organizing pneumonia mucinous cystadenoma, 87–88
(AFOP) PA, 82
AIDS. See Acquired immunodeficiency syndrome papillary adenoma, 84
(AIDS) parenchymatous tumors, 83
Airspace disease, 14 pneumocytoma “sclerosing hemangioma”, 84–86
Air trapping, 14–15 pulmonary hamartoma, 83–84
Alveolar adenoma (AA), 84 schwannoma, 82
AMI. See Adenocarcinoma minimal invasion (AMI) solitary fibrous tumor, 86
Architectural distortion, 15 squamous papilloma, 81–82
ARDS. See Acute respiratory distress syndrome Blastomycosis
(ARDS) clinical and radiologic findings, 60
Artifacts and nonspecific findings histologic findings, 60
bronchial cartilage, 26 BOOP. See Bronchiolitis obliterans/organizing
carcinoid tumorlets, 25 pneumonia (BOOP)
chemodectoma, 25, 26 Bronchial cartilage, 26
corpora amylacea, 25 Bronchiectasis, 15, 16
fresh blood, 24 Bronchiolitis obliterans/organizing pneumonia (BOOP)
intra-alveolar macrophages, 24 air spaces, 30, 31
megakaryocytes, 26 alveolar space, 31
minute meningothelial-like nodules, 25, 26 bronchioloalveolar carcinoma, 32, 93

M.M. Fraig (ed.), Diagnosis of Small Lung Biopsy: An Integrated Approach, 101
DOI 10.1007/978-1-4939-2575-9, © Springer Science+Business Media New York 2015
102 Index

Bronchiolitis obliterans/organizing pneumonia Cryptococcosis


(BOOP) (cont.) clinical and radiologic findings, 72
DAD, 31–32 clusters, 72
description, 30 differential diagnosis, 72–73
etiology, 31 fibroblasts, 72
lung transplant community, 32 fungal organisms, 72
Bronchioloalveolar carcinoma, 32 granulomatous rim and necrosis, 72, 74
Bronchopneumonia, 14, 62, 73, 74 histiocytes and giant cells, 72, 73
immunocompromised patients, 72
mucicarmine and Fontana–Masson staining, 72, 75
C Cryptogenic organizing pneumonia (COP),
Carcinoid tumorlets, 25 9, 30, 32, 40
Carcinoid tumors CT. See Computed tomography (CT)
atypical, 98
differential diagnoses, 99
large cell neuroendocrine carcinoma, 98 D
nesting/organoid pattern, 97, 98 DAD. See Diffuse alveolar damage (DAD)
neuroendocrine, 97, 98 Desquamative interstitial pneumonia (DIP)
spindle cell, 98 histopathologic findings, 44–45
Caseating granuloma, 56 peribronchial pattern, fibrosis, 43
Caseous necrosis, 53 post-obstructive pneumonia, 45
Cavitation, 18 RB, 43
Centrilobular nodules, 16 smoking history, 43
Chemodectoma, 25, 26 smoking-related diseases, 45
Chest radiograph, 13 Diffuse alveolar damage (DAD)
Chronic beryllium disease (berylliosis) and AIP, 30, 45–46
clinical and radiologic findings, 71 and ARDS, 30, 32
histologic findings, 72 histopathologic features, 32–34
Churg–Strauss syndrome interstitial pattern, 31–32
clinical and radiologic findings, 62 DIP. See Desquamative interstitial pneumonia (DIP)
histologic findings, 62
Clear cell tumor, 86–87
Coccidioidomycosis E
differential diagnosis, 59–60 EBUS. See Endobronchial ultrasound (EBUS)
fungal infection, 58 EGFR. See Epithelial growth factor receptor (EGFR)
necrotizing granulomas, 59 Emphysema, 16
needle biopsy, 59 Endobronchial ultrasound (EBUS), 6, 7, 22, 64
spherules, 59 Endobronchial wall-related tumors, 81
Computed tomography (CT) Endogenous pneumoconiosis, 35
airspace disease, 14 Epithelial growth factor receptor (EGFR), 23, 94, 95
branching endobronchial nodules, 17
“crazy-paving” pattern, 35
cystic fibrosis, 15 F
description, 13 Fiberoptic bronchoscopy (FOB), 1–2
18F-FDG PET/CT, 14 Fibroblastic plugs, 30
ground-glass opacification, 9 Fine needle aspiration biopsy, 22
high-resolution, 13, 40, 41 Flexible fiberoptic bronchoscopy, 1–2
honeycomb changes, 41 Flow cytometry, 23
needle aspiration and core biopsy, 92 FOB. See Fiberoptic bronchoscopy (FOB)
smooth interlobular, 16 Focal and diffuse infiltrative diseases
solitary lung nodule, 56 COP, 9
subpleural/perilymphatic nodules, 17 cryobiopsy, 7–8
TSCT, 14 endobronchial cryotherapy, 7
Consolidation, 16 extrinsic allergic alveolitis, 7
COP. See Cryptogenic organizing pneumonia (COP) granulomatous lung diseases, 6
Core biopsy, 1, 22, 84, 92 hpersensitivity pneumonitis (HP), 7
Corpora amylacea, 25 HRCT, 8
Crazy-paving pattern, 16 IPF, 8
Cryobiopsy, 7–8, 22–23, 41, 48 NSIP, 8
Index 103

organizing pneumonia, 9 Hamman–Rich disease, 30, 34


pulmonary function testing, 8–9 High-resolution computed tomography (HRCT), 8
routine transbronchial lung biopsy, 6 Histoplasmosis
sarcoidosis, 7 caseating granuloma, 56
smoking-related inflmatory reaction, 8–9 characterization, 56
surgical lung biopsy, 6–7 chest CT and core needle biopsy, 55, 56
transbronchial lung biopsy, 6, 7 disseminated, 58
VATS, 8 endemic fungal infection, 54
Focal solid parenchymal lesions histologic features, 57
lung masses, 5 Histoplasma organisms, 57
lymphangitic metastases, 6 solitary lung nodule, 55
metastatic malignancies, 6 surgical pathologists, 55
solitary pulmonary nodules, 6 transbronchial/needle biopsy, 56
Fungal infections Honeycombing, 16
blastomycosis, 60 Horner’s syndrome, 94
coccidioidomycosis, 58–60 Hot tub lung
granulomatous mycobacterial, 53 clinical and radiologic findings, 68
histoplasmosis, 54–58 histologic findings, 68–69
mycobacterial, 51 HRCT. See High-resolution computed tomography
(HRCT)
Hypersensitivity pneumonitis (HP)
G cholesterol crystals/clefts, 67
Goodpasture syndrome clinical and radiologic findings, 66
clinical and radiologic findings, 75 clinical presentation, 47
histologic findings, 75–76 diffuse interstitial chronic inflammation,
Granular cell tumor 66–67
cytoplasm, 83 extrinsic allergic alveolitis, 46
endobronchial wall, 83 foamy macrophages, 67
microscopic examination, 82–83 granulomatous inflammation, 66
pedunculated masses, 82 histologic triad, 67–68
Granulomas. See Pulmonary granulomas histopathologic findings, 47, 48
Granulomatosis with polyangiitis (GPA). See Wegener’s lymphocyte, 66
granulomatosis lymphoid interstitial pneumonia, 47–48
Granulomatous inflammation transbronchial lung biopsy, 67, 68
aspergillosis, 60
aspiration pneumonia, 73–75
blastomycosis, 60 I
Churg–Strauss syndrome, 62 Idiopathic pulmonary fibrosis (IPF), 8
coccidioidomycosis, 58–60 Immunohistochemical staining, 23
cryptococcosis, 72–73 Inflammatory myofibroblastic tumor
diagnostic approach, 51 anaplastic lymphoma, 87
histoplasmosis, 54–58 characterization, 87
necrotizing granulomas, 63 neoplastic process, 87
non-tubercular mycobacterial infection, 54 paraneoplastic syndrome, 87
pathologic finding, 51 small biopsies, 87
pulmonary granulomas, 63 spindle cell neoplasms, 87
rheumatoid nodule, 62–63 Interlobular septal thickening, 16–17
tuberculosis, 52–53 Interstitial lung diseases
Wegener’s granulomatosis, 60–62 AIP, 45–46
Granulomatous lung diseases autoimmune, 8
HP, 7 classification, 39, 40
sarcoidosis, 7 DIP, 39, 43–45
Ground-glass nodule, 18 HP, 46–48
Ground-glass opacity, 16 NSIP, 42–43
RB-ILD, 43–45
UIP (see Usual interstitial pneumonia (UIP))
H Intra-alveolar hemorrhage, 34–35
Halo sign, 18 Intra-alveolar macrophages, 24
Hamartoma. See Pulmonary hamartoma IPF. See Idiopathic pulmonary fibrosis (IPF)
104 Index

L idiopathic form, 8
Large cell carcinoma, 97 and UIP, 47
Lipoid pneumonia, 24–25 uniform fibrosis, 42, 43
Lung biopsy Non-tubercular mycobacterial infection
endobronchial diseases diagnose, 2–4 AIDS, 54
FOB, 1–2 histologic findings, 54, 55
nonsurgical approaches, 1, 9 multifocal bronchiectasis, 54
parenchymal diseases diagnosis, 4–9 NSIP. See Nonspecific interstitial pneumonia (NSIP)
Lymphoid interstitial pneumonia (LIP), 47–48

P
M PA. See Pleomorphic adenoma (PA)
Malignant tumors Pancoast tumor, 94
adenocarcinoma, 91–94 PAP. See Pulmonary alveolar proteinosis (PAP)
carcinoid, 97–99 Papillary adenoma
histopathologic diagnosis, 91 fibrovascular cores, 84, 85
large cell carcinoma, 97 immunohistochemical stains, 84
metastatic, 99 microscopic examination, 84
sarcomatoid carcinoma, 99 Papilloma. See Squamous papilloma
small cell carcinoma, 95–97 Parenchymal diseases
squamous cell carcinoma, 94–95 categorization, 4, 5
Masson’s bodies, 31 focal and diffuse infiltrative diseases, 6–9
Mediastinum focal solid parenchymal lesions, 5–6
anterior, 19 Parenchymatous tumors, 83
middle, 19 Part-solid nodule, 18
posterior, 19 Peribronchovascular nodule, 17
pulmonary interstitium, 19 Perilymphatic nodule, 17
Megakaryocytes, 26 PFTs. See Pulmonary function tests (PFTs)
Metastatic tumors, 99 Pleomorphic adenoma (PA)
Microbiology studies, 22–23 benign mixed tumor, 82
Miliary pattern, 17 chondroid matrix, 82
Minute meningothelial-like nodules, differential diagnosis, 82
25, 26 histopathologic examination, 82
Molecular testing, 23 salivary gland tumors, 82
Mosaic attenuation, 17 Pleura, 19–20, 41, 86, 91
Mucinous adenocarcinoma, 93 Pneumoconiosis, 37, 39
Mucinous cystadenoma, 87–88 Pneumocytoma
Mycobacterial. See Non-tubercular mycobacterial blood lakes and hemosiderin, 84
infection clusters, 85
frozen section, 85
polygonal/cuboidal, 85
N stroma/interstitium, 85
Navigational bronchoscopy, 2, 6 types, cells, 85, 86
Needle core biopsies, 22 Practical approach, 27
Neurilemmoma, 82 Pseudo atelectasis, 24
Non-necrotizing granulomatous inflammation Pseudolipoid artifact, 24–25
chronic beryllium disease (berylliosis), Pulmonary alveolar proteinosis (PAP), 35–36
71–72 Pulmonary edema, 25, 32, 37
hot tub lung, 68–69 Pulmonary function tests (PFTs), 39, 42–44
hypersensitivity pneumonitis, 66–68 Pulmonary granulomas, 54, 57, 63
sarcoidosis, 63–66 Pulmonary hamartoma
talc granulomatosis, 69–71 benign neoplasm, 83
Nonspecific interstitial pneumonia (NSIP) cartilage and intervening fat, 83
cellular, mixed and fibrotic, 42 small biopsies, 84
celular interstitial chronic inflammation, 43 soap bubble/popcorn, 83
clinical and radiological findings, 42 Pulmonary interstitium. See also Pulmonary
clinical course, 43 parenchyma
ground-glass opacification, 8 mediastinum, 19
histopathologic features, 42–43 structure, 18
Index 105

Pulmonary nodule Schwannoma


bronchoscopy, 6 cellular loose and myxoid areas, 82
cavitation, 18 immunohistochemical stains, 82
definition, 17–18 neurilemmoma, 82
ground-glass nodule, 18 Sclerosing hemangioma, 84–86
halo sign, 18 Secondary pulmonary lobule, 16
part-solid nodule, 18 Small cell carcinoma
reversed halo sign, 18 Azzopardi effect, 96
solid nodule, 18 characterization, 95
Pulmonary parenchyma chromatin, 96
airspace disease, 14 cytologic features, 96
air trapping, 14–15 extensive areas, necrosis, 95
architectural distortion, 15 imaging, 95
atelectasis, 15 immunohistochemistry, 96
bronchiectasis, 15, 16 neuroendocrine tumors, 96
centrilobular nodules, 16 Solid nodule, 6, 18
consolidation, 16 Solitary fibrous tumor
crazy-paving pattern, 16 fibroblastic proliferation, 86
emphysema, 16 immunostains, 86
ground-glass opacity, 16 lung parenchyma, 86
honeycombing, 16 mesothelioma, 86
infiltrate, 16 vascular tumor markers, 86
interlobular septal thickening, 16–17 Spindle cell carcinoid, 98
miliary pattern, 17 Squamous cell carcinoma
mosaic attenuation, 17 basaloid, 94, 95
opacity, 17 characterization, 94
peribronchovascular nodule, 17 clear cell pattern, 95
perilymphatic nodule, 17 imaging, 94
traction bronchiectasis, 16 immunohistochemistry, 95
tree-in-bud opacities, 17 keratin formation, 94
Pulmonary vasculitis metaplasia and dysplasia, 94
Goodpasture syndrome, 75–76 pancoast tumor, 94
Wegener’s granulomatosis, 76 small cell variant, 95
Squamous papilloma
characteristics, 81
R cytologic atypia, 81, 82
Respiratory bronchiolitis-interstitial lung disease dysplasia, 81, 82
(RB-ILD) microscopic examination, 81
clinical presentation, 44 Sugar tumor, 86–87
description, 43
histopathologic findings, 44–45
Reversed halo sign, 18 T
Rheumatoid nodule Talc granulomatosis
clinical and radiologic findings, 62 clinical and radiologic findings, 69–70
histologic findings, 62–63 crospovidone, 71
foreign material, 70, 71
microcrystalline cellulose stains, 71
S movat pentachrome stain, 71
Sarcoidosis transbronchial lung biopsy, 70
clinical and radiologic findings, 63–64 Thin section CT (TSCT), 14–16, 18
conservative approach, 66 Traction bronchiectasis, 16
EBUS, 64 Transbronchial biopsy, 5–7, 21–23, 41
granulomas, 65 Tree-in-bud opacities, 17
histologic features, 65 TSCT. See Thinsection CT (TSCT)
histologic hallmark, 64 Tuberculosis
microorganisms, 65 acid-fast stains, 53
transbronchial lung biopsy, 64, 65 caseating, 52
Sarcomatoid carcinoma, 99 clinical and radiologic findings, 52
106 Index

Tuberculosis (cont.) V
mycobacteria, 53 Video-assisted thoracoscopy (VATS), 8
screening, 53
transbronchial lung biopsy, 53
W
Wedge biopsy, 21, 23, 25
U Wegener’s granulomatosis
Usual interstitial pneumonia (UIP) capillaritis, 62
accelerated phase, 34 clinical and radiologic findings, 60–61
clinical course, 41–42 multinucleated giant cells, 62
ground-glass opacities, 40 polyangiitis, 61
histopathologic findings, 40–41 transbronchial lung biopsies, 62
HRCT findings, 8
PFTs, 39

You might also like