You are on page 1of 19

Review Articles

Pathology of Granulomatous Pulmonary Diseases


Yale Rosen, MD

 Context.—Because granulomas are represented in almost as tuberculous and nontuberculous Mycobacterial infec-
every disease category, the number of clinically and tions, fungal, bacterial, and parasitic infections, sarcoido-
pathologically important granulomatous pulmonary dis- sis, necrotizing sarcoid granulomatosis, berylliosis,
eases is large. Their diagnosis by pathologists is particularly hypersensitivity pneumonitis, hot tub lung, rheumatoid
challenging because of their nonspecificity. A specific nodule, bronchocentric granulomatosis, aspirated, inhaled,
diagnosis can be achieved only when a granuloma-inciting and embolized foreign bodies, drug-induced granulomas,
agent(s) (eg, acid-fast bacilli, fungi, foreign bodies, etc) are chronic granulomatous disease, common variable immu-
identified microscopically or by culture; this does not nodeficiency, and granulomatous lesions associated with
occur in most cases. Furthermore, a specific diagnosis various types of cancer.
cannot be reached in a high percentage of cases. Although Data Sources.—Review of pertinent medical literature
sarcoidosis and infectious diseases account for approxi- using the PubMed search engine and the author’s practical
mately half of pulmonary granulomatous diseases world- experience.
wide, there is significant geographic variation in their Conclusions.—Although the diagnosis of granulomatous
prevalence. lung diseases continues to present significant challenges to
Objectives.—To present updated information to serve as pathologists, the information presented in this review can
a guide to pathologic diagnosis of pulmonary granuloma- be helpful in overcoming them. The importance of
tous diseases, to address some commonly held misconcep- multidisciplinary coordination in cases where morphologic
tions and to stress the importance of multidisciplinary diagnosis is not possible cannot be overstated.
coordination. Presentation of basic aspects of granulomas (Arch Pathol Lab Med. 2022;146:233–251; doi: 10.5858/
is followed by discussion of specific disease entities, such arpa.2020-0543-RA)

T he assessment of granulomatous lung lesions seen in


surgical and autopsy specimens is often challenging.
Many excellent reviews devoted to or addressing differential
ated lesions may provide diagnostic clues. Correlation of the
pathologic findings with the patient’s medical history,
clinical, radiologic, and laboratory findings is often necessary
diagnosis of pulmonary granulomatous diseases have been to reach a diagnosis. The importance of a multidisciplinary
published.1–7 This review presents up-to-date information approach to reaching the diagnosis of granulomatous lesions
on granulomatous lung diseases and is intended to serve as in which an inciting agent cannot be identified is empha-
a practical and useful guide to diagnosis. Initial discussion of sized.
some basic features of granulomas will be followed by
discussion of individual pulmonary granulomatous diseases. OVERVIEW OF PULMONARY GRANULOMATOUS
Differential diagnosis, issues that are not widely appreciat- DISEASES
ed, and some commonly held misconceptions will be There is significant worldwide variation in the prevalence
addressed. of pulmonary granulomatous diseases. Mycobacteria are the
Identification of the etiology or disease association of most common cause of pulmonary granulomas worldwide.5
granulomatous lesions is often difficult because granulomas A retrospective review of 500 lung biopsies and resections
are nonspecific lesions whose appearance does not reveal from 10 institutions in 7 countries5 demonstrated that,
their etiology or disease association unless the inciting agent worldwide, sarcoidosis and infection accounted for 136 of
can be identified within the granuloma or by culture. 500 (27%) (range, 12%–36%) and 125 of 500 (25%) (range,
4%–48%) of cases, respectively. Mycobacterial infection was
Although the inciting agent is not identified in most cases,
diagnosed in 56 of 300 cases (19%) outside of the US versus
the morphologic features of some granulomas and associ-
16 of 200 (8%) in the US. Fungal infection was diagnosed in
38 of 200 (19%) of the US cases versus 13 of 300 (4%) in
Accepted for publication January 28, 2021. other locations. Pulmonary granulomas in immunosup-
Published online April 27, 2021. pressed individuals are almost invariably associated with
From the Department of Pathology, SUNY Downstate Health infection. Distinction between infection and noninfectious
Sciences University, Brooklyn, New York. etiologies is often the most important role of the pathologist
The author has no relevant financial interest in the products or
and has the greatest impact on clinical management in most
companies described in this article.
Corresponding author: Yale Rosen, MD, Department of Pathology, cases. A major problem encountered in the evaluation of
SUNY Downstate Health Sciences University, 450 Clarkson Ave, granulomas is that a definitive diagnosis cannot be arrived at
Brooklyn, NY 11203 (email: yrosen854@gmail.com). in a significant percentage of cases1,4,5,7,8 (Table 1).
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 233
Table 1. Granulomatous Lung Diseases Without Definitive Pathologic Diagnosis
Author(s) Cases, n Cases, % Comment
1
Ulbright and Katzenstein 22/86 26 Solitary necrotizing granulomas
Hutton Klein et al4 27/100 27 100 consecutive granulomatous cases in a pulmonary pathology
consultation practice. A specific diagnosis was favored in 34/100 (34%)
and a differential diagnosis was provided in 39/100 (39%)
Mukhopadhyay et al5 210/500 42 Granulomas of all types
Mukhopadhyay et al7 131/566 23% initially; Necrotizing granulomas. Subsequent comprehensive analysis of the 131
9% after cases following review of medical records, laboratory data including
comprehensive cultures, and radiographic data disclosed the cause of granulomas in an
review additional 79/131 cases (60%), resulting in 52 of the initial 566 cases
(9%) remaining undiagnosed
Woodard et al8 10/59 17 Granulomas of all types

DEFINITION, DEVELOPMENT, AND MORPHOLOGY OF type (peripheral nuclei) and foreign body type (central
GRANULOMAS nuclei) has no diagnostic significance and both types may be
A granuloma may be defined as ‘‘a compact (organized) present in the same specimen. Giant cells exhibit consid-
collection of mature mononuclear phagocytes (macrophages erable variation in the location of their nuclei and frequently
and/or epithelioid cells) which may or may not be do not meet the classical criteria for either Langhans or
accompanied by accessory features such as necrosis or the foreign body types.
infiltration of inflammatory leukocytes.’’9 Although necrosis Whereas epithelioid granulomas are initiated and regu-
and multinucleate giant cells are often present in granulo- lated by the immune system and are a manifestation of Type
mas, their absence does not disqualify a lesion from being IV delayed hypersensitivity, the pathogenesis of reactions to
called a granuloma. In some lesions that are recognizable as particulate foreign materials (ie, foreign body granulomas)
granulomas, the aggregates of epithelioid cells are not very probably does not involve the immune system in most
compact but are loosely arranged. These are referred to as cases. In contrast to epithelioid granulomas, foreign body
‘‘poorly formed’’ or ‘‘loose’’ granulomas and are character- granulomas consist mostly of giant cells and macrophages
istically seen in hypersensitivity pneumonitis (HP), granu- in varying proportions
lomatosis with polyangiitis (GPA), foreign body reactions, Necrosis in granulomas may occur when the granuloma-
lymphocytic interstitial pneumonia, and others. inciting agent is highly toxic to the macrophages and/or
Initial presentation of persistent and poorly degradable when a vigorous delayed hypersensitivity response is
antigens by dendritic cells to T4þ lymphocytes cells results evoked. The term ‘‘caseous necrosis,’’ which refers to the
in their transformation to Th1 lymphocytes. This is followed grossly visible, cheese-like appearance, but not to the
by complex interactions between lymphokines produced by microscopic appearance, of a necrotic lesion, has proven
the Th1 lymphocytes and cytokines produced by cells of the to be a source of substantial confusion for both pathologists
mononuclear phagocytic system that result in maturation of
and clinicians. Medlar12 stated that:
monocytes to mature macrophages, which undergo trans-
formation to epithelioid cells resulting in the formation of The term caseation is applied to the gross character-
epithelioid granulomas. Details of current concepts of the istics of the necrotic material commonly seen in
immunopathogenesis of epithelioid granulomas in sarcoid- tuberculous infection. This necrotic tissue can easily be
osis are well presented by Beijer et al.10 Compared with removed from the surrounding living tissue, leaving a
macrophages, epithelioid cells have increased secretory and cavity. It has no texture and varies in consistence from
bactericidal capability and reduced phagocytic capability. thick pus to a crumbly material not unlike cottage
Epithelioid granulomas may be surrounded by a rim of T8þ cheese.
suppressor lymphocytes. Granulomas without this lympho-
cyte rim are referred to as ‘‘naked’’ granulomas. The Although the term ‘‘caseous necrosis’’ has usually been
lymphocytes admixed with epithelioid cells within the associated with tuberculosis (TB), this type of necrosis may
granulomas are mostly T4þ lymphocytes. A study using
be seen in numerous other conditions, including fungal
quantitative stereometry determined that in the central part
infections, pulmonary angiitis and granulomatosis, malig-
of sarcoid granulomas, lymphocytes outnumber epithelioid
cells by a ratio of 2:1 and, therefore, appear to be the nant neoplasms, parasitic infections, syphilis, tularemia,
predominating cell type11; this finding requires confirma- typhoid fever, and others. The ‘‘caseous’’ or cheese-like
tion. The function of granulomas as stated by Adams9 is they appearance results from incomplete digestion of necrotic
‘‘appear to be the host’s response to a high local tissue. The terms ‘‘caseous,’’ ‘‘caseating,’’ and so on apply
concentration of a foreign substance which was not only to the gross appearance of some necrotic lesions and
destroyed by the acute inflammatory response and which have no microscopic counterpart. Therefore, stating that a
is being contained and destroyed by mononuclear phago- lesion is a ‘‘caseating granuloma’’ based on its microscopic
cytes in various stages of maturation or activation.’’ appearance is inappropriate. Since the modifiers ‘‘caseous,’’
Multinucleate giant cells, derived from fusion of mono- ‘‘caseating,’’ and so on have no diagnostic relevance and
cytes, macrophages, and epithelioid cells, rather than by may be misleading because of their association with
proliferation and nondivision, are present in most granulo- tuberculosis, it is recommended that they not be used in
mas and may assume a variety of appearances. The pathology reports to describe microscopic findings. The
traditional classification of giant cells as being of Langhans terms ‘‘necrotizing’’ and ‘‘nonnecrotizing’’ are preferable.
234 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
suppurative granulomas. Fibrin-ring and spindle-cell gran-
ulomas/pseudotumors are unusual types of nonnecrotizing
granulomas. Spindle-cell granulomas/pseudotumors, usual-
ly caused by Mycobacterium avium complex (MAC) infection
in immunocompromised patients, contain multitudes of
acid-fast bacilli (AFB). Disease associations of granuloma
subtypes are summarized in Table 2. Microscopic descrip-
tions of the granuloma subtypes are summarized in Table 3.
Nonnecrotizing granulomas usually have a noninfectious
etiology. However, some infectious diseases may also
present with nonnecrotizing granulomas. For example,
nonnecrotizing granulomas have been reported to be the
only biopsy finding in 14 of 42 cases (33%) of TB.8 The most
common pulmonary diseases typically presenting with
nonnecrotizing granulomas are sarcoidosis, hypersensitivity
pneumonitis, chronic beryllium disease, and foreign body
granulomas.
Necrotizing granulomas usually have an infectious etiol-
ogy, most caused by Mycobacteria and fungi. Exceptions
include rheumatoid nodules, GPA, and the necrotizing
sarcoid granulomatosis (NSG) variant of sarcoidosis. Pali-
saded necrotizing granulomas in the lungs are almost
always rheumatoid nodules but can also be caused by a
variety of infections.13

INCLUSIONS IN GRANULOMAS
A variety of inclusions may be present within the giant
cells of granulomas. They are nonspecific and are not
diagnostic of any disease entity.
Schaumann Bodies
Figure 1. A, Nonnecrotizing epithelioid granuloma in sarcoidosis. B, Schaumann (conchoidal) bodies, are large (25–200 lm),
Necrotizing epithelioid granuloma in tuberculosis (hematoxylin-eosin, concentrically lamellated, calcific bodies that are present
original magnification 3200 [A and B]). predominantly within giant cells in the granulomas of up to
50% of cases of sarcoidosis but also occur, and may be
Granulomas that exhibit minimal, focal necrosis will be prominent, in hypersensitivity pneumonitis, berylliosis, and
other granulomatous diseases14 (Figure 2, A). They are
discussed in the section on sarcoidosis.
composed of a mucopolysaccharide matrix impregnated
Epithelioid granulomas may be nonnecrotizing (Figure 1, with calcium salts and some iron. As the bodies enlarge, cell
A) or necrotizing (Figure 1, B); some may exhibit minimal, rupture may occur resulting in their extrusion into the
focal necrosis. Foreign body granulomas are almost always extracellular space. A crystalline component that is colorless,
nonnecrotizing. Various subtypes of necrotizing granulomas intensely birefringent, and spiculated may be seen frequent-
include palisading, suppurative, necrobiotic, and stellate, ly, either alone, or in combination with the Schaumann

Table 2. Granuloma Subtypes and Their Disease Associations


Granuloma Subtype Typical Disease Associations
Nonnecrotizing Sarcoidosis, berylliosis, HP, hot tub lung, CVID, drug reactions, foreign bodies, inhaled metals,
schistosomiasis (ova), paragonimiasis (ova)
Necrotizing TB, NTM, fungal infections, NSG, BG, CGD
Minimal, focal necrosis Sarcoidosis
Suppurative Fungal infections, GPA, CGD, aspiration, EGPA, CVID
Suppurative, stellate Cat scratch disease, lymphogranuloma venereum, tularemia, Yersinia
Palisading Rheumatoid nodule, CGD, GPA, EGPA, BG, infections, including phaeohyphomycosis, NTM,
cat scratch disease, sporotrichosis, cryptococcosis, and coccidioidomycosis
Necrobiotic Most seen postbiopsy of urinary bladder, prostate, and cervix. Associated with electrocautery
Fibrin ring Q fever, EBV, visceral leishmaniasis, toxoplasmosis, Hodgkin lymphoma, typhus, allopurinol
hypersensitivity, CMV, immune checkpoint inhibitors, and others. Seen mostly in liver and
bone marrow. Lung involvement not reported
Spindle cell granuloma/pseudotumor MAC infection, TB, and histoplasmosis (rare). Usually occurs in immunosuppressed persons
Abbreviations: BG, bronchocentric granulomatosis; CGD, chronic granulomatous disease; CMV, cytomegalovirus; CVID, common variable
immunodeficiency; EBV, Epstein-Barr virus; EGPA, eosinophilic granulomatosis with polyangiitis; GPA, granulomatosis with polyangiitis; HP,
hypersensitivity pneumonitis; MAC, Mycobacterium avium complex; NSG, necrotizing sarcoid granulomatosis variant of sarcoidosis; NTM,
nontuberculous mycobacteria; TB, tuberculosis.

Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 235
Table 3. Microscopic Descriptions of Granuloma Subtypes
Granuloma Subtype Description
Nonnecrotizing Compact, organized collection of epithelioid cells or macrophages with or without giant cells.
Necrosis absent
Necrotizing Necrosis is a prominent feature
Minimal, focal necrosis Minimal foci of necrosis focally distributed; involving a small percentage of granulomas
Suppurative Granuloma with focal necrosis with purulent exudate
Suppurative, stellate Elongated, irregular, often stellate with central suppurative necrosis
Palisading Elongated and compressed histiocytes, aligned parallel to each other and surrounding a central
zone of necrosis
Necrobiotic Collagen necrosis surrounded by palisaded histiocytes. Resembles rheumatoid nodule
Fibrin ring Small, compact collection of macrophages surrounded by a thin ring of fibrin
Spindle cell granuloma/pseudotumor Spindled macrophages simulating a neoplasm. Reported to be CD68 positive

body (Figure 2, A). As many as 70% of Schaumann bodies other disorders.19,20 They are oval or spindle-shaped and
have crystals associated with them ranging in size from 1 to approximately the same diameter as a resting lymphocyte.
20 lm. They stain for iron and calcium salts; the majority are They appear yellow-brown in sections stained with hema-
calcium oxalate admixed with calcium carbonate.15 It is toxylin-eosin (H&E) (Figure 2, E). The yellow-brown
likely that the crystals act as a nidus for the formation of the pigment has the histochemical characteristics of lipofuscin.
Schaumann body. Jones Williams16 reported finding crys- They have been reported in 11% to 68% of sarcoid lymph
talline and conchoidal bodies in 88% of cases of sarcoidosis, nodes and in 11.5% to 15% of nongranulomatous lymph
62% of chronic beryllium disease, and 6% of tuberculosis nodes, which were either normal or involved with various
cases. diseases.14,21 When stained with the Grocott methenamine
silver stain they often exhibit an appearance that is similar to
Crystalline Bodies
yeast-like budding (Figure 2, F) and can easily be mistaken
Crystalline bodies occurring without Schaumann bodies for fungi (budding yeasts). Positive staining with the Ziehl-
have been reported in 41% of cases of sarcoidosis, 15% of Neelsen (ZN) stain, and absence of a host response readily
chronic beryllium disease, and 3% of tuberculosis16 (Figure facilitate their distinction from fungi. Ultrastructural exam-
2, B). Histochemical analysis of the crystalline inclusions ination shows that the Hamazaki-Wesenberg bodies are
indicates that the majority are composed of calcium oxalate giant, intracellular, and extracellular lysosomes and residual
and calcium carbonate.15 bodies.14
Asteroid Bodies ETIOLOGY AND DISEASE ASSOCIATIONS OF
Asteroid bodies are visually striking stellate inclusions that PULMONARY GRANULOMATOUS DISEASES
are found mostly within the giant cells of foreign body
Infectious Diseases
granulomas (Figure 2, C). They may also be present in
sarcoidosis and in a variety of other granulomatous diseases, Approximately 25% of pulmonary granulomatous lesions
including tuberculosis, leprosy, histoplasmosis, schistoso- are caused by an infectious agent; Mycobacteria and fungi are
miasis, and others.17 They vary from 5 to 30 lm in size and the cause of almost all. Cultures and polymerase chain
contain as many as 30 or more rays radiating from a central reaction (PCR) are most likely to detect mycobacterial
core. Although they may be seen in granuloma-containing infection, whereas the diagnosis of most pulmonary fungal
tissues in 2% to 9% of patients with sarcoidosis,14,17 there is infections is based on histology.5 Nevertheless, AFB and
a widely held misconception that the presence of asteroid fungal stains should be performed and carefully examined in
bodies supports or indicates the diagnosis of sarcoidosis. all cases where the causative agent is not identified on the
Ultrastructural features of asteroid bodies were reported by H&E-stained slides. Culture is the ‘‘gold standard’’ for
Cain and Kraus.18 identification of infectious agents in tissue specimens.
However, culture is, unfortunately, not performed in most
Cholesterol Inclusions cases.
Slit-like cholesterol inclusions may be seen within the
giant cells of granulomas of varied etiology (Figure 2, D). Mycobacterial Infections
They may be present in as many as 17% of cases of Tuberculosis.—A lung biopsy is usually not required to
sarcoidosis14 and occasionally in other granulomatous diagnose TB because the diagnosis can typically be made
conditions. Their pathogenesis is uncertain, and they have based on exposure history, symptoms, and radiologic
no diagnostic significance. findings, ideally supplemented by the finding of AFB in
sputum smears.22 If possible, the presence of Mycobacterium
HAMAZAKI-WESENBERG BODIES tuberculosis (MTb) should be documented by cultures or
Tiny, spindle-shaped bodies, called Hamazaki-Wesenberg PCR. Lung biopsies are usually performed in those
bodies (Figure 2, E), with rare exceptions, are located individuals presenting radiographically with nodules or
extracellularly and do not present as inclusions in giant cells. masses, which may raise concern about a possible neo-
They are frequently seen in the granulomatous lymph nodes plasm.
from patients with sarcoidosis and in nongranulomatous The usual biopsy finding in TB is necrotizing granulomas
lymph nodes from patients with sarcoidosis and a variety of (Figure 1, B). However, nonnecrotizing granulomas have
236 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
Figure 2. Sarcoidosis. A, Giant cell with Schaumann body. Inset (polarized): calcium oxalate crystals with thin rims of calcification; early
Schaumann body. B, Calcium oxalate and carbonate crystals within giant cells. C, Giant cell containing 2 asteroid bodies. D, Cholesterol granuloma.
E, Hamazaki-Wesenberg bodies (yellow-brown bodies) in a lymph node. Courtesy of Runjan Chetty, MB BCh. F, Grocott methenamine silver stain of
Hamazaki-Wesenberg bodies showing their resemblance to budding yeasts (hematoxylin-eosin, original magnifications 3400 [A, A inset, and E] and
3200 [B through D]); Grocott methenamine silver stain, original magnification 3400 [F]).

been reported to be the only biopsy finding in up to 33% of examination is very time-consuming and prone to error. Its
cases8 and may be the only subtype of granuloma identified, specificity for detecting Mycobacteria is high, approaching
particularly in small biopsies with few granulomas. Occa- 100%. In 20 culture-proven mycobacterial infections,
sionally, only necrotic tissue is present. Mycobacteria were identified histologically in only 4 using
The ZN, Kinyoun acid-fast stain, and fluorochrome ZN, Fite, and auramine-rhodamine (AR) stains; none of the
staining using auramine O with or without rhodamine are stains were found to be superior.6 AFB are most likely to be
routinely used to identify AFB in tissue; neither can detected in areas of necrosis and within giant cells. Jain et
distinguish MTb from nontuberculous Mycobacteria al22 have compiled a list of tips for identifying AFB in
(NTM). Disadvantages of the ZN stain are its low sensitivity necrotizing granulomas. These include staining at least 2
ranging from 8.3% to 60% in culture positive cases12,23,24 and blocks exhibiting necrosis, selecting the blocks with the
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 237
greatest amount of necrosis, and searching within areas of immunocompromised patients.32 NTM are the most com-
necrosis where the yield is greatest using a 340 or oil mon cause of spindle-cell granulomas, a rare type of
immersion objective. False-positive AFB stains can result granuloma containing innumerable AFB; they can also be
from contamination of water used to prepare the stains with caused by MTb.
NTM.25 Filtered or distilled water should be used to prepare Because NTM are ubiquitous in the environment,
these stains. especially in soil and water, they are a common contaminant
Auramine O or AR fluorescent stains are widely used for of sputum and, to a lesser extent, of bronchoalveolar lavage
identification of AFB. Sensitivity is generally reported to be specimens and bronchial washings even if tap water is not
greater than the ZN stain and time required for examination used. When NTM grow in culture of tissue the question
is shorter. However, whether they are more or less specific arises as to whether they represent true infection or
than ZN remains controversial. In some laboratories, the AR contamination. Because lung biopsy and resection speci-
stain is used for screening and ZN is subsequently mens are considered to be sterile, a positive culture of NTM
performed on AR positives. The requirement for a fluores- from these specimens is considered clinically significant.
cent microscope makes this method unavailable for many Identification of AFB in granulomas in the tissue from which
pathologists. Because of low sensitivity, a negative AFB stain a positive culture was obtained is a further indication of true
does not exclude mycobacterial infection. infection, because it shows that the microorganisms are in a
MTb-specific DNA fragments can be identified by PCR. granuloma.22 The combination of culture positivity for NTM
Several studies of the results of PCR performed on formalin- from lung tissue and the presence of granulomas in that
fixed, paraffin-embedded tissue obtained from MTb culture– tissue with or without AFB being demonstrated, is
positive patients reported sensitivity ranging from 58% to considered sufficient to establish the diagnosis of NTM
100% (median 89%) and specificity ranging from 92% to pulmonary disease.33,34 In cases where AFB are identified in
100% (median 97%).23,26–28 Thus, PCR appears to compare pulmonary granulomas and culture is negative, PCR can be
favorably with culture for the diagnosis of tuberculosis. It used to distinguish between MTb and NTM.
can also differentiate MTb from NTM. A major advantage of Fungal Infections.—Granulomas are present in infec-
PCR is the relatively short turnaround time compared with tions caused by many species of fungi, including Histoplas-
culture, which can take up to 12 weeks, and its ability to ma, Cryptococcus, Blastomyces, Coccidioides, Paracoccidioides,
distinguish between MTb and NTM. and others. Candida and Aspergillus usually cause acute
There are a small number of reports of diagnosis of TB in inflammation but are also capable of inciting granulo-
formalin-fixed, paraffin-embedded tissue using immuno- mas.35,36 Fungal granulomas are often suppurative. Many
histochemistry with an anti-MTP64 antibody.29,30 This fungi are easily seen with routine H&E staining, but fungal
technique does not appear to be in widespread use morphology is best appreciated with the Grocott methena-
currently. mine silver stain. Mucicarmine stains the mucinous capsule
Next-generation sequencing can provide whole bacterial of Cryptococcus and helps to distinguish it from Blastomyces
genome sequences in a short timeframe (hours-days), at a that is about the same size but has a relatively thick,
relatively low cost and, increasingly, without the need for refractile cell wall and exhibits broad based budding.
culture.31 Although next-generation sequencing can be used Nonencapsulated cryptococci are not stained by mucicar-
to identify MTb in culture isolates and clinical specimens, it mine. In the author’s experience, negative fungal stains are
cannot currently be used to identify MTb in formalin-fixed, rare in fungal infections. However, in granulomatous
paraffin-embedded tissue. Pneumocystis infection, the cyst forms of Pneumocystis jiroveci
Nontuberculous Mycobacterial Infection.—NTM are may be rare and difficult to identify; Grocott methenamine
an important cause of pulmonary and extrapulmonary silver–stained step sections may be needed. Although the
infections. The subject of NTM lung disease is well reviewed ‘‘gold standard’’ for fungal identification is culture, fungi
by Jain et al22 and El-Zammar and Katzenstein.2 NTM lung that present as yeast forms in tissue can usually be identified
disease is mostly caused by MAC, a group of Mycobacteria with a high degree of confidence based on morphologic
comprising M intracellulare and M avium that are grouped features, including size, presence, or absence of budding
together because they commonly infect humans simulta- and pseudohyphae, type of budding, presence of endo-
neously. Its radiologic features overlap with those of spores, and presence of a refractile cell wall. These include
pulmonary TB. The terms ‘‘atypical Mycobacteria’’ and Histoplasma, Candida, Cryptococcus, Blastomyces, Coccidioides,
‘‘Mycobacteria other than tuberculosis’’ have also been used Paracoccidioides, and others. On the other hand, the many
in the past for these organisms. Other NTMs, such as M types of fungi that present with hyphal forms have
kansasii, M fortuitum, M chelonae, M abscesus, M gordonae, M overlapping morphology and, with some exceptions, cannot
xenopi, and others can cause pulmonary disease, but much be accurately diagnosed based on morphology alone;
less frequently than MAC.22 In this review, the term NTM culture is required. Because the detailed morphologic and
encompasses all the NTM mentioned above, but especially diagnostic features of the numerous pathogenic fungi are
MAC because they are the most common. There is beyond the scope of this review, readers are referred to
significant geographic variation in types of NTM associated publications that provide detailed coverage of fungal
with pulmonary disease.2 MAC followed by M kansasii is morphology.3,37
most frequent in the US.2 In immunosuppressed patients,
NTM infection commonly presents microscopically as sheets Nonmycobacterial Bacterial Infections
of large, foamy histiocytes laden with innumerable AFB; A variety of nonmycobacterial bacteria may produce
granulomas are usually absent. NTM pulmonary infections granulomatous inflammation, which is usually necrotizing
typically occur in immunocompetent patients and are and often suppurative. These include Brucella spp, Actino-
characterized by necrotizing granulomas containing AFB myces spp, Francisella tularensis, Bartonella henselae, Nocardia
that are indistinguishable from MTb. Necrotizing granulo- spp Yersnia enterocolitica, Listeria monocytogenes, Burkholderia
mas have also been reported to occur with NTM infection in pseudomallei (melioidosis), Coxiella burnetti, and others2,38
238 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
find and step sections may be needed. They appear as large,
round-to-oval structures averaging 200 lm in diameter.
They have a thick (5–25 lm), multilayered cuticle that
contains transverse striations. In old lesions the worms may
calcify.2
Ova of Paragonimus spp in lung tissue and ova of
Schistosoma spp in pulmonary capillaries elicit a granulo-
matous reaction. The ova are usually easily seen.
Diseases of Unknown Etiology/Idiopathy
Sarcoidosis.—Sarcoidosis is a common disease of un-
known etiology with a worldwide distribution. It involves
the lungs and intrathoracic lymph nodes in almost all cases.
There is evidence of lung involvement even in cases that
present radiographically with hilar lymphadenopathy only.39
It is the most common granulomatous disease involving the
lungs in areas of the world where the incidence of TB is low.
The pathologic features of sarcoidosis have been the subject
of several comprehensive reviews.40–44 It appears to be
triggered by a variety of unidentified antigens that can
provoke granuloma formation in persons who are geneti-
cally predisposed to mount an exaggerated immune
response. Extrathoracic involvement occurs in a small
percentage of patients and may involve any organ system.
An extensive description of sarcoidosis was published in
1999,45 but there is not yet a precise definition of this
disease.
Pathologists almost always encounter the granulomas of
sarcoidosis in bronchial, transbronchial lung, and intratho-
racic lymph node biopsy specimens. They are occasionally
seen in needle core biopsies of nodular lung lesions.
Granulomas have been reported in endobronchial biopsies
Figure 3. Cross sections of 2 immature, adult Dirofilaria immitis
in 40% to 71% of patients.46–50 In bronchial biopsies that do
worms in necrotic blood vessels within infarcted lung tissue (hematox-
ylin-eosin, original magnification 3200). not exhibit well-formed granulomas, the only finding may
be scattered giant cells with or without Schaumann bodies
Figure 4. Lymphangitic localization of granulomas around broncho- or extracellular Schaumann bodies alone. Transbronchial
vascular bundles in sarcoidosis (hematoxylin-eosin, original magnifica- lung biopsy is most frequently used to demonstrate
tion 340). granulomas in those suspected to have sarcoidosis. Overall
sensitivity ranging from approximately 50% to more than
(Table 2). Although Gram and silver stains may be helpful in 90% has been reported.46–50 The yield of granulomas is
some cases, diagnosis is usually based on identification in highest in those patients who have radiographic evidence of
cultures. lung involvement and is directly proportional to the number
of biopsy specimens obtained. Biopsy of peripheral and
Parasitic Diseases intrathoracic lymph nodes may also be performed where
Pulmonary granulomas may be produced by Dirofilaria appropriate. In cases of suspected sarcoidosis in which
immitis (dog heartworm), and the ova of Schistosoma spp granulomas are not found using the above-mentioned
and Paragonimus westermanii. biopsy procedures, surgical lung biopsies or transbronchial
Dogs, as well as other mammals, the natural hosts, harbor cryobiopsies may be performed. Jacob et al51 reported a
the adult form of D immitis in their right ventricle where sensitivity of 74% for detecting granulomas in 27 patients
they produce microfilariae that are shed into the blood- with suspected sarcoidosis with transbronchial lung cryo-
stream. Mosquitos, the intermediate hosts, become infected biopsies. Wedge resections or even lobectomies may
with microfilariae when they bite the natural host. occasionally be performed in cases presenting with single
Microfilariae develop into L3 larvae in the mosquito. When or multiple nodules that are suspicious for neoplasm.
humans are bitten by an infected mosquito, the L3 larvae Nonspecific chronic interstitial inflammation representing
develop into immature adults in subcutaneous tissue and the ‘‘alveolitis’’ of sarcoidosis appears to be the precursor of
eventually migrate to the right ventricle where they die and granuloma formation in the lungs; it was reported as a
are swept into the pulmonary arteries. They then embolize predominant or prominent finding in two-thirds of open
to the lungs where they produce infarcts surrounded by lung biopsy specimens.52 It is rarely seen in small bronchial
granulomatous inflammation with or without eosinophilia. and transbronchial biopsy specimens.
These lesions most often appear as an incidental finding of a A very characteristic feature of sarcoidosis is the tendency
solitary, well-circumscribed lesion on radiologic studies in for the granulomas to localize around bronchovascular
an asymptomatic patient.2 The immature, dead worms are bundles and fibrous septa (Figure 4). This is referred to as
found within a necrotic blood vessel(s) located in the lymphangitic or perilymphatic localization because a lym-
necrotic tissue (Figure 3), but they are sometimes difficult to phatic network is associated with these structures. Because
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 239
Figure 5. Sarcoidosis. A, Airspace granuloma. B, Granulomatous vasculitis. C, Granulomatous vasculitis. Elastic tissue stain showing focal
destruction of elastic tissue. D, Granuloma with numerous apoptotic bodies surrounding a minute focus of necrosis between arrows. E, Granulomas
with multifocal minimal necrosis at arrows. F, Large foci of necrosis in granulomas (hematoxylin-eosin, original magnifications 3100 [A and E] and
3200 [B through D and F]).

the granulomas are usually localized to the lungs and and pulmonary lymphomas may also exhibit a lymphangitic
peribronchial and mediastinal lymph nodes, it is likely that localization.
the inciting antigens, although unidentified, are tiny The granulomas of sarcoidosis are usually nonnecrotizing
respirable particles that, after entering the lungs, soon enter (Figure 1, A). They may be discrete but tend to become
the pulmonary lymphatics through which they are trans- confluent. Stains for AFB and fungi should be performed in
ported to the lymph nodes. The lymphangitic distribution of all cases to evaluate the possibility of infection. Airways and
granulomas is rarely seen in small, nonsurgical biopsy pleura are often involved but pleural effusions occur
specimens; it may be evident in cryobiopsies, surgical lung infrequently. The granulomas are often uniform in appear-
biopsies, and in autopsy lungs. Berylliosis, whose micro- ance and are mostly located in the interstitium but are also
scopic appearance may closely resemble that of sarcoidosis, occasionally seen within airspaces (Figure 5, A). The
240 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
epithelioid cells are occasionally spindle shaped. In older unless reasonable efforts to exclude an infectious etiology,
cases, the granulomas may exhibit a peripheral rim of including cultures, are undertaken. NSG is always diag-
hyalinization or fibrosis that tends to become thicker over nosed in surgical lung biopsies or resections. It was
time. In some longstanding cases the granulomas may be recommended that the use of NSG as a diagnostic term
completely replaced by scar tissue, sometimes referred to as be discontinued and replaced by ‘‘sarcoidosis with an NSG
‘‘tombstone lesions,’’ and the only indication of a preexist- pattern.’’ It was suggested that ‘‘our concept of sarcoidosis
ing granuloma is the oval shape of the scar. Calcification should now be expanded to recognize that there is a
may occur in granulomatous lesions that have undergone continuous spectrum of necrosis ranging from minimal to
extensive fibrosis. extensive.’’63
Giant cells, which may contain inclusions, are usually The diagnosis of sarcoidosis requires that other causes of
present within the granulomas. Schaumann bodies are the granulomas be reasonably excluded and clinical, radiologic,
most frequently encountered inclusions; asteroid bodies, and laboratory findings should be compatible with sarcoid-
crystalline inclusions, and cholesterol crystals may also be osis. The diagnosis should be the result of multidisciplinary
found. These inclusions are nonspecific and nondiagnostic coordination and never be made by the pathologist based
and may be seen in granulomas of any etiology. Although on microscopic findings alone. Sarcoidosis should be
Hamazaki-Wesenberg bodies are most frequently seen in
diagnosed by the patient’s primary care physician or
granuloma-containing lymph nodes in sarcoidosis, they are
pulmonologist after considering and correlating all the data
rarely seen in the lungs and within granulomas.42
supplied by the various disciplines. The biopsy report should
Granulomatous vasculitis was reported as a predominat-
ing or prominent finding in two-thirds of 128 open lung specify the presence or absence of granulomas and the
biopsies53 and in 100% of 40 autopsy lungs54 (Figure 5, B results of AFB and fungal stains. Terms such as ‘‘consistent
and C). Venous involvement is more common than arterial with’’ or ‘‘suggestive of sarcoidosis’’ should be avoided
involvement; lymphatic vessels are frequently involved. because they may be misleading. A study emphasizing this
Elastic tissue stains often reveal focal destruction of elastic point sought to determine the frequency of positive
laminae (Figure 5, C). Fibrinoid necrosis, thrombosis, and microbiologic cultures in 92 adult patients with transbron-
aneurysms are not features of the granulomatous vasculitis chial biopsy specimens exhibiting nonnecrotizing granulo-
in sarcoidosis. The vasculitis can usually be identified with mas and negative histochemical stains for microorganisms.64
routine H&E staining. However, involved blood vessels in Positive cultures for Mycobacteria and fungi were obtained in
areas of confluent granulomas and or fibrosis are often specimens from 10 patients (11%); Mycobacteria were
obscured, and elastic tissue stains may aid in their cultured in 9 of 10 and fungus (Histoplasma) in 1 of 10. If
identification. cultures had not been performed, all 92 patients would likely
Although granulomatous pulmonary vasculitis is most have been diagnosed with sarcoidosis.
often seen in sarcoidosis, it is a nonspecific lesion that is not
diagnostic of sarcoidosis. It may be seen in a variety of Hypersensitivity Diseases
conditions, including tuberculosis, GPA, the NSG variant of Berylliosis.—Berylliosis is a rare occupational lung
sarcoidosis, berylliosis, foreign body embolization in intra- disease caused by exposure and development of Type IV
venous drug abusers, schistosomiasis, and seminomas. hypersensitivity to beryllium or beryllium compounds.
Sarcoidosis is an uncommon cause of pulmonary hyper- Those at risk are involved in beryllium extraction and
tension. A small number of case reports strongly suggest production, metal machining, and working in the computer,
that in some sarcoidosis patients with pulmonary hyper- aerospace, ceramics, and electronics industries. Although
tension the cause may be marked narrowing of pulmonary the microscopic appearance has been described as closely
veins secondary to granulomatous vasculitis.55,56 mimicking sarcoidosis, including nonnecrotizing granulo-
Although the granulomas of sarcoidosis have traditionally mas, hilar lymph node involvement, and lymphangitic
been considered to be nonnecrotizing, necrosis within distribution of granulomas,65 there are some differences.
granulomas may be seen in 6% to 35% of cases.17,40,57–59 Interstitial inflammation and poorly formed granulomas,
The necrosis is usually focal, involves a small percentage of usually not a feature of sarcoidosis and resembling
the granulomas and has variously been described as
hypersensitivity pneumonitis, may be helpful in distinguish-
fibrinoid, granular, eosinophilic granular, and coagulative
ing berylliosis from sarcoidosis.66–68 The diagnosis of
(Figure 5, D and E). Apoptotic nuclei of epithelioid cells and
berylliosis is highly dependent on an occupational history
lymphocytes are often seen adjacent to and within the small
foci of necrosis (Figure 5, D) but may also been seen in of exposure and can be confirmed by demonstrating the
granulomas without necrosis.60 Rare cases of classical presence of beryllium in tissues and/or body fluids and/or
sarcoidosis may exhibit larger and even confluent areas of sensitization of lymphocytes to beryllium. Other metallic
necrosis42,61 (Figure 5, F). Suppurative necrosis is a rarity in dusts or fumes including aluminum, titanium, zirconium,
sarcoidosis. and others, may also cause pulmonary granulomas.42
Based on a review of 4 decades of the world’s literature, Hypersensitivity Pneumonitis.—HP, also known as
there is strong evidence that the lesion that was described extrinsic allergic alveolitis, represents an immune-mediated
by Liebow62 in 1973, which he called NSG, is a variant of reaction to a variety of inhaled antigens and chemicals.
sarcoidosis and is likely equivalent to nodular sarcoidosis.63 Comprehensive reviews of HP69,70 report many antigens and
The finding of granulomas that are usually confluent, chemicals, and their sources, which have been associated
necrosis of varying amounts involving granulomas, and with HP. The most common causes of HP are microbial and
granulomatous vasculitis (Figure 6, A through C) are avian antigens. Farmer’s lung, caused by bacterial and
required for the diagnosis. Because the microscopic features fungal antigens, and pigeon breeders/bird fanciers’ lung,
of NSG overlap with those of infectious diseases, especially caused by avian antigens, are the 2 most common forms of
TB and fungal infections, NSG should not be diagnosed HP. Cigarette smoking is reported to be associated with a
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 241
Figure 6. A through C, Necrotizing sarcoid granulomatosis variant of sarcoidosis. A, Localization of granulomas around bronchovascular bundles.
B, Confluent granulomas with extensive necrosis. C, Granulomatous vasculitis. D through F, Subacute hypersensitivity pneumonitis. D, Diffuse
interstitial inflammation and a poorly formed granuloma. E, Bronchiolitis with poorly formed granulomas. F, Focal organizing pneumonia at arrow
(hematoxylin-eosin, original magnifications 320 [A], 3200 [B, C, and F]), and 340 [D and E]).

decreased risk of developing HP as well as reduced are basically those of acute lung injury72; small granulomas
severity.71 were present in some cases.
HP has traditionally been classified as occurring in acute, Most lung biopsies are obtained from patients with the
subacute, and chronic forms, although there are no widely subacute form of HP (SHP). Although the diagnosis of HP
accepted criteria to distinguish these various forms.69 In the can occasionally be made or suspected on examination of
transbronchial lung biopsies, surgical lung biopsies, or
acute form symptoms arise within hours after exposure,
cryobiopsies are usually required for diagnosis.
tend to peak in 18 to 24 hours, and then subside if the
The major histologic findings in SHP are as follows:
triggering agent is removed.70 Persons with acute HP almost
never have a lung biopsy. In the small number of reported 1. Chronic interstitial inflammation that appears temporally
cases in which a lung biopsy was performed, the findings uniform and is accentuated around small airways (Figure
242 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
Table 4. Comparative Histologic Features of Sarcoidosis and Subacute Hypersensitivity Pneumonitis
Sarcoidosis Subacute Hypersensitivity Pneumonitis
Interstitial inflammation Inconspicuous, if present Always present. Peribronchiolar accentuation
Granulomas Well formed, compact, may have Small, loosely formed, no necrosis. Giant cells
focal, minimal necrosis may be present without granulomas
Schaumann bodies Frequently present Frequently present
Other inclusions May be present May be present
Lymphangitic localization of granulomas Yes No
Organizing pneumonia No Yes
Nongranulomatous inflammation of small airways Unusual Yes. Chronic inflammation 6 granulomas
Granulomatous vasculitis Common in surgical lung biopsies Rare
Pleural granulomas Up to 35%25 Not reported

6, D). Lymphocytes predominate; plasma cells are Detailed description of the pathologic findings in chronic
frequently present, but eosinophils are rare. hypersensitivity pneumonitis (CHP) are presented in a
2. Bronchiolitis (Figure 6, E) that may or may not be comprehensive review by Churg et al.74 CHP presents as a
accompanied by peribronchiolar metaplasia, bronchiol- chronic, fibrosing form of interstitial pneumonia; features of
ectasis, granulomas, and fibrosis of bronchiolar walls. SHP may be present as well. Fibrosis is required for the
3. Small, poorly formed (loose) nonnecrotizing granulomas diagnosis of CHP.74 Lung biopsy findings in CHP need to be
(Figure 6, D and E) and/or individual, scattered giant distinguished from those of a variety of idiopathic interstitial
cells are present in approximately 70% of cases.34 The pneumonias, a challenging task requiring multidisciplinary
granulomas are usually located in the interstitium or coordination. CHP may have histologic features that overlap
close to, or within, small airways. Intra-alveolar granu- with usual interstitial pneumonia, including patchy fibrosis,
lomas and giant cells can also be seen. subpleural/paraseptal localization, fibroblastic foci, and
4. Schaumann bodies and/or cholesterol crystals may be honeycombing.74 Separation of usual interstitial pneumonia
seen within the cytoplasm of giant cells; they are from CHP is crucial because usual interstitial pneumonia is
nonspecific and have no diagnostic value. The finding treated with antifibrotic agents, whereas CHP is treated with
of individual, extracellular Schaumann bodies is evidence immunosuppressive agents.74 Those with CHP have a
of preexisting granulomas. Other nonspecific inclusions significantly better prognosis after lung transplantation than
in giant cells may be present with lesser frequency than those with usual interstitial pneumonia, probably because
Schaumann bodies. they are less likely to develop bronchiolitis obliterans.75 The
5. Foci of organizing pneumonia are frequently present following histologic findings tend to favor CHP over usual
(Figure 6, F). interstitial pneumonia: upper zone, peribronchiolar, and
6. Obliterative bronchiolitis may be present in some cases. bridging fibrosis; giant cells, granulomas (usually rare),
7. Lymphoid follicles may be present in some cases. prominent interstitial inflammation, and organizing pneu-
8. In a minority of cases, SHP may present as the cellular monia.74 Despite the above-mentioned criteria, the diagno-
variant of nonspecific interstitial pneumonia. sis of CHP is difficult and often controversial.
9. Small foci of endogenous lipid pneumonia may be Hot Tub Lung.—Hot tub lung is associated with
present secondary to bronchiolar obstruction. exposure to MAC when present in aerosols from indoor
hot tubs/spas as well as swimming pools, showers,
The triad of interstitial pneumonitis, cellular bronchiolitis, humidifiers, and wind instruments.3,70,76,77 High-resolution
and poorly formed granulomas is seen in 80% of well- computerized tomography findings are usually diffuse
documented cases of SHP.73 centrilobular nodules and/or ground glass opacities. Cavi-
A history of exposure to respirable antigens or chemicals tary nodules have also been reported. It is generally
is helpful when correlated with histologic and radiographic considered to be a form of HP, but because MAC has been
findings. However, failure to identify a relevant antigenic cultured from lung tissue77 there is uncertainty as to whether
exposure is not unusual even when the histologic findings it represents an infection, hypersensitivity, or a combination
are typical of SHP. The most important differential of both.70 Biopsy usually shows nonnecrotizing granulomas
diagnostic consideration is distinguishing SHP from sar- that may be localized around airways; necrotizing granulo-
coidosis (Table 4). Nonspecific interstitial pneumonia might mas are sometimes present. Granulomas are present in the
be a diagnostic consideration in those unusual cases of SHP lumens of small bronchioles3 as well as in the interstitium
where granulomas or giant cells are not found. However, and airspaces (Figure 7). The granulomas are usually larger
the interstitial inflammation in nonspecific interstitial and more well-formed than those seen in nonmycobacterial
pneumonia is usually diffuse and not localized around small HP. Organizing pneumonia is often present. AFB stains of
airways and there is no small airway inflammation or foci of granulomas are usually negative but may be positive.3
organizing pneumonia. Lymphocytic interstitial pneumonia Lymphangitic distribution of granulomas and granuloma-
is usually characterized by dense interstitial lymphocytic or tous vasculitis are characteristic features of sarcoidosis that
lymphoplasmacytic infiltrates and nodules, sometimes are not seen in hot tub lung. Although it has been stated
exhibiting germinal centers, which do not exhibit peribron- that the exclusive interstitial location of granulomas in
chiolar localization. Poorly formed granulomas, as seen in sarcoidosis is a reliable distinguishing feature between
many lymphoproliferative disorders, may be present within sarcoidosis and hot tub lung,3 in the author’s experience,
the lymphocytic infiltrates. airspace granulomas also occur in sarcoidosis (Figure 5, A).
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 243
surrounding a minute focus of eosinophilic necrosis (Figure
8, C). Granulomatous inflammation is sometimes centered
on bronchioles; if it is a prominent finding it may be
confused with bronchocentric granulomatosis. Well-formed,
compact granulomas as occur in sarcoidosis are rarely seen;
if present, a diagnosis other than GPA should be
considered.
Vasculitis may involve arteries, veins (Figure 8, D and E),
and capillaries (Figure 8, F) and should only be diagnosed in
inflamed areas outside of areas of necrosis. Vasculitis may
involve the entire vessel wall or involvement may be focal
(Figure 8, D). Vasculitis in GPA may be nonnecrotizing or
necrotizing. The former, which is characterized by vascular
infiltration by lymphocytes and other inflammatory cells, is
not considered to be a diagnostic feature of GPA.2,3
Necrotizing vasculitis, required for the diagnosis of GPA2,3;
is characterized by suppurative necrosis, suppurative gran-
ulomas, fibrinoid necrosis, or the presence of karyorrhectic
Figure 7. Airspace granuloma and interstitial inflammation in hot tub neutrophils within the vessel wall2,3 (Figure 8, D and E).
lung (hematoxylin-eosin, original magnification 3200). Courtesy of Destruction of elastic laminae is best visualized with an
Sanjay Mukhopadhyay, MD. elastic tissue stain. Inflammation may be transmural or
involve only the intima. Necrotizing granulomatous vascu-
litis may be seen occasionally, however, in most cases of
Hot tub lung can be suspected, but not diagnosed, based on
GPA the vasculitis is nongranulomatous. Capillary involve-
biopsy findings alone. In addition to histologic findings,
ment (capillaritis) is characterized by neutrophils, usually
definitive diagnosis requires a history of indoor hot tub use
exhibiting karyorrhexis, and fibrinoid necrosis within the
or exposure to aerosolized water contaminated with MAC.
capillary walls (Figure 8, F). Capillaritis is not a specific
Vasculitic Granulomatous Diseases finding for GPA but may be the only finding in a biopsy
specimen. Patients presenting with capillaritis often have
Granulomatosis With Polyangiitis.—GPA, formerly severe hemoptysis.
Wegener granulomatosis, is a rare systemic, immune- GPA uncommonly presents with microscopic variants
mediated inflammatory process of undetermined etiology, that are characterized by the predominance of a single
mostly occurring in adults. The lesions are characterized by feature that may make diagnosis difficult. These are the
necrosis, necrotizing vasculitis, and granulomas that pre- bronchocentric, organizing pneumonia and capillaritis with
dominantly involve the upper and lower respiratory tract hemorrhage variants.3
and kidneys. Head and neck involvement are most common Diagnosis of GPA is significantly aided by the serum
followed by lung involvement that might be the only antineutrophilic cytoplasmic antibody (ANCA) test. The 2
presenting feature; hilar lymph nodes are usually not immunofluorescence patterns observed are the cytoplasmic
involved. Radiographic appearance at presentation is usually (C-ANCA), the most common, and the perinuclear (P-
multiple nodular or mass lesions that frequently exhibit ANCA) pattern. C-ANCA and P-ANCA are directed against
cavitation or, rarely, a solitary nodule with or without proteinase 3 and myeloperoxidase, respectively. Both may
cavitation. Diffuse airspace disease seen on computed occur in GPA but the C-ANCA pattern is by far the most
tomography examination usually reflects hemorrhage from common, having been reported in 84% to 99% of active,
capillaritis. generalized cases and in 60% of localized cases.78 There are
A surgical biopsy may be required in cases with atypical no significant differences in the lung biopsy findings of
clinical presentations. The most frequent atypical scenario is those expressing C-ANCA or P-ANCA.79,80 The P-ANCA
the presence of solitary or multiple nodules with or without pattern is more often seen in microscopic polyangiitis,
cavitation or cavitary infiltrates in patients without evidence polyarteritis nodosa, and eosinophilic granulomatosis with
of multisystem involvement (localized/limited GPA).6 GPA polyangiitis.
limited to lung involvement is common. Before making a diagnosis of GPA it is mandatory to
The major histologic findings are granulomatous inflam- exclude the possibility of infection using AFB and fungal
mation, necrosis, and vasculitis accompanied by a mixed stains and, ideally, cultures. The diagnosis of GPA requires
inflammatory cell infiltrate and variable amounts of fibro- multidisciplinary collaboration and cannot be made or
blastic proliferation. Organizing pneumonia is a frequent excluded based on the presence of C-ANCA positivity
finding and may occasionally be the predominating finding alone. C-ANCA may be negative when involvement is
in biopsy specimens. Necrosis appears as microabscesses, limited to the lungs and there may be false positives.
large, geographic zones of necrosis (Figure 8, A) that often Eosinophilic Granulomatosis With Polyangiitis.—Eo-
have a ‘‘dirty’’ appearance due to the presence of sinophilic granulomatosis with polyangiitis, formerly
neutrophils and necrotic debris and necrosis of collagen. Churg-Strauss syndrome, is a multisystem disorder mostly
The granulomatous inflammation may consist of scattered affecting adults and belonging to the small-vessel ANCA-
single or loose clusters of giant cells, small suppurative associated vasculitides. It is characterized by eosinophil-rich
granulomas (Figure 8, B), giant cells surrounding microab- and granulomatous inflammation often involving the
scesses, foci of geographic necrosis surrounded by palisaded respiratory tract, and necrotizing vasculitis, predominantly
histiocytes, and microgranulomas consisting of small foci of involving small-sized to medium-sized vessels.81 It is
palisaded histiocytes arranged in a cartwheel fashion and associated with asthma in 95% of patients and eosinophilia.
244 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
Figure 8. A through F, Granulomatosis with polyangiitis. A, Extensive geographic necrosis. B, Granuloma with suppurative necrosis. C, Granuloma
with a small focus of central eosinophilic necrosis and focal palisading of peripheral histiocytes. D, Necrotizing vasculitis; partial involvement of the
vessel wall. E, Necrotizing vasculitis. F, Capillaritis with acute inflammation and fibrinoid necrosis of alveolar capillary walls. G and H, Eosinophilic
granulomatosis with polyangiitis. G, Fibrinoid necrosis of a pulmonary blood vessel with surrounding eosinophilic pneumonia. H, Eosinophilic
pneumonia (hematoxylin-eosin, original magnifications 320 [A] and 3200 [B through H]).

Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 245
The lungs are one of the most frequently involved sites.
Aside from asthma, patients may present with sinusitis,
allergic rhinitis, and nasal polyposis. The diagnosis is based
mostly on the following clinical criteria: asthma, eosino-
philia, neuropathy, nonfixed pulmonary infiltrates, parana-
sal sinus abnormalities, and a biopsy containing a blood
vessel with extravascular eosinophils.82 P-ANCA is present
in approximately 70% of cases.83 Biopsies are usually
obtained from extrapulmonary sites, especially skin. In the
lungs a combination of eosinophilic pneumonia, granulo-
matous inflammation, and necrotizing vasculitis is consid-
ered pathognomonic, but that combination is present
infrequently.83,84 Granulomas are usually necrotizing and
often appear as small groups of palisaded histiocytes
surrounding foci of central necrosis. The vasculitis involves
arteries and veins and exhibits either granulomatous
inflammation with or without prominent giant cells or a Figure 9. Bronchocentric granulomatosis, idiopathic (hematoxylin-
nongranulomatous mixed chronic inflammatory cell infil- eosin, original magnification 3100).
trate with many eosinophils.2 Fibrinoid necrosis of blood
vessel walls in often seen (Figure 8, G). Eosinophilic center where the bronchiolar lumen is located. The etiology
pneumonia may occur alone or may accompany necrotizing of the nonasthmatic cases is varied and, in many cases,
vasculitis, but usually without granulomatous inflammation2 unknown; blood and tissue eosinophilia are usually absent.
(Figure 8, H).
The necrotizing granulomas are centered on bronchioles
Autoimmune Granulomas (Figure 9) and there is often palisading of histiocytes. There
may be partial or complete involvement of the circumfer-
Rheumatoid Nodule.—Pulmonary rheumatoid nodules ence of the bronchiole. The presence of small remnants of
are rare, representing only 0.2% of cases of pulmonary bronchiolar epithelium in the center of the granuloma
granulomatous diseases.5 The pulmonary nodules are provides good evidence for bronchiolar involvement. If
palisaded granulomas with a central area of eosinophilic
there is no bronchiolar epithelium within the granuloma,
or basophilic necrosis surrounded by palisaded histiocytes;
location of the granuloma next to a pulmonary artery or
they are identical in appearance to those encountered in
arteriole strongly suggests that the granuloma involves a
subcutaneous tissue. Most patients also have subcutaneous
bronchiole. Elastic tissue stains are often helpful in
nodules. The lung nodules are usually multiple and
identifying pulmonary arteries and bronchioles that may
peripheral, subpleural location is common2; the pleura
be obscured by dense inflammation. Pulmonary arterioles
may also be involved. There may an associated nonnecrotiz-
ing vasculitis. The presence of multinucleate giant cells, may be secondarily inflamed.
neutrophils, and eosinophils admixed with the palisaded The diagnosis of BG can be made with confidence in
histiocytes would be unusual and should suggest another asthmatic patients with eosinophilia if an infectious etiology
diagnosis.2 As with any necrotizing granulomatous condi- has been reasonably excluded. Pathologists should be
tion, the possibility of infection should be excluded with hesitant to diagnose BG in nonasthmatic patients, even if
special stains for microorganisms and cultures, if possible. special stains and cultures for microorganisms are negative,
The diagnosis of pulmonary rheumatoid nodule can be because infection is still possible and other noninfectious
made with confidence if an infectious etiology can be conditions may simulate BG. Bronchocentric granulomas
reasonably excluded and the patient is known to have may occur in GPA, rheumatoid arthritis, and infections.
rheumatoid disease. If the history is not known or if the Tuberculosis and other infections, including MAC, and
patient is known not to have rheumatoid disease, the histoplasmosis, may present with bronchocentric granulo-
possibility of rheumatoid nodule can be included in the mas.87 GPA can be distinguished from BG by the presence
differential diagnosis that would also include infection, of necrotizing vasculitis, which is not a feature of BG.
GPA, and others (Table 2). Secondary vasculitis, which is nonnecrotizing, may occur in
BG. Rheumatoid nodules are usually subpleural but can
Mixed Allergic and Idiopathic Granulomatous Reactions simultaneously involve other areas of the lung including
Bronchocentric Granulomatosis.—Bronchocentric airways. BG, GPA, and rheumatoid nodules may exhibit
granulomatosis (BG) is characterized by necrotizing gran- peripheral palisading of histiocytes. The pathology report
ulomas that primarily involve and destroy bronchioles. should provide a differential diagnosis that includes BG,
Approximately half of the cases occur in asthmatics with infection, and others depending on the patient’s medical
allergic bronchopulmonary fungal disease, mainly aspergil- history.
losis, associated with blood and tissue eosinophilia.85 In
Foreign Body Reactions
these patients, BG is generally considered to be a
manifestation of fungal hypersensitivity.85,86 Eosinophilic Foreign bodies can reach the lung parenchyma via
pneumonia and/or mucoid impaction of bronchi, both aspiration, embolization, and inhalation; they usually incite
additional manifestations of allergic bronchopulmonary a nonnecrotizing granulomatous reaction. In contrast to
fungal disease, may also be present. Except in rare cases, epithelioid granulomas, foreign body granulomas are
fungi are not found in the granulomas. In those cases where composed of giant cells and macrophages in varying
fungi are found, they are usually present in the necrotic numbers.
246 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
Figure 10. A, Aspirated vegetable fragment with giant cells and acute inflammation. B, Pale blue-green staining talc crystals with dense fibrosis and
giant cell reaction in pleura; postpleurodesis. Movat pentachrome stain. Courtesy of Sanjay Mukhopadhyay, MD. C, Extravascular embolized
microcrystalline cellulose with giant cell reaction in an intravenous drug abuser. D, Embolized crospovidone in an airspace with giant cell reaction in
an intravenous drug abuser (hematoxylin-eosin, original magnifications 3100 [A] and 3200 [C and D]; Movat pentachrome, original magnification
3200 [B]).

Aspiration.—Aspiration occurs in persons who have present in the stomach at the time of aspiration, foreign
conditions that impair their gag, cough, and swallowing bodies may be difficult or impossible to find. A clue to
reflexes. Aspirated foreign materials usually consist of food aspiration in the absence of foreign material and granulo-
items, especially vegetable fragments and skeletal muscle. mas is the presence of scattered giant cells along with acute
Barium, pill fragments, and other materials may also be and/or organizing pneumonia. This finding, in the proper
aspirated. Most aspirated particles greater than 10 lm in clinical setting, should encourage a search for foreign
diameter are deposited in the throat and upper airways and material by obtaining deeper sections and/or examination
cannot penetrate the lower tissues of the respiratory tract. If of additional tissue. When the stomach is empty, only
the individual survives an episode of aspiration, pneumonia gastric juice will be aspirated; this usually results in diffuse
follows as a reaction to foreign material, gastric acid, and alveolar damage.88
bacteria originating from the upper aerodigestive tract. Aspiration pneumonia is most often detected at autopsy
Aspiration pneumonia can be diagnosed with certainty only but can also be diagnosed in biopsies. It often goes
by identifying foreign bodies. It is characterized by acute and unrecognized by both clinicians and pathologists. In a
organizing inflammation usually accompanied by one or review of 59 surgical lung specimens exhibiting aspiration
more of the following: abscess formation, suppurative pneumonia, aspiration was suspected clinically in only 4 of
granulomas, and a giant cell reaction. Aspirated vegetable 45 (9%) of cases in which a clinical impression or differential
material usually incites a giant cell reaction, whereas skeletal diagnosis was provided.89 In a report of 100 consecutive
muscle fibers usually do not. Vegetable fragments can be granulomas in a pulmonary pathology consultation practice,
recognized by their latticework arrangement of square or it was determined that aspiration pneumonia was the
rectangular large cells with thick cell walls composed of condition most frequently unrecognized by the referring
cellulose. They are often infiltrated by inflammatory cells pathologist.4
and surrounded by giant cells (Figure 10, A). The Embolization.—Embolization of foreign bodies to the
appearance of aspirated foreign bodies varies significantly lungs that incite a granulomatous reaction occurs most
depending upon the degree of digestive changes and frequently as the result of intravenous drug abuse and
degenerative changes following aspiration. Skeletal muscle complications of therapeutic procedures. The landmark
fibers often lose their striations. If little food material was publication by Tomashefski and Hirsch in 198090 called
Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 247
attention to pulmonary lesions produced by intravenous tumor necrosis factor-alpha antagonists, interferon, or
injection of solutions prepared from tablets or capsules polyethylene glycol interferon therapeutics and immune
intended for oral consumption. The most frequently injected checkpoint inhibitors. The authors stated that the clinical/
medications are opioids and amphetamines. Aside from the radiologic presentations of ‘‘drug-induced sarcoidosis’’ are
active ingredient, pills/tablets contain fillers/excipients that usually close to sarcoidosis but there was no specific
provide bulk, adhesion, and other properties. Although mention of the presence or absence of lung granulomas.
cornstarch and talc were commonly used as fillers in the However, because the reactions were said to resemble
past, microcrystalline cellulose (a binder) and crospovidone sarcoidosis, it seems reasonable to assume that there was
(a disintegrant) are mostly encountered at present. When lung involvement. Of the 59 drugs identified, the 10 most
the fillers enter the lung via the pulmonary arterial strongly associated with inciting granulomas are interferon-
circulation, they are trapped in arterioles and capillaries alpha 2a, interferon-alpha 2b, ribavirin, peginterferon-alpha
and incite a florid giant cell reaction in and around the 2a, peginterferon 2b, interferon-alpha, interferon-alpha 1a,
vessels and in perivascular parenchyma. Cornstarch gran- pembrolizumab, bosentan, and ipilimumab. Other drugs
ules have a spherical shape and exhibit Maltese cross that have been implicated in causing pulmonary granulomas
birefringence. Talc crystals are large and have a sheet-like or are the Bacillus Calmette-Guerin vaccine, etanercept, ever-
plate-like appearance (Figure 10, B) and may exhibit a olimus, sirolimus, fluoxetin, isoniazid, imatinib, mesal-
yellowish tint in H&E-stained sections. Microcrystalline amine, mesylate, nivolumab, sulfasalazine, and
cellulose appears as bundles of long, rod-like crystals ustekinumab. The terminology that has been used for these
(Figure 10, C). The distinction between microcrystalline drug reactions includes the terms ‘‘sarcoidosis-like reac-
cellulose and talc in H&E-stained sections may be difficult. tions’’ and ‘‘drug-induced-sarcoidosis.’’ In the author’s
This distinction can be made with the modified Russell opinion, the term ‘‘drug-induced granulomatous disease’’
Movat pentachrome stain that stains microcrystalline seems preferable because sarcoidosis is not yet defined.
cellulose yellow and talc, light blue-green91 (Figure 10, B).
The modified Russell Movat pentachrome stain is techni- Granulomas Associated With a Defective Enzyme
cally challenging and may give inconsistent results unless Chronic Granulomatous Disease.—Chronic granulo-
performed by an experienced histotechnologist. Crospovi- matous disease (CGD) is a diverse group of rare, hereditary
done fragments stain dark blue with H&E and have a very diseases characterized by production of defective phagocyte
distinctive, unique, coral-like appearance (Figure 10, D). All nicotinamide adenine dinucleotide phosphate oxidase by
the filler materials, except crospovidone, exhibit strong
cells of the immune system, thus impairing the ability of
birefringence when examined with polarized light.
neutrophils and macrophages to kill ingested pathogens.100
Silicone leakage from breast implants may rarely embolize
Approximately 90% of those with CGD are males who have
to the lungs. Total parenteral nutrition-associated crystalline
mutations on the X-linked CYBB gene coding for
precipitates resulting in pulmonary vascular occlusions and
gp91phox.100
alveolar granulomas have been reported.92
CGD is diagnosed in childhood, usually before age 5; it is
Inhalation.—Many types of inhaled particulate matter
rare in adults. Patients with CGD typically experience
are capable of inciting pulmonary granulomas. Lung injury
recurrent bouts of fungal and bacterial infections, most
associated with inhalation of opioids (usually cocaine or
heroin) and amphetamines is usually caused by the talc with frequently involving the lungs. The histopathologic features
which these drugs are mixed. The talc crystals incite a of lung biopsy specimens obtained from 20 patients with
granulomatous reaction that is not perivascular and consists CGD were described in detail by Moskaluk et al.101
primarily of giant cells; a similar reaction occurs with Granulomas were present in all cases. They were suppura-
inhaled crack-cocaine.93 Cocaine users may also develop tive, necrotizing, and nonnecrotizing. Occasional large foci
spontaneous pneumothorax.94 The presence of granulomas of confluent, geographic necrosis was present surrounded
of undetermined etiology in surgical specimens of lung by palisaded histiocytes. Abscess formation was more
obtained for treatment of spontaneous pneumothorax, frequently associated with fungal infections than bacterial
should raise the possibility of cocaine abuse. Inhaled metals infections. In addition to granulomas and abscesses, there
that promote granuloma formation include aluminum, was diffuse chronic inflammation with foci of fibrosis. Fungi
barium, beryllium, cobalt copper, gold, rare earths (lantha- and bacteria were cultured from 11 of 20 (56%) and 6 of 20
nides), titanium, and zirconium.95 Inhalation of hair spray (30%) of specimens, respectively. Aspergillus spp were the
may incite the formation of granulomas.96 A history of most frequently isolated fungi. Pathologists can suspect the
exposure is essential for consideration of the diagnosis of diagnosis of CGD based on knowing the patient’s age,
inhalation-induced granulomas. The medical literature history of recurrent lung infections, and the histologic
contains a multitude of case reports of many other materials findings. However, there are no histologic findings that are
that, when inhaled, cause pulmonary granulomas. diagnostic of CGD.

Drug-Induced Granulomas Granulomas Associated With Primary Antibody Deficiency


A variety of drugs are sometimes associated with Common Variable Immunodeficiency.—Combined
production of granulomas. The medical literature contains variable immunodeficiency (CVID), a rare disease, is the
mostly individual case reports as well as few small case most common symptomatic primary antibody deficiency
series.97,98 A review of the World Health Organization disease.102 It occurs in children and adults. Hypogamma-
Pharmacovigilance Database by Cohen Aubart et al99 found globulinemia leads to recurrent bacterial infections. The
59 drugs that produce granulomas that the authors referred lower and upper respiratory tracts are the major sites of
to as ‘‘drug-induced sarcoidosis.’’ Overall, 85.5% of these infections. Bacterial pneumonia is the most frequent acute
drug reactions were considered ‘‘serious’’ and 3.5% were infection; viral infections may also occur. Chronic lung
fatal. Most of the drugs associated with granulomas were disease, including bronchiectasis, chronic obstructive pul-
248 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
monary disease, and asthma, as well as chronic sinusitis, preferable (ie, ‘‘cancer-related granulomatosis’’ or ‘‘sar-
develops in many patients.102 At least 10% to 20% of CVID coid-like reaction’’).
patients develop interstitial lung disease (ILD) resulting
from immune dysregulation.103 ILD in these patients CONCLUSIONS
includes follicular bronchiolitis, nodular lymphoid hyper- Granulomas are ubiquitous, occurring in almost every
plasia, granulomatous lung disease, lymphocytic interstitial disease category. Although sarcoidosis and mycobacterial
pneumonia, nonspecific interstitial pneumonia, and orga- infection each account for approximately 25% of pulmonary
nizing pneumonia. The radiologic and histologic heteroge- granulomas worldwide, there is significant geographic
neity of ILDs in CVID and other types of primary antibody variation in their incidence. Pathologic diagnosis is often
deficiency disease has led to the coining of the umbrella challenging, and a definitive diagnosis, which can only be
term ‘‘GLILD,’’ granulomatous lymphocytic interstitial lung achieved by demonstrating an etiologic agent within
disease, which encompasses granulomatous disease and all granulomas or by culture, is not achieved in a substantial
forms of pulmonary lymphoid hyperplasia.104 The term is percentage of pulmonary granulomas. The presence or
said to be applicable to any type of interstitial lymphocytic absence of necrosis in granulomas are not completely
infiltrate and/or granuloma in the lung of a patient with a reliable indicators of the presence or absence of infection.
primary antibody deficiency disease if other conditions that Multidisciplinary interaction and coordination in cases
can produce these findings, including infections, other where an etiologic agent is not identified microscopically
defined ILDs, and malignant lymphoproliferative diseases, or by culture often helps to establish a diagnosis; its
have been excluded.105 Granulomas are usually nonnecro- importance cannot be overemphasized.
tizing. After recently studying 34 surgical lung biopsy cases
References
of CVID and 4 of IgA deficiency (IgAD) Larsen et al106
1. Ulbright TM, Katzenstein AL. Solitary necrotizing granulomas of the lung:
concluded that ‘‘GLILD is neither a specific nor a useful differentiating features and etiology. Am J Surg Pathol. 1980;4(1):13–28.
entity and biopsies from CVID and IgAD deficient patients 2. El-Zammar OA, Katzenstein AL. Pathological diagnosis of granulomatous
should be diagnosed simply by microscopic pattern(s) lung disease: a review. Histopathology. 2007;50(3):289–310.
3. Mukhopadhyay S, Gal AA. Granulomatous lung disease: an approach to the
observed.’’ There are no specific histologic findings that differential diagnosis. Arch Pathol Lab Med. 2010;134(5):668–690.
enable a diagnosis of CVID. 4. Hutton Klein JR, Tazelaar HD, Leslie KO, Colby TV. One hundred
consecutive granulomas in a pulmonary pathology consultation practice. Am J
Granulomas Associated With Cancer Surg Pathol. 2010;34(10):1456–1464.
5. Mukhopadhyay S, Farver CF, Vaszar LT, Dempsey OJ, Popper HH, Mani H.
Pulmonary granulomatous reactions associated with Causes of pulmonary granulomas: a retrospective study of 500 cases from seven
cancer may be encountered in a variety of circumstances. countries. J Clin Pathol. 2012;65(1):51–57.
6. Mukhopadhyay S, Aubry MC. Pulmonary granulomas: differential
The most common is the occasional presence of granulomas diagnosis, histologic features and algorithmic approach. Diagn Histopathol.
immediately adjacent to any subtype of lung carcinoma and/ 2013;19(8):288–297.
or in regional draining lymph nodes; they are usually 7. Mukhopadhyay S, Wilcox BE, Myers JL, Bryant SC, Buckwalter SP,
nonnecrotizing epithelioid granulomas or single or multiple Wengenack N. Pulmonary necrotizing granulomas of unknown cause: clinical
and pathologic analysis of 131 patients with completely resected nodules. Chest.
giant cells. Their pathogenesis is poorly understood; it is 2013;144(3):813–824.
assumed that the inciting agents are tumor antigens. Similar 8. Woodard BH, Rosenberg SI, Farnham R, Adams DO. Incidence and nature
granulomatous reactions have been reported with carcino- of primary granulomatous inflammation in surgically removed material. Am J
Surg Pathol. 1982;6(2):119–129.
mas arising in any organ. Brincker107 reported granuloma- 9. Adams DO. The biology of the granuloma. In: Ioachim HL, ed. Pathology of
tous reactions in 165 of 3770 (4.4%) of carcinomas. In Granulomas. New York, NY: Raven; 1983:1–20.
heavily keratinized squamous carcinoma, a foreign body 10. Beijer E, Veltkamp M, Meek B, Moller DR. Etiology and immunopatho-
genesis of sarcoidosis: novel insights. Semin Respir Crit Care Med. 2017;38(4):
giant cell reaction to extruded keratin is sometimes seen. 404–416.
Granulomatous reactions have been reported in 210 of 1551 11. Kosjerina Z, Zaric B, Vuckovic D, Lalosevic D, Djenadic G, Murer B. The
(13.5%) and 8 of 110 (7.3%) of Hodgkin and non-Hodgkin sarcoid granuloma: ‘‘epithelioid’’ or ‘‘lymphocytic-epithelioid’’ granuloma?
Multidiscip Respir Med. 2012;7(1):11.
lymphomas, respectively108; they occur in T-cell lymphomas 12. Medlar EM. A study of the process of caseation in tuberculosis. Am J
and B-cell lymphomas. Patients with chronic, active Pathol. 1926;2(4):275–290.
sarcoidosis are at increased risk of developing lympho- 13. Su WP, Kuechle MK, Peters MS, Muller SA. Palisading granulomas caused
mas109; this has been referred to as the ‘‘sarcoidosis- by infectious diseases. Am J Dermatopathol. 1992;14(3):211–215
14. Rosen Y, Vuletin JC, Pertschuk LP, Silverstein E. Sarcoidosis from the
lymphoma syndrome.’’108 Pulmonary granulomas may be pathologist’s vantage point. Pathol Annu 1979;14(Part 1):405–439.
associated with administration of therapeutic agents, such 15. Visscher D, Churg A, Katzenstein AL. Significance of crystalline inclusions
as BCG and interferons, and other drugs used to treat cancer in lung granulomas. Mod Pathol. 1988;1(6):415–419.
16. Jones Williams W. The nature and origin of Schaumann bodies. J Pathol
(see section on drug-induced granulomas). Bacteriol. 1960;79:193–201.
It is well known that a systemic granulomatous condition 17. Ricker W, Clark M. Sarcoidosis; a clinicopathologic review of 300 cases,
that may involve the lungs, sometimes referred to as including 22 autopsies. Am J Clin Pathol. 1949;19(8):725–749.
18. Cain H, Kraus B. The ultrastructure and morphogenesis of asteroid bodies
‘‘sarcoidosis,’’ can develop before, concomitant with, or in sarcoidosis and other granulomatous disorders. In: Williams JW, Davies BH
after a diagnosis of certain solid tumors. These tumors eds. Eighth International Conference on Sarcoidosis and Other Granulomatous
include lymphoma, melanoma, testicular germ cell tumors Diseases. Alpha Omega; 1980:30–37.
19. Hamazaki T. Über ein neues, säuerfeste Substanz führendes Spindelkör-
(especially seminoma), and carcinomas of the cervix, liver, perchen der Menschlischen Lymphdrüsen. Virchows Arch (Pathol Anat). 1938;
lung, uterus, breast, and head and neck.110–112 The termi- 301;490–522.
nology used for these granulomatous conditions is some- 20. Wesenberg W. Uber säurefeste ‘‘Spindelkörper Hamazaki’’ bei Sarkoidose
der Lymphknoten und über doppellichtbrechende Zelleinschlüsse bei Sarkoidose
what confusing and controversial and includes der Lungen. Arch Klin Exp Dermatol. 1966;227(1):101–107.
‘‘sarcoidosis,’’ ‘‘sarcoid reaction,’’ and ‘‘sarcoid-like reac- 21. Doyle WF, Brahman HD, Burgess JH. The nature of yellow-brown bodies
tion.’’ Since there is not yet a precise definition of in peritoneal lymph nodes. Arch Pathol. 1973;96(5):320–326.
sarcoidosis, it is inappropriate to label these granulomatous 22. Jain D, Ghosh S, Teixeira L, Mukhopadhyay S. Pathology of pulmonary
tuberculosis and non-tuberculous mycobacterial lung disease: facts, misconcep-
reactions as sarcoidosis. Until we know more about these tions, and practical tips for pathologists. Semin Diagn Pathol. 2017;34(6):518–
conditions and sarcoidosis, descriptive terminology is 529.

Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 249
23. Hillemann D, Galle J, Vollmer E, Richter E. Real-time PCR assay for 51. Jacob M, Bastos HN, Mota PC, et al. Diagnostic yield and safety of
improved detection of Mycobacterium tuberculosis complex in paraffin- transbronchial cryobiopsy in sarcoidosis. ERJ Open Res. 2019;5(4):00203–2019.
embedded tissues. Int J Tuberc Lung Dis. 2006;10(3):340–342. 52. Rosen Y, Athanassiades TJ, Moon S, Lyons HA. Nongranulomatous
24. Kommareddi S, Abramowsky CR, Swinehart GL, Hrabak A. Non- interstitial pneumonitis in sarcoidosis. Relationship to development of epithelioid
tuberculous mycobacterial infections: comparison of the fluorescent auramine- granulomas. Chest. 1978;74(2):122–125.
O and Ziehl-Neelsen techniques in tissue diagnosis. Hum Pathol. 1984;15(11): 53. Rosen Y, Moon S, Huang CT, Lyons HA. Granulomatous pulmonary
1085–1089. angiitis in sarcoidosis. Arch Pathol Lab Med. 1977;101(4):170–174.
25. Vaerewijck MJ, Huys G, Palomino JC, Swings J, Portaels F. Mycobacteria in 54. Takemura T, Matsui Y, Saiki S, Mikami R. Pulmonary vascular involvement
drinking water distribution systems: ecology and significance for human health. in sarcoidosis: a report of 40 autopsy cases. Hum Pathol.1992;23(11):1216–
FEMS Microbiol Rev. 2005;29(5):911–934. 1223.
26. Park JS, Kang YA, Kwon SY, et al. Nested PCR in lung tissue for diagnosis of 55. Smith LJ, Lawrence JB, Katzenstein AA. Vascular sarcoidosis: a rare cause
pulmonary tuberculosis. Eur Respir J. 2010;35(4):851–857. of pulmonary hypertension. Am J Med Sci. 1983;285(1):38–44.
27. Seo AN, Park HJ, Lee HS, et al. Performance characteristics of nested 56. Crissman JD, Koss M, Carson RP. Pulmonary veno-occlusive disease
polymerase chain reaction vs real-time polymerase chain reaction methods for secondary to granulomatous venulitis. Am J Surg Pathol.1980;4(1):93–99
detecting mycobacterium tuberculosis complex in paraffin-embedded human 57. Longcope WT, Freiman DG. A study of sarcoidosis; based on a combined
tissues. Am J Clin Pathol. 2014;142(3):384–390. investigation of 160 cases including 30 autopsies from The Johns Hopkins
28. Lee HS, Park KU, Park JO, Chang HE, Song J, Choe G. Rapid, sensitive, and Hospital and Massachusetts General Hospital. Medicine. 1952;31(1):1–132.
specific detection of Mycobacterium tuberculosis complex by real-time PCR on 58. Thomson AD. The pathology of sarcoidosis. Postgrad Med J. 1958;34(391):
paraffin-embedded human tissues. J Mol Diagn. 2011;13(4):390–394. 248–253.
29. Kohli R, Punia RS, Kaushik R, Kundu R, Mohan H. Relative value of 59. Carrington CB, Gaensler EA, Mikus JP, Schachter AW, Burke GW, Goff
immunohistochemistry in detection of mycobacterial antigen in suspected cases AM. Structure and function in sarcoidosis. Ann N Y Acad Sci. 1976;278:265–283.
of tuberculosis in tissue sections. Indian J Pathol Microbiol. 2014;57(4):574–578. 60. Kunitake R, Kuwano K, Miyazaki H, Hagimoto N, Nomoto Y, Hara N.
30. Purohit MR, Sviland L, Wiker H, Mustafa T. Rapid and specific diagnosis of Apoptosis in the course of granulomatous inflammation in pulmonary sarcoid-
extrapulmonary tuberculosis by immunostaining of tissues and aspirates with osis. Eur Respir J. 1999;13(6):1329–1337.
anti-MPT64. Appl Immunohistochem Mol Morphol. 2017;25(4):282–288. 61. Strickland-Marmol LB, Fessler RG, Rojiani AM. Necrotizing sarcoid
31. Jeanes C, O’Grady J. Diagnosing tuberculosis in the 21st century - dawn of granulomatosis mimicking an intracranial neoplasm: clinicopathologic features
a genomics revolution? Int J Mycobacteriol. 2016;5(4):384–391. and review of the literature. Mod Pathol. 2000;13:909–913.
32. Farhi DC, Mason UG III, Horsburgh CR Jr. Pathologic findings in 62. Liebow AA. The J. Burns Amberson lecture–pulmonary angiitis and
disseminated Mycobacterium avium-intracellulare infection. A report of 11 cases. granulomatosis. Am Rev Respir Dis. 1973;108(1):1–18.
Am J Clin Pathol. 1986;85(1):67–72. 63. Rosen Y. Four decades of necrotizing sarcoid granulomatosis: what do we
33. Griffith DE, Aksamit T, Brown-Elliott BA, et al.; ATS Mycobacterial know now? Arch Pathol Lab Med. 2015;139(2):252–262.
Diseases Subcommittee; American Thoracic Society; Infectious Disease Society 64. Hsu RM, Connors AF Jr, Tomashefski JF Jr. Histologic, microbiologic, and
of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention clinical correlates of the diagnosis of sarcoidosis by transbronchial biopsy. Arch
of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007; Pathol Lab Med. 1996;120(4):364–368
175(4):367–416. 65. Churg AM, Green FHY. Occupational lung disease. In: Churg AM, Myers
34. Mukhopadhyay S. Role of histology in the diagnosis of infectious causes of JL, Tazelaar HD, Wright JL, eds. Thurlbeck’s Pathology of the Lung. 3rd ed. New
granulomatous lung disease. Curr Opin Pulm Med. 2011;17(3):189–196. York, NY: Thieme; 2005:769–862.
35. Dermawan JKT, Ghosh S, Keating MK, Gopalakrishna KV, Mukhopadhyay 66. Jones Williams W. Beryllium disease. Postgrad Med J. 1988; 64(753):511–
S. Candida pneumonia with severe clinical course, recovery with antifungal 516.
therapy and unusual pathologic findings: a case report. Medicine (Baltimore). 67. Freiman DG, Hardy HL. Beryllium disease. The relation of pulmonary
2018;97(2):e9650. pathology to clinical course and prognosis based on a study of 130 cases from the
36. Dermawan JK, Ghosh S, Mukhopadhyay S. Expanding the spectrum of U.S. beryllium case registry. Hum Pathol. 1970;1(1):25–44.
chronic necrotising (semi-invasive) aspergillosis: a series of eight cases presenting 68. Williams WJ. A histological study of the lungs in 52 cases of chronic
as radiologically solid lung nodules mimicking malignancy. Histopathology. beryllium disease. Br J Ind Med. 1958;15(2):84–91.
2020;76(5):685–697 69. Spagnolo P, Rossi G, Cavazza A, et al. Hypersensitivity pneumonitis: a
37. Procop GW, Pritt BS. Fungal infections. In: Pritt BS, Mathison BA, Milner comprehensive review. J Investig Allergol Clin Immunol. 2015;25(4):237–250.
DA, et al., eds. Atlas of Fundamental Infectious Disease Histopathology. 70. Miller R, Allen TC, Barrios RJ, et al. Hypersensitivity pneumonitis a
Northfield, Illinois: College of American Pathologists: 2018:91–148. perspective from members of the Pulmonary Pathology Society. Arch Pathol Lab
38. Scheutz AN, Pritt BS. Bacterial infections. In: Pritt BS, Mathison BA, Milner Med. 2018;142(1):120–126.
DA, et al., eds. Atlas of Fundamental Infectious Disease Histopathology. 71. Selman M, Buendı́a-Roldán I. Immunopathology, diagnosis, and
Northfield, Illinois: College of American Pathologists; 2018:19–90. management of hypersensitivity pneumonitis. Semin Respir Crit Care Med.
39. Rosen Y, Amorosa JK, Moon S, Cohen J, Lyons HA. Occurrence of lung 2012;335:543–554.
granulomas in patients with stage I sarcoidosis. AJR Am J Roentgenol. 1977; 72. Hariri LP, Mino-Kenudson M, Shea B, et al. Distinct histopathology of
129(6):1083–1085. acute onset or abrupt exacerbation of hypersensitivity pneumonitis. Hum Pathol.
40. Mitchell DN, Scadding JG, Heard BE, Hinson KF. Sarcoidosis: 2012;43(5):660–668.
histopathological definition and clinical diagnosis. J Clin Pathol. 1977;30(5): 73. Coleman A, Colby TV. Histologic diagnosis of extrinsic allergic alveolitis.
395–408. Am J Surg Pathol. 1988;12(7):514–518.
41. Sheffield EA. Pathology of sarcoidosis. Clin Chest Med. 1997;18(4):7 41– 74. Churg A, Bilawich A, Wright JL. Pathology of chronic hypersensitivity
54. pneumonitis. What is it? What are the diagnostic criteria? Why do we care? Arch
42. Rosen Y. Pathology of sarcoidosis. Semin Respir Crit Care Med. 2007; Pathol Lab Med. 2018;142(1):109–119.
28(1):36–52. 75. Kern RM, Singer JP, Koth L, et al. Lung transplantation for hypersensitivity
43. Rossi G, Cavazza A, Colby TV. Pathology of sarcoidosis. Clin Rev Allergy pneumonitis. Chest. 2015; 147(6):1558–1565.
Immunol. 2015;49(1):36–44. 76. Barrios RJ. Hypersensitivity pneumonitis: histopathology. Arch Pathol Lab
44. Ma Y, Gal A, Koss M. Reprint of: The pathology of pulmonary sarcoidosis: Med. 2008;132(2):199–203.
update. Semin Diagn Pathol. 2018;35(5):324–333. 77. Khoor A, Leslie KO, Tazelaar HD, Helmers RA, Colby TV. Diffuse
45. Statement on sarcoidosis. Joint Statement of the American Thoracic pulmonary disease caused by nontuberculous mycobacteria in immunocompe-
Society (ATS), the European Respiratory Society (ERS) and the World Association tent people (hot tub lung). Am J Clin Pathol. 2001;115(5):755–762.
of Sarcoidosis and Other Granulomatous Disorders (WASOG) adopted by the 78. Katzenstein ALA. Katzenstein and Askin’s Surgical Pathology of Non-
ATS Board of Directors and by the ERS Executive Committee, February 1999. Am Neoplastic Lung Disease. 4th ed. Philadelphia, PA: Elsevier; 2006.
J Respir Crit Care Med. 1999;160(2):736–755. 79. Gal AA, Salinas FF, Staton GW Jr. The clinical and pathological spectrum
46. Kieszko R, Krawczyk P, Michnar M, Chocholska S, Milanowski J. The yield of antineutrophil cytoplasmic autoantibody-related pulmonary disease. A
of endobronchial biopsy in pulmonary sarcoidosis: connection between comparison between perinuclear and cytoplasmic antineutrophil cytoplasmic
spirometric impairment and lymphocyte subpopulations in bronchoalveolar autoantibodies. Arch Pathol Lab Med. 1994;118(12):1209–1214.
lavage fluid. Respiration. 2004;71(1):72–76. 80. Gaudin PB, Askin FB, Falk RJ, Jennette JC. The pathologic spectrum of
47. Shorr AF, Torrington KG, Hnatiuk OW. Endobronchial biopsy for pulmonary lesions in patients with anti-neutrophil cytoplasmic autoantibodies
sarcoidosis: a prospective study. Chest. 2001;120(1):109–114. specific for anti-proteinase 3 and anti-myeloperoxidase. Am J Clin Pathol. 1995;
48. Koerner SK, Sakowitz AJ, Appelman RI, Becker NH, Schoenbaum SW. 104(1):7–16.
Transbronchial lung biopsy for the diagnosis of sarcoidosis. N Engl J Med. 1975; 81. Nguyen Y, Guillevin L. Eosinophilic granulomatosis with polyangiitis
293(6):268–270. (Churg-Strauss). Semin Respir Crit Care Med. 2018;39(4):471–481.
49. Koonitz CH, Joyner LR, Nelson RA. Transbronchial lung biopsy via the 82. Masi AT, Hunder GG, Lie JT, et al. The American College of Rheumatology
fiberoptic bronchoscope in sarcoidosis. Ann Intern Med. 1976;85(1):64–66. 1990 criteria for the classification of Churg-Strauss syndrome (allergic granulo-
50. Gilman MJ, Wang KP. Transbronchial lung biopsy in sarcoidosis. An matosis and angiitis). Arthritis Rheum. 1990;33(8):1094–1100.
approach to determine the optimal number of biopsies. Am Rev Respir Dis. 1980; 83. Katzenstein AL. Diagnostic features and differential diagnosis of Churg-
122(5):721–724. Strauss syndrome in the lung. A review. Am J Clin Pathol. 2000;114(5):767–772.

250 Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen
84. Koss MN, Antonovych T, Hochholzer L. Allergic granulomatosis (Churg- 99. Cohen Aubart F, Lhote R, Amoura A, et al. Drug-induced sarcoidosis: an
Strauss syndrome): pulmonary and renal morphologic findings. Am J Surg Pathol. overview of the WHO pharmacovigilance database. J Intern Med. 2020;288(3):
1981;5(1):21–28. 356–362.
85. Katzenstein AL, Liebow AA, Friedman PJ. Bronchocentric granulomatosis, 100. Chiriaco M, Salfa I, Di Matteo G, Rossi P, Finocchi A. Chronic
mucoid impaction, and hypersensitivity reactions to fungi. Am Rev Respir Dis. granulomatous disease: clinical, molecular, and therapeutic aspects. Pediatr
1975;111(4):497–537. Allergy Immunol. 2016;27(3):242–253.
86. Bosken CH, Myers JL, Greenberger PA, Katzenstein AL. Pathologic features 101. Moskaluk CA, Pogrebniak HW, Pass HI, Gallin JI, Travis WD. Surgical
of allergic bronchopulmonary aspergillosis. Am J Surg Pathol. 1988;12(3):216– pathology of the lung in chronic granulomatous disease. Am J Clin Pathol. 1994;
222. 102(5):684–691.
87. Myers JL, Katzenstein AL. Granulomatous infection mimicking broncho-
102. Cinetto F, Scarpa R, Rattazzi M, Agostini C. The broad spectrum of lung
centric granulomatosis. Am J Surg Pathol. 1986;10(5):317–22.
diseases in primary antibody deficiencies. Eur Respir Rev. 2018;27(149):180019.
88. Dail DH. Bronchial and transbronchial diseases. In Dail DH, Hammar SP,
eds. Pulmonary Pathology. 2nd ed. New York, NY: Springer-Verlag; 1994:79–119. 103. Prasse A, Kayser G, Warnatz K. Common variable immunodeficiency-
89. Mukhopadhyay S, Katzenstein AL. Pulmonary diseases due to aspiration of associated granulomatous and interstitial lung disease. Curr Opin Pulm Med.
food and other particulate matter: a clinicopathologic study of 59 cases 2013;19(5):503–509.
diagnosed on biopsy or resection specimens. Am J Surg Pathol. 2007;31(5): 104. Park JH, Levinson AI. Granulomatous-lymphocytic interstitial lung disease
752–759. (GLILD) in common variable immunodeficiency (CVID). Clin Immunol. 2010;
90. Tomashefski JF Jr, Hirsch CS. The pulmonary vascular lesions of 134(2):97–103.
intravenous drug abuse. Hum Pathol. 1980;11(2):133–145. 105. Hurst JR, Verma N, Lowe D, et al. British Lung Foundation/United
91. Sigdel S, Gemind JT, Tomashefski JF Jr. The Movat pentachrome stain as a Kingdom Primary Immunodeficiency Network consensus statement on the
means of identifying microcrystalline cellulose among other particulates found in definition, diagnosis, and management of granulomatous-lymphocytic interstitial
lung tissue. Arch Pathol Lab Med. 2011;135(2):249–254. lung disease in common variable immunodeficiency disorders. J Allergy Clin
92. McNearney T, Bajaj C, Boyars M, Cottingham J, Haque A. Total parenteral Immunol Pract. 2017;5(4):938–945
nutrition associated crystalline precipitates resulting in pulmonary artery 106. Larsen BT, Smith ML, Tazelaar HD, Yi ES, Churg A. GLILD revisited:
occlusions and alveolar granulomas. Dig Dis Sci. 2003;48(7):1352–1354. pulmonary pathology of common variable and selective iga immunodeficiency.
93. Hirche TO, Lambrecht E, Wagner TO. Crack-syndrome: the pulmonary Am J Surg Pathol. 2020;44(8):1073–1081.
complications of inhaled cocaine. A review a propos a case report. Pneumologie. 107. Brincker H. Sarcoid reactions in malignant tumours. Cancer Treat Rev.
2002;56(11):684–688. 1986;13(3):147–156.
94. Ciriaco P, Rossetti F, Carretta A, et at. Spontaneous pneumothorax in
108. Brincker H. The sarcoidosis-lymphoma syndrome. Br J Cancer 1986;54(3):
cocaine users. QJM. 2019;112(7):519–522.
467–473.
95. Newman LS. Metals that cause sarcoidosis. Semin Respir Infect. 1998;
109. de Charry F, Sadoune K, Sebban C, et al. Association of lymphoma and
13(3):212–220.
96. Gebbers JO, Burkhardt A, Tetzner C, Rüdiger HW, von Wichert P. ‘‘Hair- granulomatosis: a case series. Rev Med Interne. 2016;37:453–459.
spray lung.’’ Clinical and morphological findings. Schweiz Med Wochenschr. 110. Cohen PR, Kurzrock R. Sarcoidosis and malignancy. Clin Dermatol. 2007;
1980;110(16):610–615. 25(3):326–333.
97. Larsen BT, Chae JM, Dixit AS, Helmers RA. Methotrexate pneumonitis: 111. Pavic M, Debourdeau P, Vacelet V, Rousset H. Sarcoidosis and sarcoid
review of the literature and histopathological findings in nine patients. Am J Surg reactions in cancer. Rev Med Interne. 2008;29(1):39–45.
Pathol. 2019;43(10):1331–1340. 112. Butt S, Alzebdeh R, Kable TD, Soubani AO. Non-caseating granulomas in
98. Rambhia PH, Reichert B, Scott JF, et al. Immune checkpoint inhibitor- patients after the diagnosis of cancer: clinical characteristics and outcome.
induced sarcoidosis-like granulomas. Int J Clin Oncol. 2019;24(10):1171–1181. Sarcoidosis Vasc Diffuse Lung Dis. 2011;28(1):444–449.

Arch Pathol Lab Med—Vol 146, February 2022 Pathology of Granulomatous Pulmonary Diseases—Rosen 251

You might also like