Pneumonia




(1)
Institute of Pathology, Medical University Graz, Graz, Austria

 




8.1 Alveolar Pneumonias (Lobar and Bronchopneumonia)


The lung is constantly exposed to airborne infectious agents due to the large surface area of approximately 100 m2. Therefore pneumonia is one of the most common lung diseases. Understanding infection requires understanding the routes of infections, the way invading organisms infect epithelial cells, as well as defense mechanisms of the lung tissue acquired during evolution.

By the double arterial supply via pulmonary and bronchial arteries, neutrophil granulocytes or lymphocytes and monocytes can be directed into an area of infection rapidly. In addition the diameter of the pulmonary capillaries of approximately 5–6 μm requires adaptation of leukocytes and thus also slows their passage time, providing more time for contact with adhesion molecules expressed on endothelial cells, required for migration into the infected tissue [1].


Defense system:

The mucociliary escalator system can remove infectious organisms before they might act on the epithelia. The more viscous layer of mucus is at the surface, the more liquid layer at the ciliary site. Bacteria, for example, stick within this viscous mucus and can be transported toward the larynx. The cough reflex in addition helps to expel this material from the airways. An example how important this system works can be seen in patients with immotile cilia syndrome, where an inherent gene defect causes uncoordinated ciliary beating and results in defective clearance of mucus and subsequent recurrent infections [2, 3].


Innate immune system:

The innate immune system consists of complement activation (often via alternative pathway), surfactant apoproteins capable of bacterial inactivation, and the cellular constituents such as macrophages, granulocytes, and epithelial cells. Here we will briefly discuss this system. For more detailed information, the reader is referred to the vast amount of immunological reviews on this subject.

There are three known activation pathways for complement: the alternative, the classic, and the lectin pathway. Opsonization seems to be the most important function of complement C3. This leads to enhanced phagocytosis of bacteria. The system seems to be self-regulated as phagocytosis of apoptotic neutrophils by macrophages leads to less C3 activation and cytokine release by macrophages and consequently less inflammation [4].

Several surfactant apoproteins (SP) are produced by type II pneumocytes and secreted toward the alveolar surface. Two of them SPA and SPD are members of the collectin family proteins. At their C-terminal end, they have a lectin moiety, which is able to recognize bacterial oligosaccharides (galactosylceramide, glucosylceramide) present on the capsule of bacteria such as staphylococci. This binding causes aggregation and growth arrest of the bacteria and enhances phagocytosis by alveolar macrophages [5, 6].

Epithelial cells form a barrier for the entry of infectious organisms and thus protect the underlying mesenchymal structures, essential for lung function. Although many organisms have developed binding sites for respiratory epithelia, such as ICAM1 used by rhinoviruses, the epithelia have developed response mechanisms such as cytokine release, for example, proinflammatory interleukin 1β (IL1β), tumor necrosis factor α (TNFα), IL6, IL16, chemokines as IL8, macrophage inflammatory protein (MIP1α), RANTES, granulocyte-macrophage colony-stimulating factor (GMCSF), and others [7]. By the release of these mediators, neutrophils, macrophages, lymphocytes, and especially also cytotoxic T and NK cells are attracted and might initially already kill the invading organisms.


Monocytes/macrophages and granulocytes:

Macrophages are the primary source of defense against any type of infectious organism. Macrophages constantly patrol throughout the lung, ingesting every inhaled foreign material. Macrophages in contrast to monocytes live longer due to a genetic shift toward antiapoptosis by downregulating PTEN [8]. Macrophages also express Toll-like receptors (TLR2, TLR4) and CD14 and interact with SPA and CD44 to exert different functions such as release of antibacterial proteins/peptides [9]. Granulocytes interact with macrophages: if large amount of bacteria are inhaled, macrophages direct neutrophils to the site of infection, whereas small amounts of bacteria might be cleared by macrophages alone. Removal of apoptotic neutrophils requires macrophages, and this in turn decreases the inflammatory response and neutrophil influx [4]. Neutrophils are able to kill many phagocytosed bacteria by producing large amounts of oxygen radicals (superoxide anions) in their lysosomes and fusing them with the phagosomes. Neutrophils are produced in the bone marrow and released from there by cytokine stimuli. Once they enter the circulation, their apoptotic program is activated. They enter the infectious site using adhesion molecules on endothelia in due time and exert their function. This is facilitated by integrins and also other adhesins. Once within the interstitium, neutrophils move along gradients of chemokines and also acidic pH.

Eosinophils are specifically seen in parasitic infections. This is usually mediated by T lymphocytes and will be discussed below.


Adaptive immune system:

The adaptive immune system is a late invention in evolution. It requires different types of lymphocytes, such as B lymphocytes for an antibody-mediated reaction and T lymphocytes and NK cells for a direct cell-mediated toxic reaction. In addition this system also requires classical dendritic cells for antigen processing and antigen presentation; these cells get in contact with invading organisms at the site of first contact or in lymph nodes.

Within the bronchial mucosa, IgA-producing B lymphocytes are found. Secreted IgA is a complex, where two molecules of IgA are joined by a secretory component. In combination with other molecules such as albumin, transferrin, ceruloplasmin, and IgG, these are antioxidants and have a mucosal defense function. These molecules are increased secreted in lung injury and inflammation [10]. In cigarette smokers the immune barrier function is impaired by a decreased release of secretory component, which in turn also decreases the transcytosis of IgA [11]. One of the most important functions of IgA secreted at the lining fluid is opsonization of different bacteria [12].

Different types of dendritic cells can be found in the bronchial and alveolar system such as classical, follicular, Langerhans, and interdigitating reticulum cells. These cells are thought to play a role in antigen uptake and processing. Dendritic cells also direct the type of immune reaction by interacting with different Toll receptors. Under inflammatory conditions a Thelper1 response is favored, whereas Thelper2 responses require another mechanism [13]. Dendritic cells, for example, confronted with mycobacteria will induce differentiation of CD4+ to CD4+17+ cells and also induce Toll receptor 9 expression resulting in granuloma formation [14]. Some subpopulations of dendritic cells can induce immune tolerance and exhaustion, which might play a role in certain diseases, but this will be discussed in another chapter.


8.1.1 Clinical Symptoms of Pneumonias


There are some key features characterizing pneumonias, such as fever, cough, and fatigue. Fever will give some information about possible organisms: above 39.5 °C most likely this is caused by a viral infection, whereas bacterial pneumonias present with temperatures between 38 and 39 °C. Cough can be productive with either serous or purulent expectoration. Laboratory evaluation will show inflammatory parameters, such as leukocytosis, etc.

Radiologically the lung will show ground glass opacities and consolidations, depending on the age of the inflammatory infiltrate. Clinically pneumonias are separated into typical and atypical pneumonia. Atypical pneumonia can have different meanings, either an atypical infiltration pattern on CT scans or atypical presentation with rare infectious organisms.


8.1.2 Alveolar Pneumonias (Bronchopneumonia and Lobar Pneumonia; Adult and Childhood)


Although infectious pneumonia is a common disease, biopsies and surgical resections are rarely seen in pathologic practice. Most of these cases are diagnosed clinically and treated accordingly by antibiotics. If biopsied or resected, these cases usually turn out as unusual pneumonia caused by unusual organisms. Pneumonias are commonly seen at autopsy. The evaluation of infectious organisms will be discussed after the granulomatous pneumonias.


8.1.2.1 Gross Morphology


Pneumonia develops in stages, starting with hemorrhage. The lung is dark red, consistency is firm, and on the cut surface, there is some granularity seen, corresponding to fibrin cloths out of the alveoli (Fig. 8.1). In the next stage, the color of the lung changes to gray and grayish yellow. This is induced by the influx of leukocytes, dying of leukocytes, and release of lipid substances (Fig. 8.2). The consistency of the lung is comparable to liver tissue, hence the old name “hepatization.” Finally in the best scenario, the exudate is reabsorbed, the alveoli are filled with air, and the lung changes back to normal (lysis). Most often these classical stages are not anymore seen, because pneumonia is immediately treated with antibiotics and therefore do not develop into the yellow “hepatization” stage but resolve out of the gray-red one. However, complications of bronchopneumonia can be seen such as abscess formation (Fig. 8.3) and pneumonia with infarcts due to infectious vasculitis (see below; Fig. 8.4).

A299455_1_En_8_Fig1_HTML.jpg


Fig. 8.1
Early pneumonia with hemorrhage; autopsy specimen


A299455_1_En_8_Fig2_HTML.jpg


Fig. 8.2
Macroscopy of purulent bronchopneumonia. At lower right there is abscess formation; the pleura shows purulent pleuritis


A299455_1_En_8_Fig3_HTML.jpg


Fig. 8.3
Purulent pneumonia with abscess formation


A299455_1_En_8_Fig4_HTML.jpg


Fig. 8.4
Purulent pneumonia with multiple infarcts due to infectious vasculitis

Histology and development of bronchopneumonia: Bronchopneumonia in the initial stages starts with an influx of macrophages from the interstitial cell pool as well as from the blood vessels (monocytoid cells; Fig. 8.5). Capillaries are widened (hyperemia) and the endothelial gaps are opened. Fluid from the blood enters into the alveolar spaces (inflammatory edema) and proteins start to coagulate (fibrin cloths). This initial stage is followed by an entry of red blood cells, which undergo lysis, contributing to fibrinogenesis. In this stage fibrin nets are seen mixed with red blood cells, scattered macrophages, and neutrophils. This corresponds to the macroscopic picture of hemorrhagic pneumonia (dark red cut surface, heavy lung, edematous fluid rinsing from the cut surface). After 1 day dense infiltrations by neutrophils appear, mixed with fibrin nets completely filling the alveolar spaces (Fig. 8.6). Capillaries are still hyperemic and widened. Macroscopically the cut surface changes to a gray-red color, due to the massive infiltration by granulocytes. Since the alveoli are completely filled by cells and fibrin, the consistency is similar to liver (hepatic consolidation or hepatization). Granulocytes ingesting and degrading bacteria also die because of liberation of toxic lysosomal enzymes accumulate lipids within their cytoplasm, which macroscopically gives the cut surface a yellow tone (usually by day 2–3; Fig. 8.7). After 6–7 days clearance of the alveoli starts: neutrophils have degraded the bacteria, macrophages clear the debris from dying neutrophils, fibrin is lysed by the enzymes from macrophages and granulocytes, and finally the alveoli are filled by air again. Under normal condition the pneumonia resolves within 10–14 days without remnants of the infectious episode.

A299455_1_En_8_Fig5_HTML.jpg


Fig. 8.5
Early bronchopneumonia with influx of macrophages into the alveolar lumen. H&E, bar 20 μm


A299455_1_En_8_Fig6_HTML.jpg


Fig. 8.6
Full blown bronchopneumonia. There is necrosis of the bronchial mucosa and dense infiltration of the bronchial wall and the alveolar tissue by neutrophils. H&E, bar 200 μm


A299455_1_En_8_Fig7_HTML.jpg


Fig. 8.7
Purulent bronchopneumonia due to bacterial infection; H&E, bar 100 μm; inset gram stain of the same area showing gram-positive cocci. Gram, bar 5 μm

If for several reasons no resolution occurs, acute bronchopneumonia will undergo organization. The resulting morphologic picture is organizing pneumonia (see below).


8.1.2.2 Variants of Bronchopneumonias (Purulent Pneumonia (PN))


Lobar pneumonia is characterized by a uniform inflammatory infiltration of the lung. Bacteria are distributed early on by an edema within a whole lobe or several segments. The pneumonia therefore will show the same timely development in all areas involved. This means that the developmental stage of the inflammation is identical in each area investigated. Most often biopsies or resection specimen will present with dense neutrophilic infiltrations and fibrin cloths filling the alveoli. Bacteria can easily be identified using a Gram stain (Fig. 8.7).

Bronchopneumonia in contrast will show different developmental stages in different areas, depending on the amount of bacteria present in a given segment. This will result in a colorful picture on macroscopy with dark red, grayish, and even yellowish areas and the same on histology: areas of hemorrhage, areas of mixed fibrinous and granulocytic infiltrations, areas of granulocytic debris, and macrophage infiltration.

Pneumonias with abscess formation are another form of bronchopneumonia, which most often is seen in infections with certain species of bacteria. These abscesses are based on localized necrosis, either directly induced by the bacteria or by an interaction of bacteria with the coagulation system.


8.1.3 Diffuse Alveolar Damage (DAD) and Acute Interstitial Pneumonia


Clinically acute interstitial pneumonia (AIP, also adult respiratory distress syndrome (ARDS)) is characterized by an acute onset of severe hypoxia, with the radiological appearance of white lung. Histologically there is edema and fibrinous exudate, widened edematous alveolar septa (see also below acute fibrinous pneumonia). Later on hyaline membranes are formed – this was called diffuse alveolar damage (DAD) (Fig. 8.8). Depending on the cause of DAD, scattered neutrophilic and/or eosinophilic granulocytes can be found in bacterial, toxic, or drug-induced DAD, or few lymphocytes are seen in viral and rickettsia infections, respectively [15, 16]. Inflammatory infiltrates may be even absent such as in various kinds of shock. Rarely cases of “idiopathic AIP” have been reported. Probably some of these cases represent cases of undiagnosed SLE or drug toxicity. In the author’s experience in all cases sent for consultation and primarily labeled as idiopathic DAD, an etiology could finally be established. So it might be questioned if idiopathic DAD does exist [17].

A299455_1_En_8_Fig8_HTML.gif


Fig. 8.8
Diffuse alveolar damage (DAD)/acute interstitial pneumonia in this case induced by Puumala virus. There is edema, mild infiltration by lymphocytes, and development of hyaline membranes. H&E, bar 100 μm

Hamman-Rich described an interstitial pneumonia with fulminant course leading to death in their six cases within 6 months. In the author’s description, there was no hyaline membrane mentioned but a proliferation of fibroblasts. Since the tissues from these cases were all lost, this disease cannot be reconstructed and remains an enigma [18].

The sequence of events in DAD is largely dependent on the cause: Toxic metabolites of drugs or released collagenase and elastase from necrotizing pancreatitis will cause endothelial damage, followed by leakage of the small peripheral blood vessels. This causes edema, followed by pneumocyte cell death due to hypoxia. Serum proteins will pass into the alveolar lumina, coagulate there, and by the breathing movements are compressed into hyaline membranes (Figs. 8.9 and 8.10). In case of airborne disease, e.g., infection or inhaled toxins, pneumocytes type I die followed by type II. Due to the lack of surfactant lipids, the alveoli collapse. The basement membrane is either preserved or also destroyed (especially in viral infection). This again causes leakage of capillaries, edema with/without bleeding, protein extravasation into the alveoli, and finally formation of hyaline membranes.

A299455_1_En_8_Fig9_HTML.jpg


Fig. 8.9
Drug-induced DAD (neuroleptic). In (a) areas of interstitial infiltrations by lymphocytes and histiocytes are seen, as well as fibrin cloths in alveoli. There is also alveolar hemorrhage. In (b) there is endothelial damage and fibrin cloth, which points to the etiology (toxic substance from circulation). In (c) fibrin cloths are seen within the septa as well as outside in alveoli, and in (d) there are hyaline membranes already in organization. H&E, bar 100 μm, and 20 μm in (bd)


A299455_1_En_8_Fig10_HTML.jpg


Fig. 8.10
DAD in cardiac shock. There is congestion of blood in the capillaries, also hyaline membranes have developed (a); in (b) there is another typical feature of shock, namely, intravascular fibrin clothing. H&E, bars 50 μm and 20 μm, respectively

The lethality of DAD is still high despite improvements, which have been made in the past decade. In some cases the progression of the disease might be blocked by antiprotease treatment [19]. In more recent time extracorporeal oxygenation or NO treatment has shown some benefit. If the patient survives the acute phase, DAD will be organized, which is essentially an organizing pneumonia, by some authors also labeled organizing DAD: granulation tissue grows into the alveoli and hyaline membranes are incorporated into the plugs. Remnants of hyaline membranes can be demonstrated several weeks after the initial injury (Figs. 8.11, 8.12, and 8.13). If a tissue biopsy or an autopsy specimen is available early on in the course of the disease, the etiology might be elaborated: in viral infection inclusion bodies can be seen, which can present either as nice large inclusion bodies (CMV, RSV) or by red-violet stained nucleic acids forming ill-defined speckles in nuclei and/or cytoplasm (adenovirus) [20]. This is followed by atypical proliferation and transformation of pneumocytes type II (Fig. 8.14). Typically the infected cell shows enlargement, an atypical large bizarre nucleus, and an accentuated nuclear membrane due to increased nucleic acid traffic induced by the virus. These cellular features can last for several months. In contrast to atypical pneumocyte hyperplasia (AAH), these atypical cells are singles and do not form a continuous layer along the alveolar wall. Rickettsia infection results in less pronounced proliferation of pneumocytes. In shock and drug-induced DAD, the endothelia will undergo apoptosis and necrosis, and fibrin cloths might be seen in capillaries (Fig. 8.10). In these cases the alveolar septa are widened and edematous. Inflammatory cells are scarce or absent. In later stages of drug pneumonia, scattered eosinophils are encountered – their function being completely unknown.

A299455_1_En_8_Fig11_HTML.jpg


Fig. 8.11
DAD in organization. Macroscopic picture showing areas of consolidation. Not much normal lung tissue is left. On the cut surface, numerous tiny little nodules are seen, which represent granulation tissue


A299455_1_En_8_Fig12_HTML.jpg


Fig. 8.12
DAD in organization, this is essentially an organizing pneumonia. Hyaline membranes are still visible but organized by granulation tissue, which grows within alveoli and finally will fill the lumina


A299455_1_En_8_Fig13_HTML.jpg


Fig. 8.13
DAD in organization. In this case the granulation tissue has filled the alveoli, leaving only slit-like spaces. Remnants of hyaline membranes are still visible. H&E, bar 20 μm


A299455_1_En_8_Fig14_HTML.gif


Fig. 8.14
DAD in viral infection; in (a) proven infection by adenovirus type 5 (infected cell arrow); there are no inclusion bodies, because these viruses form tiny pseudocrystalline intracytoplasmic structures. H&E, bar 20 μm, in (b) cytomegalovirus infection combined with Pneumocystis jirovecii. Note the characteristic large intranuclear inclusion bodies in CMV. Giemsa, ×630




  • What are the characteristics of DAD?



    • Edematous fluid accumulation in alveoli and in the interstitium (depending on the time course)


    • Fibrin cloths in alveoli with/without hyaline membranes


    • Scarce inflammatory infiltrates (neutrophils and/or lymphocytes, etiology dependent)


    • Minor diagnostic but etiologically important features are damage of pneumocytes, endothelial cells, fibrin thrombi in small blood vessels, and regeneration ± atypia

Acute fibrinous and organizing pneumonia (AFOP) was recently described as a variant of DAD: the dominant pattern is accumulation of intra-alveolar fibrin and concomitant organizing pneumonia [21]. Also pneumocyte type II hyperplasia, edema, and inflammatory infiltrates were described. Clinically the symptoms were identical to ARDS/AIP. The main difference stated by the author was the absence of hyaline membranes and the presence of fibrin cloths. The underlying causes were similar to classical DAD/AIP, so the author concluded that this might be a variant of DAD. However, some aspects have never been clarified: Fibrin exudation and clothing is seen early in DAD (see also Fig. 8.9), so the earliest phase of DAD does not present with hyaline membranes – these are formed later on due to respiration, which compresses fibrin into hyaline membranes. Rarely DAD might also present with a multifocal pattern, which includes a timely heterogeneity: acute fibrinous exudation in one, organizing DAD in another area [22]. Within the underlying cause, similar diseases as in DAD were found, including rare cases of acute hypersensitivity pneumonia [21, 23].


8.1.4 Lymphocytic Interstitial Pneumonia (LIP)


LIP almost vanished from the literature in the last 5 years. The major problem is the separation from NSIP. When NSIP was described, it was never clearly separated from LIP [24]. When comparing my own cases and reports from the literature, it becomes evident that differences do exist: in LIP the lymphocytic and plasmacytic infiltration is dense, hyperplasia of the bronchus-associated lymphoid tissue (BALT) is common, and within lymph follicles germinal centers are usually present [24]. The infiltration in LIP is more diffuse, architectural distortion is common, and scarring does occur. Histiocytic and monocytic cellular infiltrations are much less pronounced compared to NSIP. Lymphoepithelial lesions do occur similar to lymphomas, in some entities aggressively infiltrating and destroying the epithelium; in other cases no epithelial disruption does occur. In contrast to NSIP, the architecture of the peripheral lung is remodeled, especially in later stages (Fig. 8.15).

A299455_1_En_8_Fig15_HTML.gif


Fig. 8.15
Lymphocytic interstitial pneumonia. Dense lymphocytic infiltrates with ill-formed primary lymph follicles are seen in (a). In (b) the infiltration was dominated by CD4+ lymphocytes ruling out hypersensitivity pneumonia in this case. In (c) the infiltrate is composed of lymphocytes, plasma cells. Focally fibrosis has started with proliferating myofibroblasts. H&E, bar 50 μm (a) and ×200 (c), immunohistochemistry for CD4, bar 100 μm in (b)

The clinical presentation depends on the underlying disease, and the CT scan usually shows ground glass opacities, in subacute and chronic stages, and also areas of fibrosis.

On gross morphology scattered areas of consolidations are seen.

Within the etiologic spectrum, similar diseases are found as in NSIP: autoimmune diseases especially collagen vascular diseases, allergic diseases as extrinsic allergic alveolitis/hypersensitivity pneumonia (EAA/HP) (acute and subacute), allergic drug reactions, HIV infection, and in children different types of immunodeficiency (T-cell defect, NK-cell defect). The most important differential diagnoses, however, are extranodal marginal zone lymphoma of MALT/BALT type and lymphomatoid granulomatosis type I. In all cases the clonality has to be evaluated and a lymphoma needs to be excluded by proof of multiclonality.

LYG type I can be difficult to separate: large blasts are rare and can be obscured within a dense infiltrate by small lymphocytes. The lymphocytic infiltration is polyclonal, so this does not help in the separation. Therefore a search for EBV-positive blasts is essential. Also it is important to exclude posttransplant lymphoproliferative disease [25], which can present in a similar pattern (large lymphoid cells usually EBV positive). However, it should be reminded that some of the autoimmune diseases have a high propensity of developing non-Hodgkin lymphomas later in the course [26]. Within the autoimmune diseases, Sjøgren’s disease most often presents with LIP pattern [27, 28].



  • What are the morphologic characteristics?



    • Diffuse dense lymphoplasmocytic infiltrates in alveolar septa and bronchial/bronchiolar walls. In some cases the lymphocytic infiltration can form concentric rows encasing capillaries and venules.


    • Hyperplasia of BALT with well-formed follicular centers.


    • Focal fibrosis and scarring with distortion of the peripheral lung architecture.


    • Lymphoepithelial lesions.


    • Eccentric sclerosis of vessel walls with narrowing of lumina: this is usually a sign of deposition of immune complexes in the vessel walls and should prompt the search for diseases associated with the production of autoantibodies, such as Sjøgren’s disease and systemic sclerosis.


8.1.4.1 Immunohistochemistry


Every case of LIP needs an evaluation for clonality using antibodies or in situ hybridization for kappa and lambda. As soon as a lymphoma is ruled out, further evaluation can be directed toward the underlying etiology. In a first step, lymphocytes should be subtyped into B and T lymphocytes and furthermore into CD4+ and CD8+ T lymphocytes. An evaluation of regulatory T cells using FOXP3 antibodies will also help in sorting the etiology. EAA/HP is dominated by CD8+ T lymphocytes at least in acute stages, whereas in autoimmune diseases the lymphocytic infiltrate is usually mixed. The absence of Treg cells can be of help for the diagnosis of some of the autoimmune diseases, such as rheumatoid arthritis.


8.1.5 Giant Cell Interstitial Pneumonia (GIP; See Also Under Pneumoconiosis)


GIP has a quite narrow etiologic spectrum either being caused by hard metal dust or by viral infection. The former will be discussed later. Several viruses can cause GIP, the classical one being measles virus. However, in contrast to pneumoconiosis in infections, the giant cells are mixed epithelial as well as macrophagocytic. The epithelial giant cells (Hecht cells) are transformed pneumocytes type II in whom nuclear division was not followed by cell division giving rise to multinucleation [29]. The additional features are identical to DAD as described above. Especially within the epithelial cells, viral inclusion bodies can be found (Fig. 8.16). Besides measles, also respiratory syncytial virus (RSV) can present with this picture predominantly in children [30].

A299455_1_En_8_Fig16_HTML.jpg


Fig. 8.16
Giant cell interstitial pneumonia (GIP) here in a 2-year-old girl, which died due to measles pneumonia. There is DAD with hyaline membrane formation, but in addition there are multiple multinucleated giant cells, which show intranuclear violet-red viral inclusion bodies. H&E, ×400

Alveolar and interstitial pneumonias can be induced by a wide variety of organisms. According to that they can be classified as bacterial, viral, rickettsia, or parasitic. Parasitoses will be covered in eosinophilic pneumonias (Chap. 10).

Infectious pneumonias in childhood are quite common but are rarely biopsied. There are some differences in so far as the density of leukocytic infiltrations is much less compared to the adult form.

Opportunistic infections as part of the infectious pneumonias in immunocompromised patients will be mentioned in the tables under the different organisms and in the chapter on transplantation pathology. Disorders related to therapeutic intervention, chemotherapeutic drug, and radiation injury will be discussed in toxic reaction due to drugs and inhalation.


8.1.6 The Infectious Organisms


Bacterial pneumonias are most often purulent; the dominant inflammatory cell is the neutrophil. In early stages the infiltration starts with macrophages and fibrin exudation followed by infiltration by neutrophils. Abscess formation is common; cavitation is induced by some bacteria and most likely is induced by vasculitis and thrombosis. A few bacteria cause DAD and fibrinous pneumonia, other lymphocytic pneumonia – these tissue reactions can point to the underlying type of infection (Table 8.1). A scattered type of neutrophilic infiltration is seen in some infections such as Nocardia or Legionella pneumonia (Fig. 8.17) and a mixed infiltration of leukocytes but dominated by macrophages seen in such rare bacterial infections as listeriosis (Fig. 8.18, Table 8.1).

A299455_1_En_8_Fig17_HTML.jpg


Fig. 8.17
(a) Bacterial pneumonia with scattered nodular aggregates of neutrophils. This should prompt one for special stains such as silver stains and modified acid-fast stains. (b) The infectious organism in this case was identified as Nocardia asteroides. (a) H&E, ×50, (b) Fite stain, bar 10 μm


A299455_1_En_8_Fig18_HTML.jpg


Fig. 8.18
Acute bacterial pneumonia with unusual features. There are histiocytic granulomas in the bronchial wall extending into the lumen, a dense macrophagocytic infiltration in alveoli, and a mixture of lymphocytic and neutrophilic infiltrations within the alveolar septa and bronchial wall. Special stains and culture identified the organisms as Listeria monocytogenes. H&E, bar 50 μm



Table 8.1
Gram-negative and gram-positive bacteria and types of pneumonias


















































































































Type of bacterium

Tissue reaction

Proof by

Children/adult

Actinomyces israelii and other subspec.

Abszeding PN, necrotizing histiocytic/epithelioid granulomatous PN

Pos. Gram or Brown-Brenn stain

No/yes

Bacillus anthracis

DAD, hemorrhage, necrotizing purulent PN

IHC, culture

No/yes wool sorters disease

Chlamydia pneumoniae, Chlamydophila psittaci

Lymphocytic and eosinophilic bronchitis, DAD, LIP

IHC, ISH, PCR (two forms: EB and RB

Yes/no

Corynebacterium diphtheriae

Pseudomembranous bronchitis, purulent PN

Gram pos

Yes/yes

Haemophilus influenzae

Purulent PN

Gram neg, methylene blue, IHC

Yes/yes

Klebsiella pneumoniae

Abszeding necrotizing purulent PN (lobar or lobular)

Gram neg, culture

Yes/yes

Legionella pneumophila

Fibrinous, hemorrhagic, necrotizing purulent PN

Warthin-Starry, Brown-Hopps, EM, IHC

No/yes

Listeria monocytogenes

DAD (transplacental transmission), purulent PN

Gram pos

Yes/no

Burkholderia pseudomallei, B. cepacia

Abszeding purulent PN, histiocytic granulomatous PN

Gram neg

Rarely/rarely (opportunistic)

Mycobacterium tuberculosis complex (see also below)

Granulomatous PN, necrotizing PN

ZN, RA, IHC, PCR

Yes/yes

Non-tuberculosis Mycobacteria (MOTT)

Granulomatous PN

ZN, RA, IHC, PCR

Yes/yes

Mycoplasma pneumoniae

Lymphocytic necrotizing bronchiolitis, LIP, DAD

IHC, PCR

No/yes

Nocardia asteroides

Purulent absceding PN

Modified ZN, GMS, Brown-Brenn (Gram pos), PCR

Rare/yes

Pseudomonas aeruginosa

Hemorrhagic PN, absceding purulent PN. Combined purulent vasculitis and cavitation

Gram neg, Brown-Hopps, culture

Rare/yes

Rhodococcus equi

Absceding PN, cavitation

PAS, GMS, Brown-Hopps (Gram pos)

No/yes (opportunistic, HIV)

Staphylococcus aureus

Purulent PN

Gram pos

No/yes

Streptococcus pneumoniae, S. viridans *

Purulent PN, DAD (infants), *abscesses

Gram pos

Yes/yes

Treponema pallidum

Epithelioid and neutrophilic granulomatous PN with abscesses and vasculitis

Warthin-Starry
 

Francisella tularensis

DAD with neutrophils and macrophages, later epithelioid cell granulomas, necrosis, cavitation

Gram neg, Brown-Hopps, Warthin-Starry, IHC

No/yes

Bordetella pertussis

Necrotizing bronchitis, bronchiolitis, peribronchiolar purulent PN

Gram neg, Giemsa, PCR

Yes/no


*GRAM gram stain, GMS Grocott methenamine silver impregnation, ZN Ziehl-Neelsen stain, RA rhodamine-auramine stain, PAS periodic acid-Schiff reaction, IHC immunohistochemistry, PCR polymerase chain reaction, EM electron microscopy, EB elementary body, RB reticulate body

Fungal pneumonias are caused by a variety of fungal organisms. Most often fungal infection does not proceed into infections of deep organs, but stay confined to the skin, oral cavity, or the upper respiratory tract. In immunocompromised patients or in infants, however, fungal infections can cause lethal widespread multiorgan infections (Figs. 8.19 and 8.20). Since many of the fungi have developed some capsular structures and also can undergo different developmental stages, the host tissue often needs to develop different strategies to keep the infection under control. In the normal host, fungi are usually controlled by an influx of neutrophils, which are capable of eliminating the fungi before they can cause pneumonia. In conditions where the fungi cannot be controlled, such as in bronchiectasis, the lung encases the infection by granulation tissue, starting as an organizing pneumonia, and later on a fibrous capsule separates the infectious focus from the normal lung – a mycetoma has been formed (Fig. 8.21). Normally there is a steady-state situation, i.e., no invasion of the fungus in deep areas of the lung occur, but the lung cannot get rid of the fungus. However, there are rare conditions where invasion does occur and a chronic slowly progressing pneumonia develops – called chronic necrotizing mycosis. A few fungi pathogenic in humans can cause life-threatening infections: an example is mucormycosis. Again infection most often occurs in immunocompromised patients. The patients develop cough, occasionally mild hemorrhage, fever and shortness of breath is common. The major problem is that this fungus does not respond to many antifungal drugs therefore amphotericin B is applied, which has many toxic side effects. Pneumonia in Mucor infection presents with an infiltration of macrophages and neutrophils, necrosis is widespread, pleura is often involved, or the infection can even enter the pleural cavity (Fig. 8.22, Table 8.2).

A299455_1_En_8_Fig19_HTML.jpg


Fig. 8.19
(a) Purulent pneumonia due to fungal infection in a child being treated for leukemia. Note that the hyphae have already reached the blood vessels, which is a risk for developing sepsis. Although the size of the hyphae, the 45° angle of growth, and the septation would favor an Aspergillus type of fungus, be aware that many other fungi can look alike. In (b) the fungus could be identified as Aspergillus niger, due to the presence of conidia. H&E, ×200, bar 20 μm


A299455_1_En_8_Fig20_HTML.jpg


Fig. 8.20
Infection in an immunocompromised patient treated with high-dose corticosteroids. Within the alveoli there is a foamy eosinophilic material, which is suggestive for Pneumocystis infection. In the inset Pneumocystis jirovecii is demonstrated by Grocott methenamine silver stain. H&E, ×200, Grocott, ×400


A299455_1_En_8_Fig21_HTML.jpg


Fig. 8.21
Mycetoma in a preformed bronchiectasis. Overview shows necrosis and a dense infiltrate in the wall of this bronchus. In the inset numerous hyphae are shown and the neutrophilic reaction within the bronchial wall. H&E, ×12.5 and 100, respectively


A299455_1_En_8_Fig22_HTML.jpg


Fig. 8.22
Mucormycosis here with widespread necrosis and nuclear debris from leukocytes. The organisms can be seen on H&E (left side) but better by silver impregnation (right side). H&E, bar 50 μm, Grocott methenamine, bar 50 μm



Table 8.2
Fungal organisms causing pneumonias






















































Type of fungus

Tissue reaction

Proof by

Children/adult

Aspergillus fumigatus, A. flavus, A. niger, other spec.

BCG, necrotizing bronchitis, mycetoma, chronic necrotizing PN, purulent PN with vasculitis

GMS, PAS, PCR

Yes/yes

Mucor 5 species

Necrotizing purulent PN, pleural involvement common

GMS, PCR

Yes/yes

Blastomyces dermatiditis

Purulent PN with abscesses, epithelioid granulomatous PN

GMS, IHC

No/yes

Candida species

Purulent PN, focal abscess

PAS, GMS, calcofluor-white, IHC

Yes/yes; immunocompromised

Coccidioides immitis

Purulent PN with microabscesses, necrotizing epithelioid granulomatous PN

GMS, IHC, ISH,

No/yes

Cryptococcus neoformans

Necrotizing epithelioid granulomatous PN

GMS, PAS, Fontana-Masson, IHC, Mucicarmine

No/yes

Histoplasma capsulatum,

Necrotizing epithelioid or histiocytic, granulomatous PN

GMS, Wright-Giemsa, IHC

Yes/yes

Paracoccidioides brasiliensis

Necrotizing epithelioid granulomatous PN, may be mixed with purulent PN

GMS, IHC

Yes/yes


GMS Grocott methenamine silver impregnation, PAS periodic acid-Schiff reaction, IHC immunohistochemistry, PCR polymerase chain reaction

Finally the reaction of the lung tissue against some specific forms of fungi can also be granulomatous. This reaction can be an innate immune reaction with histiocytes, macrophages, and foreign body giant cells or develop into a specific immune reaction with lymphocytes and epithelioid granuloma formation. However, this specific immune reaction depends on a functioning non-impaired immune system capable of producing different types of T lymphocytes (see below). Many fungi exhibit an angioinvasive growth behavior, i.e., their hyphae will grow toward arteries and veins directed by increase of pO2 and immediately will invade through the vessels wall, resulting in sepsis. There exist also an allergic mycosis, called allergic bronchopulmonary mycosis (ABPA, ABPM), which is based on a sensitization against fungal proteins; this will be discussed in Chap. 10.

Respirotropic viruses and Rickettsia cause viral and rickettsial pneumonias. One of the most common tissue reactions is DAD with hyaline membranes. In virus infections only scattered lymphocytes are seen in tissue sections, but in BAL there can be a lymphocytosis with up to 30 % of lymphocytes, predominantly CD8+ ones. Some viruses such as influenza type A strains can destroy the basal lamina of the epithelial layer and the capillaries by their enzymes. In these cases diffuse hemorrhage is seen with bleeding from capillaries, giving the macroscopic surface of the mucosa a dark red color. The distribution of inflammatory changes is also important: influenza virus usually causes trachea-broncho-pneumonia, whereas adenovirus is more likely causing bronchiolo-pneumonia. In cases of less virulent types of strains of viruses, a lymphocytic interstitial pneumonia can be seen (Figs. 8.23, 8.24, and 8.25). As a rule one should always try to find viral inclusion bodies. They can be prominent and easily seen as in CMV or HSV infection, whereas in adenovirus infection this can be difficult, because of intracytoplasmic bodies. Since the virions are very small and invisible, the package is ill defined. Viral inclusion bodies are stained violet red due to their high content of either DNA or RNA, and viral inclusions change the internal structure of a nucleus: the nuclear membrane is less sharp, and the chromatin structure is blurred.

A299455_1_En_8_Fig23_HTML.jpg


Fig. 8.23
Adenovirus-induced pneumonia. (a) There are many transformed pneumocytes with large nuclei. A few show dark stained nuclei and a red-violet cytoplasm. This should prompt further evaluation for viruses. (b) Immunohistochemistry for adenovirus showing many infected cells with granular cytoplasmic inclusion bodies. H&E, bar 10 μm; Immunohistochemistry, bar 20 μm pneumonia.


A299455_1_En_8_Fig24_HTML.jpg


Fig. 8.24
Hantavirus-induced pneumonia. There is edema, mild lymphocytic infiltration, only few pneumocytes show enlargement of nuclei, and abnormal chromatin pattern. H&E, ×250 (Courtesy of Prof. Walker, Galveston)


A299455_1_En_8_Fig25_HTML.jpg


Fig. 8.25
Pneumonia induced by Chlamydia trachomatis. The lung tissue is infiltrated by scattered lymphocytes, the pneumocytes II show abnormal nuclei some with blurred contours, and a red-violet cytoplasm. By immunohistochemistry the Chlamydia infection was proven. H&E, bar 50 μm, Immunohistochemistry, bar 20 μm


8.1.6.1 HIV Infection and the Lung


Clinically early pulmonary involvement appears as interstitial infiltration with progression to nodular tumor masses obliterating the lung. As with other viral infections, mild diffuse alveolar damage to frank interstitial fibrosis is the prominent finding [31]. However, due to the specific attack of the virus toward CD4+ lymphocytes, concomitant infections are common. This also will change the histology of HIV-induced pneumonia. There can be an overlay by Pneumocystis jirovecii or cytomegalovirus pneumonia, a lymphoid interstitial pneumonia, and a desquamative interstitial pneumonia [32]. Children as well as adults can be involved. Early interstitial fibrosis and even complete resolution of the pulmonary changes can be seen early on in the disease development. Kaposi’s sarcoma as a consequence of long-standing HIV infection is one of the most serious complications in these patients (this will be discussed in the tumor chapter) [33] (Table 8.3).


Table 8.3
Virus and Rickettsia, causing pneumonia















































































Type of virus

Tissue reaction

Proof by

Children/adult

Adenovirus

Hemorrhagic PN, DAD

IHC or ISH

Yes/yes

Cytomegalovirus

DAD, hemorrhagic PN

H&E, IHC, ISH

Yes/rare (AIDS, immunocompromised)

Echovirus

Hemorrhagic PN, DAD

IHC, ISH

Yes/no

Epstein-Barr virus

Mild lymphocytic PN

IHC, ISH

Yes/no

Hantavirus

Hemorrhagic PN

ISH, PCR

No/yes

Herpes simplex virus

DAD, hemorrhagic PN with necrosis

ISH, PCR, IHC

Yes/yes

Influenza/parainfluenza virus

DAD

ISH, PCR, IHC, cell culture

Yes/yes

Measles virus

GIP, DAD

H&E, ISH, IHC

Yes/no

Respiratory syncytial virus

DAD, hemorrhagic PN, GIP

H&E, IHC, ISH

Yes/no

Rubella virus

LIP, DAD

ISH, PCR

Yes, congenital/no

Hemorrhagic fever viruses (Ebola, Marburg HF, Kyasanur HF, Omsk HF)

Hemorrhagic PN

ISH, PCR

No/yes

Human immunodeficiency virus (HIV)

DAD, LIP, interstitial fibrosis

ISH, PCR

Yes/yes

Rickettsia rickettsii, R. prowazekii, R. typhi

Edema, DAD, LIP, vasculitis

IHC, PCR

No/yes


IHC immunohistochemistry, ISH in situ hybridization, PCR polymerase chain reaction, HF hemorrhagic fever

Pneumonia in children occurs in two peak ages: in early childhood and later in school children. Whereas pneumonia in school children is not much different from that in adults, pneumonia in early childhood is different. In small children the infiltration by leukocytes is much less pronounced compared to adults; however, the symptoms are much more pronounced. When calculating the density of leukocytes in alveolar septa, a mild infiltration by lymphocytes can be accompanied by dramatic shortness of breath and severe hypoxia, even requiring assisted ventilation. Infection in children in the first 2 years of life can happen as intrauterine infection or as an infection shortly after birth (Fig. 8.26).

A299455_1_En_8_Fig26_HTML.jpg


Fig. 8.26
Perinatal infection and pneumonia with EBV in a 6-month-old child. Left side photograph shows a mild lymphocytic infiltration, predominantly peribronchiolar. Right: In situ hybridization for EBV. H&E, ×100, ISH, ×100


8.1.6.2 Transplacental Infection Causing Pneumonias in Childhood


Infections can occur in children already in the fetal period via transplacental infection. Some of these infections such as measles when occurring during the first 3 months of gestation will cause developmental defects especially in the brain. Bacterial and fungal infections will not occur in this period, because for an infection a fully developed placenta is necessary. Whereas bacterial infections via the placenta will cause placentitis and amniitis [34] and cause premature delivery or intrauterine death, infections with viruses and Rickettsia will be transmitted to the fetus. Most common although in general rare infections are caused by ureaplasma (different serotypes), CMV, EBV, and Chlamydia trachomatis and pneumoniae [3440]. The disease is also known under the name of Wilson-Mikity syndrome (Fig. 8.27).

A299455_1_En_8_Fig27_HTML.jpg


Fig. 8.27
Wilson-Mikity syndrome is another viral infection here in a 2-month-old baby. The child was transplacentally infected by the mother and developed pneumonia. Note the thickening of the alveolar septa by a lymphocytic infiltration but in addition also a proliferation of smooth muscle cells (a). In (b) the muscular proliferation is highlighted by Movat stain. Normal are single cells whereas here two to four layers of smooth muscle cells are seen. (c) In situ hybridization for CMV turned out positively. H&E, ×100, Movat pentachrome stain, ×100, ISH, ×200


8.1.7 Bronchopulmonary Dysplasia (BPD)


Bronchopulmonary dysplasia is a specific condition found in premature children. Inflammation is a major contributor to the pathogenesis of BPD, which is often initiated by a respiratory distress response and exacerbated by mechanical ventilation and exposure to supplemental oxygen [41]. Similar to Wilson-Mikity syndrome, infectious organisms such as ureaplasma and CMV have been reported to cause BPD [34, 42, 43]. In BPD sometimes remnants of infant DAD can be seen (hyaline membranes; Fig. 8.28), but the characteristic feature is interstitial fibrosis (Fig. 8.29).

A299455_1_En_8_Fig28_HTML.jpg


Fig. 8.28
Bronchopulmonary dysplasia (BPD) in a prematurely born child, which died with respiratory distress syndrome. There are hyaline membranes pointing to previous DAD but in addition mild inflammatory lymphocytic infiltrates and most important fibroblast proliferation in the septa. H&E, ×150


A299455_1_En_8_Fig29_HTML.jpg


Fig. 8.29
BPD in another prematurely born child. Here fibrosis of the interstitium is striking. Prematurity of the lung is evident by hyperplastic type II pneumocytes. H&E, bar 50 μm


8.1.8 Aspiration Pneumonia


Aspiration in children can be seen in two different forms: meconium aspiration during delivery causing severe respiratory distress and postnatal aspiration, most often as silent nocturnal aspiration in breast-fed babies. Risk factors for severe meconium aspiration are fetal distress and birth asphyxia [44, 45]. The diagnosis is most often made at autopsy. In addition to DAD, also a foreign body granulomatous reaction might be seen, depending on the time the child has survived. In silent nocturnal aspiration, children swallow milk from breast-feeding and aspirate small amounts. This causes scattered ground glass opacities on CT scan and lipid pneumonia on histology. However, the diagnosis can be made by bronchoalveolar lavage: macrophages laden with lipid droplets in their cytoplasm in more than 10 % are diagnostic in this setting (Fig. 8.30).

A299455_1_En_8_Fig30_HTML.jpg


Fig. 8.30
Silent nocturnal aspiration. The suspected clinical diagnosis was confirmed by BAL showing >10 % of macrophages with lipid droplets in their cytoplasm. Oil red O stain, bar 20 μm


8.1.9 HIV Infection


HIV infection transmitted by HIV-positive mothers can cause also HIV in the child. It has been shown that HIV-infected women as well as HIV-infected family members coinfected with opportunistic pathogens might transmit these infections more likely to their infants than women without HIV infection, resulting in increased acquisition of such infections in the young child [46]. Otherwise HIV infection in children is morphologically similar to that in adults. Within the spectrum of opportunistic infections, Pneumocystis jirovecii is the most common.


8.2 Granulomatous Pneumonias



8.2.1 Introduction


The name granuloma is derived from the Latin word granulum, which means grain. The ending -oma is a Greek ending, used to designate a nodular swelling. Therefore granuloma is a nodular, well-circumscribed macroscopic lesion. With the invention of microscopy, this term has been extended to small nodular aggregates of cells. Over the decades the definition has undergone different interpretations. Some use granuloma strictly for well-circumscribed lesions, whereas others also designate a more loose aggregate of inflammatory cells as granuloma.

Epithelioid cell granulomas originally were recognized as a granulomatous inflammatory reaction elicited by infectious organisms. The first organisms identified were Mycobacterium tuberculosis and bovis and Treponema pallidum [47]. In the nineteenth century, Schaumann, Besnier, and Boeck recognized another epithelioid cell granulomatosis, which, due to the macroscopic resemblance to dermal sarcoma, they called sarcoidosis [48]. In the following decades, various epithelioid cell granulomatoses have been added, and even in the 1990s, new diseases have been reported, like zirconiosis [4952].


8.2.2 What Influences Granuloma Formation? Why Necrosis?


The formation of epithelioid cell granulomas requires a combination of at least two different sets of stimulants: (a) stimulants for granuloma formation and (b) stimulants for epithelioid and Langhans cell differentiation. So what are the driving forces?

Granuloma formation is an old phylogenetic process by which complex organisms protect themselves against invading organisms or toxic substances. The invader or a toxic substance is isolated by granulation tissue or is phagocytosed and degraded simply by macrophages as part of the innate immune system. If these cells can kill the invading organism, no further defense line is required. If the invader cannot be ingested and degraded by these cells, histiocytes and macrophages can form foreign body giant cells, which are more efficient in phagocytosis and degradation. These cells together form foreign body granulomas. In every case the invading organism cannot be killed by phagocytosis, another defense line is activated, which includes immune mechanisms. This more powerful line of defense is the epithelioid cell granuloma. The driving forces, which induce granuloma formation, are the macrophages, the antigen-presenting cells, such as Langerhans and dendritic reticulum cells, and the T and B lymphocytes [5356]. Among the different cytokines released are interleukins 1β, 2, 3, 8, 10, 12, 17, macrophage migration inhibitory factor 1 (MIF1), IFNγ, and TNFα. How these factors act and interact is still not understood; however, macrophages and lymphocytes are activated and immobilized. This is followed by the cytokine-induced transformation of macrophages into epithelioid and foreign body giant cells [5762]. Giant cells can be either formed by fusion of macrophages or by nuclear division without cell division. Foreign body giant cells further on differentiate into Langhans giant cells. This process of transformation is maintained by the same secretory factors, which are produced in larger quantities by the epithelioid cells and by infiltrating lymphocytes [63].

But why do we find non-necrotizing and necrotizing epithelioid cell granulomas even in the presence of the same organism?

Different substances either actively liberated from mycobacteria or passively by degradation can induce granuloma formation. Among them are trehalose-6,6′-dimycolate, lipoarabinomannan, and 65 kDa antigen of mycobacterial capsule (a chaperonin) [61, 62]. These products stimulate granuloma formation by the induction of cytokine gene expression, mainly IL1β or TNFα. In addition they have other effects, like induction of apoptosis, enhancing coagulation, and together release TNFα, which subsequently induce necrosis by occlusion of small blood vessels. The mycobacterial chaperonin also stimulates monocytes to express mRNA for TNFα and to release IL6 and IL8, cytokines which are chemoattractants for lymphocytes. In some patients necrotizing and non-necrotizing epithelioid cell granulomas, induced by M. tuberculosis, can be found side by side. The underlying mechanism is not completely understood. One possible explanation might be the mycobacterial burden: large amounts of mycobacteria release large quantities of coagulation factors and thus induce infarct-like necrosis. Another explanation is within the interaction of virulent stains of mycobacteria and host defense cells [63].

When we go back to morphology, we can see three different settings, in which we encounter necrosis: M. tuberculosis escape the immune defense, multiply, invade vessel walls, and are in part degraded by leukocytes, and by this a massive liberation of capsule constituents occurs; epithelioid cell granulomas develop in vessels walls and obstruct or occlude the vessel lumen, and ischemic necrosis follows; an imbalance of the virulence of the mycobacteria and the immune defense capability of the host is in favor of the invading organism. These factors together might lead to higher concentration of TNFα, as well as trehalose-6,6′-dimycolate, lipoarabinomannan, and chaperonin. In addition vasculitis-associated and released thrombogenic factors may synergistically act together to induce this characteristic caseous necrosis (Fig. 8.31).

A299455_1_En_8_Fig31_HTML.jpg


Fig. 8.31
Macroscopy of nodular tuberculosis with many large and small nodules with caseous necrosis

When classifying granulomatous pneumonias, we will discern epithelioid from histiocytic granulomas and as a second step differentiate infectious from noninfectious forms.


8.2.3 Morphologic Spectrum of Epithelioid Cell Granulomas


Epithelioid cell granulomas are a specific form of granulomas, composed of epithelioid cells, giant cells, and lymphocytes (epithelioid: epithel = the stem of epithelium and oid = similar to). This type of granuloma can be induced by a variety of quite different stimuli. Epithelioid and giant cells are specialized members of the monocyte/macrophage lineage, the first a differentiated secretory cell (Fig. 8.32) and the second a specialized phagocytic cell (Fig. 8.33). Giant cells can be either formed by cell fusion or by incomplete cell division (no cytoplasmic division). Both ways have been proven experimentally [56, 64, 65]. First foreign body giant cells are formed, which later reorganize into Langhans cells. These are characterized by a nuclear row opposite to the phagocytic pole of the cell. Lymphocytes are usually layered at the outer granuloma shell and can be numerous or sparse. Phenotypically these are T lymphocytes, whereas B lymphocytes are loosely arranged outside the granulomas. T-helper-1 and T-helper-2 and cytotoxic T lymphocytes (CD8+) can be present in the granulomas, with the composition depending on the type of underlying disease. This will be discussed later.

A299455_1_En_8_Fig32_HTML.jpg


Fig. 8.32
Cytology of epithelioid cells. The nuclei are curved, the cytoplasmic border is ill defined. Giemsa, bar 10 μm


A299455_1_En_8_Fig33_HTML.jpg


Fig. 8.33
Cytology of a giant cell with numerous nuclei. Pap stain, ×400

We can encounter different stages of granuloma formation: first we see a loose aggregation of macrophages, histiocytes, lymphocytes, and even neutrophils. During each step the granuloma becomes more compact, and the margins are better circumscribed. During aging, epithelioid cell granulomas might undergo fibrosis and hyalinization (Figs. 8.34, 8.35, and 8.36). However, in some diseases like extrinsic allergic alveolitis, the epithelioid cell granulomas remain less well delineated and tend to be more loosely arranged. Also a spillover of lymphocytes into adjacent alveolar septa is seen.

A299455_1_En_8_Fig34_HTML.jpg


Fig. 8.34
Early epithelioid cell granuloma, here in a case of sarcoidosis. Note the scattered lymphocytes within and outside the granuloma. H&E, bar 50 μm


A299455_1_En_8_Fig35_HTML.jpg


Fig. 8.35
Well-developed epithelioid cell granuloma in sarcoidosis. Epithelioid and Langhans cells are easily seen; lymphocytes are now scarce. H&E, bar 20 μm


A299455_1_En_8_Fig36_HTML.jpg


Fig. 8.36
Old epithelioid cell granuloma in a patient with long-standing sarcoidosis. Almost all cells vanished, a few epithelioid cells are seen. Trichrome stain, ×150

A very important finding is central necrosis, defining the necrotizing epithelioid cell granuloma. Small necrobiotic foci or few apoptotic cells are not regarded as necrosis. The necrosis is either stained eosinophilic with minimal amounts of nuclear debris, or may contain larger amounts of nuclear debris, or stained blue violet by H&E. In early necrosis neutrophils can be found. The descriptive term caseous necrosis is often used; however, it should be reminded that this term was invented to describe these necroses macroscopically: a caseous necrosis is characterized by a yellowish color and soft, cheese-like consistency (Fig. 8.31).


8.2.4 The Causes of Epithelioid Cell Granulomas and Their Differential Diagnosis


Pathologists usually differentiate granulomatoses by their morphologic appearance: if there is an epithelioid cell granuloma with necrosis, primarily infectious diseases are to be discussed, whereas in non-necrotizing granulomas, other diagnoses are to be added. Although this rule will be true in most cases, it should be reminded that sometimes necrosis is not associated with infection, as in necrotizing sarcoid granulomatosis and some cases of bronchocentric granulomatosis.

The distribution pattern of the granulomas may assist in sorting out specific diseases: the distribution of granulomas along lymphatic vessels is quite characteristic in sarcoidosis, whereas an airspace-oriented pattern is seen in most infectious epithelioid cell granulomatoses. However, the distribution pattern might not be apparent in transbronchial biopsies.


8.2.4.1 Infectious Epithelioid Cell Granulomas



Tuberculosis

Members of the M. tuberculosis complex, i.e., M. tuberculosis, M. bovis and BCG, M. africanum, and M. microti, cause tuberculosis. These mycobacteria belong to a group of fast-growing mycobacteria (Table 8.4). Virulence of these mycobacteria varies from medium- to high-virulent strains. Depending on the virulence on the one hand and the competence of the hosts’ immune system, the morphology is reflected by widespread necrosis or by non-necrotizing epithelioid cell granulomas (Figs. 8.37, 8.38, and 8.39). The faith of the granulomas depends on stabilization or destabilization of this balance between virulence of the mycobacteria and the immune system of the host (see schema below): improved immune competence combined with antituberculous therapy is accompanied by inhibition of mycobacterial growth, stabilization of granulomas, fibrosis, and hyalinization. The opposite results when a decrease of immunocompetence and increase of virulence occur. This is reflected by necrosis up to necrotizing pneumonia with abscess formation and the inability to mount a granulomatous response, as it can be seen in end-stage AIDS patients infected with M. tuberculosis.


Table 8.4
Types of mycobacteria in tuberculosis type, which are pathogenic for humans

















M. tuberculosis

M. bovis and Bacillus Calmette-Guerin

M. africanum with subtypes M. suricattae and M. mungi

M. microti

M. canetti

M. pinnipedii


A299455_1_En_8_Fig37_HTML.jpg


Fig. 8.37
Tuberculosis with large necrosis and concomitant alveolar proteinosis, which results in widespread distribution of the mycobacteria. This condition is based on an impaired immune function and usually also highly virulent strains of M. tuberculosis. H&E, bar 0.1 mm


A299455_1_En_8_Fig38_HTML.jpg


Fig. 8.38
Tuberculosis in an immunocompromised patient. There is widespread necrosis and the granuloma formation is impaired. The granuloma wall is broken down at two areas in this section, and mycobacteria can escape the host’s immune defense. H&E, ×100


A299455_1_En_8_Fig39_HTML.jpg


Fig. 8.39
Tuberculosis in a normal host. One of the granulomas present with a small focus of necrosis, whereas most of the granulomas not. Transbronchial biopsy, H&E, bar 50 μm


A299455_1_En_8_Sch1_HTML.jpg

Schema: The balance of the host’s immune system capability and the virulence of the mycobacterial strain: extensive necrosis in tuberculosis associated with alveolar proteinosis points to impaired immune reaction, whereas a good functioning immune system and slowly growing mycobacteria will result in healing or scar.

A wide variety of responses and patterns can occur in tuberculosis. Infection in the European population is frequent; up to 90 % of the population acquire a mycobacterial infection in early adulthood; however, only 1–3 % of this population will present with symptoms. In the majority of the population, this infection will cause tiny granulomas in the mid and upper portion of the lower lobes. These granulomas undergo fibrosis, and a scar is all what can be found quite frequently in this location at autopsies decades later.

Clinical symptoms are cough, night sweats, temperature around 38 °C, and fatigue. Radiologically tuberculosis presents with single or multinodular densities but also often simulates lung cancer. Even on CT scan, the differential diagnosis cannot be made with certainty.

In patients presenting with tuberculosis, the initial form is most often a multinodular disease with caseous necrosis but located in one of the lung lobes (usually lower lobes). Depending on the ability of the patient’s immune system, vasculitis can occur. Under tuberculostatic treatment this type of tuberculosis usually heals leaving scars and bronchiectasis. These in later life can be the preformed cystic structures prone to mycetoma.

In rare instances the primary infection had destroyed large areas of the lung and the necrotic focus cannot be replaced by scar tissue. In this case the necrotic focus is encased by granulation tissue, which is subsequently replaced by scar tissue. In the center the necrotic focus is still present, and mycobacteria are viable. This lesion is called tuberculoma (Fig. 8.40).

A299455_1_En_8_Fig40_HTML.jpg


Fig. 8.40
Tuberculoma detected incidentally during X-ray and removed because clinically suspected for malignancy. Resection specimen formalin fixed

Secondary tuberculosis can occur in some patients in later life either as an exacerbation from a tuberculoma or by a secondary infection. In these cases the upper lobes are more often affected. Usually in this condition, miliary tuberculosis occurs: mycobacteria get access to the blood vessels causing vasculitis, and the organisms are disseminated within the lung but also to other organs (Fig. 8.41).

A299455_1_En_8_Fig41_HTML.jpg


Fig. 8.41
Miliary tuberculosis, autopsy specimen. Numerous small nodules are scattered in this lung, each representing a granuloma with necrosis

There are some complications from tuberculosis, such as hemorrhage, when the necrotizing granuloma destroys the wall of larger pulmonary arteries. This will cause diffuse bleeding and ultimately the death of the patient (Fig. 8.42). Another complication is access of the granulomas and their mycobacterial content to larger airways, which will result in aerogenous spreading of the organisms, but also infection of other humans within the patient’s living area (Fig. 8.43).

A299455_1_En_8_Fig42_HTML.jpg


Fig. 8.42
Autopsy specimen showing massive hemorrhage from an erosion of a large pulmonary artery caused by caseous tuberculosis


A299455_1_En_8_Fig43_HTML.jpg


Fig. 8.43
Tuberculosis-induced necrosis has opened this bronchus and the infectious organisms can now be distributed through the airways but also will be expectorated and can infect other people. H&E, ×100

Diagnosis is established first by the demonstration of an epithelioid granulomatous reaction, followed by the proof of mycobacteria within the granuloma or in cytological material (BAL, smear) and by culture or PCR. This will be discussed in detail at the end.


Mycobacteriosis

This is an infection with atypical mycobacteria (other than M. tuberculosis complex (MOTT)). It was once a rare disease, causing epithelioid cell granulomas in newborn and young children. It now has become a well-recognized disease in patients suffering from AIDS, or in otherwise immunocompromised patients. Many different mycobacteria can induce predominantly non-necrotizing epithelioid cell granulomas, among them M. avium-intracellulare, M. fortuitum, M. gordonae, M. kansasii, and M. xenopi, to name just the more common species. Some cause local disease, like skin lesions by M. marinum, whereas others cause systemic disease, like M. avium (Fig. 8.44). The diagnosis of mycobacteriosis can be made by acid-fast stains but in most instances requires culture or molecular biology techniques for species definition. In cases of severe immunodeficiency, the host’s reaction might be impaired, which results in the inability to form epithelioid cell granulomas. In these cases macrophage granulomas are found, similar to granulomas in lepromatous lepra.

A299455_1_En_8_Fig44_HTML.jpg


Fig. 8.44
Mycobacteriosis with non-necrotizing epithelioid cell granulomas. Note the proximity of the granulomas to the airspace, which points to an airborne infection. M. gordonae was identified by acid-fast stain and PCR. H&E, ×160

The reproductive cycle of MOTT species is quite variable: M. avium-intracellulare is a very slow-growing organism, which requires a culture for up to 11 weeks until the organism can be identified, whereas M. fortuitum is a fast-growing organism, which can be identified within 2 weeks. Necrotizing granulomas are usually found in these fast-growing species.

Recently a new disease was described as hot tub lung disease. Mycobacteria of the MOTT complex were identified as the causing agent [66, 67]. If this is an infectious disease caused by slow-growing MOTT, species in otherwise immunocompetent patients or a hypersensitivity reaction is not clear. An answer to this question is complicated as a hyperreactivity or allergic reaction can occur in mycobacterial infections as part of the immune defense and thus is not a proof of an allergy (Fig. 8.45). Biopsies from patients suffering from this type of disease will show exposure-related symptoms, i.e., increase of symptoms during the weekend (exposure to mycobacteria in hot tub) and relief of symptoms during the week. Morphologically the lesions present as non-necrotizing epithelioid cell granulomas, similar to classical mycobacteriosis with slow-growing mycobacteria such as M. avium-intracellulare (Table 8.5).
Jun 26, 2017 | Posted by in RESPIRATORY | Comments Off on Pneumonia

Full access? Get Clinical Tree

Get Clinical Tree app for offline access