4 neumonitis por hipersensibilidad 2012 dr. selman ya leido muy bueno.pdf

Upload: denisse-diaz

Post on 01-Jun-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    1/12

    Immunopathology, Diagnosis, and Managementof Hypersensitivity Pneumonitis

    Moisés Selman, M.D. 1 Ivette Buendía-Roldán, M.D. 1

    1 Instituto Nacional de Enfermedades Respiratorias   “Ismael Cosío

    Villegas,” México

    Semin Respir Crit Care Med 2012;33:543–554.

    Address for correspondence and reprint requests   Moisés Selman,

    M.D., Instituto Nacional de Enfermedades Respiratorias, Tlalpan 4502,

    CP 14080, México DF, México (e-mail: [email protected]).

    Hypersensitivity pneumonitis (HP) is a complex syndrome

    that results from the exposure to a wide variety of   nely

    dispersed antigens of size suitable for reaching the alveolar

    spaces (usually particles smaller than 5 µm) that include

    mammalian and avian proteins, fungi, thermophilic bacteria,

    and certain small-molecular-weight chemical compounds that

    combine with host proteins to form haptens.1 The incidence

    and prevalence of HPare dif cult to estimate precisely because

    the disease represents a syndrome with different causative

    agents, and epidemiological studies lack uniform diagnostic

    criteria.Interestingly, however, the incidence of HP is low if we

    consider that the offending antigens are numerous and widely

    distributed around the world. Actually, an extensive number of 

    etiologicagents and sources of antigenscapableof inducing HP

    have been described, and the listof environments and agents is

    always increasing (► Tables 1, 2 , 3). Thus, for example, in the

    last 10 years it has been demonstrated that heated water

    colonized by Mycobacterium avium may provoke HPin hot tub

    users.2 Likewise, Mycobacterium immunogenum may contam-

    inate metal working  uid (MWF), causing disease when they

    become aerosolized, mostly in automotive industries.3 Work-

    ers with exposure to MWF are generally in the manufacturing

    sector in processes like metal machining, forging, and stamp-

    ing. HP has also been described in individuals exposed to

    Cytophaga   endotoxin in a nylon plant.4 In these workers

    Cytophaga wasisolated fromthe plant air-conditioning system,

    and patients showed precipitins to  Cytophaga antigen. Finally,

    indirect exposure through a partner has been found to cause

    HP, which can complicate the identication of the offending

    antigen.

    Thelow incidence of HP suggests that geneticor additional

    environmental factors are necessary to develop the disease.

    Keywords

    ►   hypersensitivity 

    pneumonitis

    ►   extrinsic allergic

    alveolitis

      lung 

    brosis

     Abstract   Hypersensitivity pneumonitis (HP) is an inammatory interstitial lung disease caused by a wide variety of organic particles and certain small-molecular weight chemical

    compounds that provoke an exaggerated immune response in susceptible individuals.

    The clinical manifestations are heterogeneous and have been classically described as

    acute, subacute and chronic. The chronic form has an insidious onset over a period of 

    months or years, with progressive dyspnea and often evolves to brosis. The pathology 

    is characterized by a bronchiolocentric interstitial mononuclear cell inltration, non-necrotizing poorly formed granulomas, cellular pneumonitis and variable degrees of 

    brosis. However, morphological diagnosis of HP is complicated because the subacute/

    chronic forms may be dif cult to distinguish from idiopathic pulmonary  brosis/usual

    interstitial pneumonia and nonspecic interstitial pneumonia. In general, diagnosis of 

    HP represents a challenge for clinicians that need to weigh a constellation of clinical,

    laboratory, radiographic and (when available) pathological evidence for each patient to

    assess the certainty of the diagnosis. The cornerstone of therapy is antigen avoidance.

    Although clinical trials are scanty, corticosteroids are usually indicated based upon

    expert opinion. In this review we summarize the current evidence regarding the

    diagnostic criteria and therapeutic strategies as well as the immunopathological

    mechanisms putatively implicated in the development of the disease.

    Issue Theme   Orphan Lung Diseases;

    Guest Editor, Jay H. Ryu, M.D.

    Copyright © 2012 by Thieme Medical

    Publishers, Inc., 333 Seventh Avenue,

    New York, NY 10001, USA.

    Tel: +1(212) 584-4662.

    DOI   http://dx.doi.org/

    10.1055/s-0032-1325163.

    ISSN  1069-3424.

    543

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    2/12

    Also, it is likely that a number of mild cases are misdiagnosed

    as viral infections, and some severe advanced cases as idio-

    pathic pulmonary   brosis (IPF) or other   brotic lung

    disorders.

    According to several studies the prevalence of farmer’s

    lung, one of the best-known types of HP, can be estimated in

    0.5 to 3% of exposed farmers.5 Pigeon breeder’s disease (PBD),

    another common form of HP, seems to occur frequently

     Table 1  Fungal and Bacterial Antigens Implicated in Hypersensitivity Pneumonitis

    Disease Antigen Source

    Farmer’s lung   Faeni rectivirgula   Moldy hay, grain, silage

    Ventilation pneumonitis;humidier lung; air conditionerlung

    Thermoactinomyces vulgaris,Thermoactinomyces sacchari,Thermoactinomyces candidus

    Klebsiella oxytoca

    Contaminated forced-air systems; waterreservoirs

    Bagassosis   T. vulgaris   Moldy sugarcane (ie, bagasse)

    Mushroom worker’s lung   T. sacchari    Moldy mushroom compost

    Enoki mushroom worker’s lung(Japan)

    Penicillium citrinum   Moldy mushroom compost

    Suberosis   Thermoactinomyces viridis, Aspergillusfumigatus, Penicillium frequentans Penicillium

     glabrum

    Moldy cork

    Detergent lung; washingpowder lung

    Bacillus subtilis enzymes Detergents (during processing or use)

    Malt worker’s lung   Aspergillus fumigatus, Aspergillus clavatus   Moldy barley 

    Sequoiosis   Graphium, Pullularia, and Trichoderma spp.,

     Aureobasidium pullulans

    Moldy wood dust

    Maple bark stripper’s lung   Cryptostroma corticale   Moldy maple bark

    Cheese washer’s lung   Penicillium casei, A. clavatus   Moldy cheese

    Woodworker’s lung   Alternaria spp., wood dust Oak, cedar, and mahogany dust, pine andspruce pulp

    Hardwood workeŕ  s lung   Paecilomyces   Kiln-dried wood

    Paprika slicer’s lung   Mucor stolonifer    Moldy paprika pods

    Sauna taker’s lung   Aureobasidium spp., other sources Contaminated sauna water

    Familial HP   B. subtilis   Contaminated wood dust in walls

    Wood trimmer’s lung   Rhizopus spp., Mucor  spp. Contaminated wood trimmings

    Composter’s lung   T. vulgaris, Aspergillus   Compost

    Basement shower HP   Epicoccum nigrum   Mold on unventilated shower

    Hot tub lung   Mycobacterium avium complex Hot tub mists; mold on ceiling

    Wine maker’s lung   Botrytis cincrea   Mold on grapes

    Woodsman’s disease   Penicillium spp. Oak and maple trees

    Thatched roof lung   Sacchoromonospora viridis   Dead grasses and leaves

    Tobacco grower’s lung   Aspergillus spp. Tobacco plants

    Potato riddler’s lung Thermophilic ac tinomycetes, F. rectivirgula,T. vulgaris, Aspergillus  spp.

    Moldy hay around potatoes

    Summer-type pneumonitis   Trichosporon cutaneum   Contaminated old houses

    Dry rot lung   Merulius lacrymans   Rotten wood

    Stipatosis   Aspergillus fumigatus, T. actinomycetes   Esparto dustMachine operator’s lung   Mycobacterium immunogenum, Pseudomona

     uorescensAerosolized metalworking  uid

    Residential provokedpneumonitis

     Aureobasidium pullulans   Residential exposure

    Humidier lung   Naegleria gruberi, Acanthamoeba polyphaga, Acanthamoeb a castellani, Bacillus  spp., others

    Contaminated water from home humidier,ultrasonic misting fountains

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán544

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    3/12

    among bird fanciers taking care of hundreds or thousands of 

    pigeons (ie 20 to 20,000 per 100,000 persons at risk).1

    However, the prevalence of PBD among people with only a

    few birds at home is uncertain. Recently, in a large, general-

    population-based cohort of HP patients, the overall incidence

    rate was  1 per 100,000 in the UK population.6 The disease

    appears to be a rare interstitial lung disease (ILD) in children,and a recent report in Denmark showed an incidence of 2/

    year and a point prevalence of 4/1,000,000 children.7 Inci-

    dence/prevalence seems to be even lower according to the

    few pediatric cases reported worldwide and in relation to a

    study of lung biopsy from 101 immunocompetent children

    with diffuse interstitial lung disease in a North American

    cohort derived from 13 pediatric centers over a 4 year period

    in which only two children with HP were identied.8

    Pathogenic Mechanisms

    HP is an immunopathological disorder occurring in suscepti-

    ble individuals where both humoral and cellular mechanisms

    participate in the development of the lung lesions. However,

    the genetic basis of the disease is poorly understood. Some

    studies indicate that gene polymorphisms of major histocom-patibility complex (MHC) class II alleles are implicated in the

    risk to develop the disease.9–13 Also, polymorphisms in the

    transporters associated with antigen processing (TAP) genes

    may also be involved.14 TAP play an important role trans-

    porting peptides across the endoplasmic reticulum mem-

    brane for MHC class I molecule assembly. Almost every T cell

    response is controlled by the molecular interaction between

    the clonotypically expressed αβ  T cell receptor and cognate

     Table 2  Chemicals Implicated in Hypersensitivity Pneumonitis

    Disease Antigen Source

    Pauli’s reagent alveolitis Sodium diazobenzene sulfate La borator y reagent

    Chemical worker’s lung Isocyanates; trimellitic anhydride Polyurethane foams, spray paints, elastomers,special glues

    Dental techniciań  s lung Methyl methacrylate Polishing and grinding prostheses

    Vineyard sprayer’s lung Copper sulfate Bordeaux mixture

    Pyrethrum HP Pyrethrum Pesticide

    Epoxy resin lung Phthalic anhydride Heated epoxy resin

    UNKNOWN

    Bible printer’s lung Moldy typesetting water

    Coptic lung (mummy handler’s lung) Cloth wrappings of mummies

    Grain measurer’s lung Cereal grain

    Coffee worker’s lung Coffee bean dust

    Tap water lung Contaminated tap water

    Tea grower’s lung Tea plants

    Mollusk shell HP Sea snail shell

    Swimming pool worker’s lung Aerosolized endotoxin from poolWater sprays and fountains

     Table 3  Animal Proteins Implicated in Hypersensitivity Pneumonitis

    Disease Antigen Source

    Pigeon breeder’s or pigeon fancier’sdisease

    Avian droppings, feathers, serum Parakeets, budgerigars, pigeons, chickens,turkeys

    Pituitary snuff taker’s lung Pituitary snuff Bovine and porcine pituitary proteins

    Fish meal worker’s lung Fish meal Fish meal dust

    Bat lung Bat serum protein Bat droppings

    Furrier’s lung Animal fur dust Animal pelts

    Animal handler’s lung, laboratory worker’s lung, insect proteins

    Rats, gerbils Urine, serum, pelts, proteins

    Miller’s lung   Sitophilus granarius (ie, wheat weevil) Dust-contaminated grain

    Lycoperdonosis Puffball spores Lycoperdon puffballs

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán   545

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    4/12

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    5/12

    respiratory infection. Moreover, patients with proved HP may

    show a rise of inuenza viruses or  Mycoplasma pneumoniae

    antibody titers.40,41 Infarmers, the acute form ofHP should be

    distinguished from an inuenza-like illness resulting from

    the nonimmunological reaction to inhaled mold dusts (bac-

    terial or fungal toxins), called organic dust toxic syndrome. 42

    These patients, however, have no precipitins to antigens and

    usually present with normal clinical  ndings on respiratoryexamination and chest radiographs.

    Subacute HP

    The subacute form of HP usually results from continual low-

    level exposure to inhaled antigens or more likely from the

    progression of an undiagnosed acute HP. This clinical form is

    characterized by gradual development of productive cough

    and dyspnea over several days to weeks. Some patients

    present with fever during the  rst weeks, and fatigue, an-

    orexia, and weight loss are commonly observed. Physical

    examination usually reveals tachypnea and bibasilar inspira-

    tory crackles. Wheezing, provoked by small airway obstruc-

    tion, is not common, but it occurs in some patients. Thedifferential diagnosis includes granulomatous lung disorders,

    primarily sarcoidosis, lymphocytic interstitial pneumonia

    (LIP), drug-induced lung disease, idiopathic cellular nonspe-

    cic interstitial pneumonia (NSIP), and respiratory bronchi-

    olitis/interstitial lung disease (RB-ILD) (a smoker-related lung

    disorder) or other bronchiolar disorders.1

    Chronic HP

    It has been proposed that the chronic presentation of HP may

    result from two different clinical scenarios: (1) from unrec-

    ognized acute/subacute episodes (recurrent chronic HP) and

    (2) from a slowly progressive disease in patients exposed tolow levels of antigen and without history of acute episodes

    (insidious chronic HP). Chronic HP presents as prolonged and

    relentlesspneumonitis with progressivedyspnea on exertion,

    cough, fatigue, malaise, and weight loss. Despite chronicity,

    these patients may stabilize or even improve after antigen

    avoidance and antiinammatory/immunosuppressive treat-

    ment. However, they often progress, evolving to diffuse

    brosis and end-stage lung disease. Digital clubbing may be

    seen in advanced  brotic HP and may help to predict poorer

    outcome. Chronic HP may mimic idiopathic pulmonary bro-

    sis (IPF), and in this case the differential diagnosis may be

    extremely dif cult. Interestingly, some patients with HP,

    primarily farmers with recurrent acute episodes, developan obstructive lung disease with emphysematous changes

    instead of lung  brosis. Differential diagnosis of chronic HP

    includes brotic NSIP and, importantly, IPF.43

    It is important to emphasize that, despite the wide use of 

    this clinical (acute, subacute, chronic) classication, signi-

    cant overlap between these interrelated categories often

    occur, which is at least partially due to the lack of clear and

    standardized criteria to differentiate between these various

    forms. The classication is further complicated because

    chronic HP may still be active and progressive. Actually,

    cluster analysis of a large group of HP patients failed to

    identify these three categories and proposed only two.44 In

    the  rst category, patients present with recurrent systemic

    symptoms a few hours following antigen exposure (that is,

    acute HP). The subacute presentation was considered only a

    variant of the acute form of the disease, with similar but

    attenuated symptoms remaining for daysor weeks.40 Patients

    in the second category correspond to chronic ones.

     Acute Exacerbations in Chronic HPSome patients with chronic HP may occasionally experience

    an acute exacerbation (as described in IPF) characterized by

    sudden and rapid deterioration of dyspnea, severe increase of 

    hypoxemia, and the appearance of new ground-glass opaci-

    ties or consolidation. This severe process seems to be associ-

    ated with male sex, smoking habit, more   brosis and less

    ground-glass opacities in initial high-resolution computed

    tomographic (HRCT) scan, and worse pulmonary function

    tests at the time of diagnosis. Lung biopsy/autopsy during the

    acute exacerbation reveals predominantly diffuse alveolar

    damage (DAD) but also organizing pneumonia.45,46

    Pulmonary Function Tests

    Lung function abnormalities play an important role in deter-

    mining the severity of the disease during the  rst evaluation

    and follow-up but are neither specic nor diagnostic for HP

    because similar modications are found in many other inter-

    stitial lung diseases (ILDs).1 HP patients show a restrictive

    ventilatory defect characterized by a decrease of forced vital

    capacity (FVC) and total lung capacity (TLC). However, be-

    cause HP affects the small airways, a mixed obstructive/

    restrictive functional pattern may be observed in some

    patients, with decrease in forced expiratory   ow in the

    midexpiratory phase.

    47,48

    An early reduction of diffusingcapacity for carbon monoxide (DL CO) is often found. HP

    patients exhibit hypoxemia at rest that usually worsens

    with exercise. Patients with mild/moderate disease may be

    normoxemic at rest, but oxygen desaturation is usually

    observed with exercise.

    The correlation between pulmonary functional abnormal-

    ities and the prognosis of HP has not been properly evaluated,

    but it is the general belief that patients with more severe

    abnormalitieshave a worse outcome. However,some patients

    with severe impairment may recover, whereas others with

    relatively mild functional abnormalities at the onset of dis-

    ease may develop progressive pulmonary  brosis or airway

    obstruction.

    Imaging

    Chest radiographs can be useful in the diagnostic evaluation

    of suspected HP, but there are no specic  ndings, and some

    patients with HP may have normal chest radiographs. Abnor-

    malities in acute and subacute HP patients include poorly

    dened small nodules throughout both lungs and ground-

    glass attenuation, either patchy or diffuse. Airspace consoli-

    dation can also be observed. In advanced chronic HP, ndings

    also include pulmonary brosis with linear interstitial opaci-

    ties, lung distortion, and honeycombing. In these cases,

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán   547

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    6/12

    cardiomegaly may develop as a result of chronic pulmonaryhypertension and cor pulmonale.

    Computed Tomography

    Computed tomography (CT) and HRCT provide a better-

    quality and more precise estimation of the pattern, extent,

    and distribution of the disease and correlate better with

    clinical and histopathological parameters.49

    In acute HP, HRCT manifestations include a diffuse and

    hazy increased parenchymal density and patchy or wide-

    spread airspace opacication (►Fig. 1). The characteristic

    features of subacute HP consist of patchy or diffuse bilateral

    ground-glass attenuation, poorly de

    ned small round cen-trilobular nodules (usually

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    7/12

    whereas asymptomatic (antigen-exposed) subjects without

    sensitization had intermediate rates of clearance.59 However,

    the usefulness of this method is uncertain because there is

    overlap in clearance rates of the radioisotope between ILD

    patients and normal subjects and because cigarette smokingand other variables inuence the respiratory clearance of 

    99mTc-DTPA.

    Laboratory Tests

    Routine laboratory tests are not useful either for diagnosis or

    to monitor disease activity or progression.

    Serum-precipitating IgG antibodies against the offending

    antigen are usually detectable. However, the presence or

    absence of these antibodies should be taken carefullybecause

    similar antibodies may be found in exposed but asymptom-

    atic individuals, and false-negative results may occur, mainly

    in chronic cases.60,61 Avian antigen-specic IgG and IgA canbe easily detectable in saliva samples,62,63 which may be a

    useful approach, especially in children, and can also facilitate

    sampling for epidemiological studies.

    Bronchoalveolar Lavage

    Patients with HP usually display an increase in the total cell

    count with a remarkable elevation in the percentage of 

    lymphocytes, usually T cells (►Fig. 4). Importantly however,

    an increase in BAL lymphocytes is also observed in some

    asymptomatic HP-antigen-exposed individuals. In these

    cases, it is unclear whether the increase of lymphocytes

    represents a   “normal”   inammatory response or whether

    the apparent   “normal”  individuals have a low-intensity al-

    veolitis without clinical consequences.64 Although for a long

    time it was considered that CD8þ T cell subset was predomi-

    nant, recent evidence demonstrates that the CD4:CD8 ratiovaried according to several variables, including, the type of 

    causative antigen, the clinical status (ie, acute/subacute vs

    chronic), smoking habit, and others.32,65 A few B lymphocytes

    and plasmacellscan also bepresent inBAL uids, mainly after

    recent antigen exposure or subacute active disease.66,67 On

    the other hand, an increase of neutrophils together with

    lymphocytes characterizes the acute attacks.68,69 There is

    Figure 3   Three different levels of a high-resolut ion computed tomographic scan in a patient with chronic hypersensitivity pneumonit is. (A) On a

    background of ground-glass attenuation, extensive reticular opacities and traction bronchiectasis are frequently observed. (B) There is also

    architectural distortion of the lung parenchyma. A remarkablending in some chronic advanced patients is the presence of honeycombing (B and

    C arrows).

    Figure 4   Bronchoalveolar lavage cell prole in hypersensitivity 

    pneumonitis is characterized by a noteworthy increase of lymphocytes

    (hematoxylin and eosin; 40

    ).

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán   549

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    8/12

    also an increase of mast cells that may be useful (together

    with the increase of lymphocytes) for the differential diagno-

    sis with other ILDs.70 Actually, although there is no unique

    nding for HP, the described changes may eventually help todistinguish HP from other frequent ILDs.71

    Morphology

    Little is know about the pathological changes of the acute

    form of HP. A recent review of cases with new and rapid onset

    of symptoms reported that, in addition to the bronchiolocen-

    tric lymphoplasmacytic inltrate,  brin deposition and neu-

    trophilic inltrates are usually present.72 However, hyaline

    membranes, pneumocyte atypia, and   broblastic prolifera-

    tion as attributes of acute lung injury or diffuse alveolar

    damage were not found.Subacute HP is characterized by a bronchiolocentric pneu-

    monitis with interstitial lymphoplasmacytic inltrates, cel-

    lular bronchiolitis, and small, poorly differentiated, loosely

    arranged granulomas73 (►Fig. 5). In the absence of granulo-

    mas, the histopathological pattern in subacute/chronic HP

    can be very similar to that observed in NSIP.74 Occasional

    areas of organizing pneumonia with Masson bodies are often

    seen. Bronchiole pathology may vary, and while proliferative

    bronchiolitis obliterans has been described in farmer’s lung,

    constrictive bronchiolitis is primarily seen in PBD.75,76 Occa-

    sionally, peribronchiolar metaplasia (peribronchiolar prolif-

    eration of bronchial epithelium along thickened

    peribronchiolar alveolar walls) can be the primary histologi-cal  nding in the lung biopsy.77

    Chronic HP is characterized by variable degrees of  brotic

    changes, and in advanced cases,  brosis can be severe, with

    destruction of the lung architecture, and honeycomb changes

    that may be dif cult to distinguish from usual interstitial

    pneumonia (UIP) (►Fig. 6).78,79 Also, some chronic patients

    may show a relatively homogeneous linear   brosis resem-

    bling   brotic NSIP.79,80 In patients in whom the pattern of 

    brosis is consistent with UIP or NSIP, bronchiolocentric

    localization of the brotic lesions and the presence of Schau-

    mann bodies, giant multinucleated cells, or granulomas, may

    support the diagnosis of chronic HP. It has been recently

    suggested that cathepsin-K is a sensitive immunohistochem-

    ical marker for detection of microgranulomas.81 Bridging

    brosis between centrilobular and subpleural areas, and

    areas close to the interlobular septa is frequently seen inchronic HP. Interestingly, patients with concurrent histopath-

    ological features of pulmonary alveolar proteinosis and HP

    have been recently reported, although the putative linkage is

    unknown.82

    Diagnosis

    Clinicians should maintain a high index of suspicion for HP in

    patients with clinical, radiological, and functional features of 

    ILD.An environmental antigen is essential for the development

    of HP and must be considered when reviewing a patient’s

    clinical history. Unfortunately, it is often impossible to identifythe causal antigen, or the interval between exposure and the

    onset of symptoms is so long that the cause-and-effect rela-

    tionship is dif cult to establish. Alternatively, some patients

    have a positive history of exposure to HP antigens, and even

    specic circulating antibodies, but they have another ILD.

    The elements that contribute to diagnosis have been

    previously published.1 Diagnosis of acute HP based on (1)

    evidence of exposure (usually substantial), documented by

    history and specic antibodies against the offending antigen;

    (2) a   ulike syndrome; (3) increased BAL neutrophils and

    lymphocytes; and (4) signicant improvement after remov-

    ing the patient from the suspected environment, and wors-

    ening after reexposure.Diagnosisof subacute HP includes (1)evidence of exposure

    and specic antibodies against the offending antigen; (2)

    progressive dyspnea; (3) BAL lymphocytosis (usually>40% in

    nonsmokers); (4) ground-glass opacities, poorly dened cen-

    trilobular nodules, and mosaic attenuation on inspiratory

    images and of air trapping on expiratory CT images; (5)

    restrictive functional pattern plus hypoxemia and reduced

    diffusingcapacity forcarbon monoxide (DL CO); and (6) partial

    improvement after removing the patient from the suspected

    environment, and worsening after reexposure.

    Diagnosis of chronic HP is based on (1) evidence of 

    exposure and specic antibodies against the offending

    Figure 5   Photomicrographs of subacute hypersensitivity pneumonitis in lungs showing two common and characteristic histopathological

    features. (A)Interstitial lymphocytic inltrate anda poorly formedgranuloma (arrowhead). (B)The granuloma is typicallyfound in peribronchiolar

    localization (arrow). Epithelioid and multinucleated giant cells form a loosely organized aggregate without the compact architecture that

    characterizes other granulomatous disorders such as sarcoidosis (hematoxylin and eosin).

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán550

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    9/12

    antigen (or antigen-induced lymphocyte proliferation); (2)

    clinical behavior of chronic ILD; (3) BAL lymphocytosis; (4)

    reticular opacities superimposed to subacute changes on

    HRCT; (5) restrictive functional pattern plus hypoxemia and

    reduced DL CO; and (6) lung biopsy if there is insuf cient

    evidence for diagnosis.

     Therapeutic Approach and Prognosis

    Early diagnosis andavoidance of further antigenexposure are

    crucial in the management of these patients. Although some

    of them report complete remission of the disease despite

    subsequent exposure to the offending antigen, in most cases

    continued antigen inhalation is one of the identied causes of 

    an adverse prognosis.

    Corticosteroids are recommended in acute, severe, and

    subacute/chronic disease.However, long-term ef cacy of these

    agents has not been determined in appropriate clinical trials.

    Old evidence suggests that in farmer’s lung corticosteroids didnot inuence the long-term outcome of the disease.83

    In subacute/chronic progressive cases, the empirical

    scheme consists of 0.5 mg/kg/d of prednisone for a month

    followed by a gradual reduction until a maintenance dose of 

    10 to 15 mg per day is reached.1 This treatment should be

    discontinued if there is no clinical or functionalresponse after

    1 year.

    Inhaled steroids have been occasionally used to reduce the

    severe side effects of prolonged systemic steroid therapy;

    however, evidence of ef cacyis scanty.84–86 Patients with hot

    tub lung provoked by Mycobacterium avium complex organ-

    isms contaminating hot tub water have been treated with

    corticosteroids or with antimycobacterial therapy or both or

    even simply by avoiding further exposure, with signicant

    improvement.87,88 Also, no controlled studies of pediatric HP

    have been performed, and treatment regimens are mainly

    extrapolated from adults. Monthly high-dose intravenous

    methylprednisolone constituted the basic treatment in asmall cohort of patients in Denmark (15 mg/kg OD [every

    day] for 3 days). Additionally, oral prednisolone was initiated

    immediately, and other immunosuppressive drugs such as

    azathioprine or cyclosporine were employed in severe cases

    or in those with no obvious improvement in lung function

    within 2 to 3 months or in cases of relapse.7 Most children

    improved, and no mortality was seen.

    Thalidomide, an antiinammatory and immunomodula-

    tory drug, has proved useful in some immunopathological

    disorders, including sarcoid skin lesions.89 This drug inhibits

    the production of TNF-α, IL-12p40, and IL-18 by alveolar

    macrophages obtained from patients with diverse ILDs, in-

    cluding HP.90 However, there are no controlled clinical trialsin HP. Finally, in chronic progressivebrotic HP patients, lung

    transplantation should be considered.

    If the diagnosis is correctly and timely done, outcome is

    usually good in acute and subacute HP. By contrast, patients

    with chronic disease often progress to irreversible pulmonary

    brosis, and 30% of them die within a few years of diagno-

    sis.91,92 In general, the risk of mortality increases with evi-

    dence of brosis at lungbiopsyor HRCTor with a more severe

    respiratory impairment on pulmonary function tests.91–94

    A report from the National Center for Health Statistics

    showed that overall age-adjusted death rates increased signi-

    cantly between 1980 and 2002, from 0.09 to 0.29 per million.95

    Figure 6   (A, B, C) Photomicrographs of three patients with chronic hypersensitivity pneumonitis. Lung tissues show chronic interstitial

    mononuclear inammation,  broblastic foci, interstitial  brosis, and distortion of the lung architecture. These lesions may be misinterpreted as

    usual interstitial pneumonia. ((A) Masson trichrome staining, (B, C) hematoxylin and eosin staining.)

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán   551

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    10/12

    Proportionate mortality ratios were signicantly high for  agri-

    cultural production,  livestock and  agricultural production, crop.

    The death rate was greater in farming states, where farmer’s

    lung accounted for nearly 40% of all HP deaths. Importantly

    however, the study demonstrated that nearly 56% of HP deaths

    were due to unspecied organic dust/etiological agents.

    Conclusions

    HP is a complex and multifaceted disease that should be

    included in the differential diagnosis of any patient consult-

    ing with an ILD. Early diagnosis and avoiding further antigen

    exposure improve survival. The diagnosis requires a high

    index of suspicion by the clinician and rests on the combina-

    tion of  ndings from environmental/occupational exposure

    history, clinical behavior, serology, HRCT, BAL, and, if neces-

    sary, lung biopsy. Treatment with corticosteroids is usually

    indicated, but their long-term effect in this disease is unclear.

    In chronic patients with progressive brosis and refractory to

    medical therapy, lung transplantation should be considered.

    Experimental and clinical evidence points toward a T cell–

    mediated mechanism driving HP, but the role of the different

    T cell subsets in the development of the disease needs to be

    elucidated. A better understanding of the immunopathologi-

    cal mechanisms may help to nd HP-specic biomarkers and

    may also open new therapeutic strategies.

    References1   Selman M. Hypersensitivity pneumonitis: a multifaceted deceiv-

    ing disorder. Clin Chest Med 2004;25(3):531–547, vi

    2   Hanak V, Kalra S, AksamitTR, Hartman TE,TazelaarHD, RyuJH. Hot

    tub lung: presenting features and clinical course of 21 patients.Respir Med 2006;100(4):610–615

    3  Tillie-Leblond I, Grenouillet F, Reboux G, et al. Hypersensitivity

    pneumonitis and metalworking  uids contaminated by mycobac-

    teria. Eur Respir J 2011;37(3):640–647

    4   Nordness ME, Zacharisen MC, Schlueter DP, Fink JN. Occupational

    lung disease related to cytophaga endotoxin exposure in a nylon

    plant. J Occup Environ Med 2003;45(4):385–392

    5   Girard M, Lacasse Y, Cormier Y. Hypersensitivity pneumonitis.

    Allergy 2009;64(3):322–334

    6   Solaymani-Dodaran M, West J, Smith C, Hubbard R. Extrinsic

    allergic alveolitis: incidence and mortality in the general popula-

    tion. QJM 2007;100(4):233–237

    7   Buchvald F, Petersen BL, Damgaard K, et al. Frequency, treatment,

    and functional outcome in children with hypersensitivity pneu-

    monitis. Pediatr Pulmonol 2011;46(11):1098–11078   DishopMK, Askin FB,Galambos C, et al. Children’s Interstitial Lung

    Disease (chILD) Network: classication of diffuse lung disease in

    older children and adolescents: a multi-institutional study of the

    Children’s Interstitial Lung Disease Network [abstract]. Mod

    Pathol 2007;20:287–288

    9  Flaherty DK, Braun SR, Marx JJ, Blank JL, Emanuel DA, Rankin J.

    Serologically detectable HLA-A, B, and C loci antigens in farmer’s

    lung disease. Am Rev Respir Dis 1980;122(3):437–443

    10   Rittner C, Sennekamp J, Mollenhauer E, et al. Pigeon breeder’s

    lung: association with HLA-DR 3. Tissue Antigens 1983;21(5):

    374–379

    11   Selman M, Terán L, Mendoza A, et al. Increase of HLA-DR7 in

    pigeon breeder’s lung in a Mexican population. Clin Immunol

    Immunopathol 1987;44(1):63–70

    12   Ando M, Hirayama K, Soda K, Okubo R, Araki S, Sasazuki T. HLA-

    DQw3 in Japanese summer-type hypersensitivity pneumonitis

    induced by Trichosporon cutaneum. Am Rev Respir Dis

    1989;140(4):948–950

    13   Camarena A, Juárez A, Mejía M, et al. Major histocompatibility

    complex and tumor necrosis factor-alpha polymorphisms in pi-

    geon breeder’s disease. Am J Respir Crit Care Med 2001;

    163(7):1528–1533

    14   Aquino-Galvez A, Camarena A, Montaño M, et al. Transporter

    associated with antigen processing (TAP) 1 gene polymorphisms

    in patients with hypersensitivity pneumonitis. Exp Mol Pathol

    2008;84(2):173–177

    15   BridgemanJS, Sewell AK, Miles JJ, Price DA,Cole DK. Structural and

    biophysical determinants of  αβ T-cell antigen recognition. Immu-

    nology 2012;135(1):9–18

    16   Wucherpfennig KW, Sethi D. T cell receptor recognition of self and

    foreign antigens in the induction of autoimmunity. Semin Immu-

    nol 2011;23(2):84–91

    17  Hoppin JA, Umbach DM, Kullman GJ, et al. Pesticides and other

    agricultural factors associated with self-reported farmer’s lung

    among farm residents in the Agricultural Health Study. Occup

    Environ Med 2007;64(5):334–341

    18  Dakhama A, Hegele RG, Laamme G, Israël-Assayag E, Cormier Y.

    Common respiratory viruses in lower airways of patients withacute hypersensitivity pneumonitis. Am J Respir Crit Care Med

    1999;159(4 Pt 1):1316–1322

    19   Israël-Assayag E, Dakhama A, Lavigne S, Laviolette M, Cormier Y.

    Expression of costimulatory molecules on alveolar macrophages

    in hypersensitivity pneumonitis. Am J Respir Crit Care Med

    1999;159(6):1830–1834

    20   Depierre A, Dalphin JC,PernetD, Dubiez A, Faucompré C, Breton JL.

    Epidemiological study of farmer’s lung in   ve districts of the

    French Doubs province. Thorax 1988;43(6):429–435

    21  Warren CPW. Extrinsic allergic alveolitis: a disease commoner in

    non-smokers. Thorax 1977;32(5):567–569

    22   Baur X, Richter G, Pethran A, Czuppon AB, Schwaiblmair M.

    Increased prevalence of IgG-induced sensitization and hypersen-

    sitivity pneumonitis (humidier lung) in nonsmokers exposed to

    aerosols of a contaminated air conditioner. Respiration 1992;59(4):211–214

    23   Arima K, Ando M, Ito K, et al.Effect ofcigarette smoking onprevalence

    of summer-type hypersensitivity pneumonitis caused by Tricho-

    sporon cutaneum. Arch Environ Health 1992;47(4):274–278

    24   DalphinJC, Debieuvre D, PernetD, et al.Prevalence andrisk factors

    forchronic bronchitisand farmer’s lung in Frenchdairy farmers. Br

     J Ind Med 1993;50(10):941–944

    25   MorellF, RogerA, Reyes L, Cruz MJ,Murio C, MuñozX. Bird fancier’s

    lung: a series of 86 patients. Medicine (Baltimore) 2008;

    87(2):110–130

    26   Blanchet MR, Israël-Assayag E, Cormier Y. Inhibitory effect of 

    nicotine on experimental hypersensitivity pneumonitis in vivo

    and in vitro. Am J Respir Crit Care Med 2004;169(8):903–909

    27   Sopori ML, Kozak W, Savage SM, Geng Y, Kluger MJ. Nicotine-

    induced modulation of T Cell function. Implications for inamma-

    tion and infection. Adv Exp Med Biol 1998;437:279–289

    28   Nizri E, Irony-Tur-SinaiM, LoryO, Orr-Urtreger A, Lavi E, BrennerT.

    Activation of the cholinergic anti-inammatory system by nico-

    tine attenuates neuroinammation via suppression of Th1 and

    Th17 responses. J Immunol 2009;183(10):6681–6688

    29   Ohtsuka Y, Munakata M, Tanimura K, et al. Smoking promotes

    insidious and chronic farmer’s lung disease, and deteriorates the

    clinical outcome. Intern Med 1995;34(10):966–971

    30  Vogelmeier C, Krombach F, Münzing S, et al. Activation of blood

    neutrophils in acute episodes of farmer ’s lung. Am Rev Respir Dis

    1993;148(2):396–400

    31   Laamme C, Israël-Assayag E, Cormier Y. Apoptosis of bronchoal-

    veolar lavage lymphocytes in hypersensitivity pneumonitis. Eur

    Respir J 2003;21(2):225–231

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán552

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    11/12

    32   Barrera L, Mendoza F, Zuñiga J, et al. Functional diversity of T-cell

    subpopulations in subacute and chronic hypersensitivity pneu-

    monitis. Am J Respir Crit Care Med 2008;177(1):44–55

    33  Yamasaki H, Ando M, Brazer W, Center DM, Cruikshank WW.

    Polarized type 1 cytokine prole in bronchoalveolar lavage T cells

    of patients with hypersensitivity pneumonitis. J Immunol

    1999;163(6):3516–3523

    34   Glimcher LH. Trawling for treasure: tales of T-bet. Nat Immunol

    2007;8(5):448–450

    35   Simonian PL,Roark CL,WehrmannF, et al.Th17-polarized immune

    response in a murine model of hypersensitivity pneumonitis and

    lung  brosis. J Immunol 2009;182(1):657–665

    36   Selman M, Pardo A, Barrera L, et al. Gene expression

    proles distinguish idiopathic pulmonary brosis from hypersen-

    sitivity pneumonitis. Am J Respir Crit Care Med 2006;173(2):

    188–198

    37   Lazarevic V, Glimcher LH. T-bet in disease. Nat Immunol 2011;

    12(7):597–606

    38   Kolls JK, Lindén A. Interleukin-17 family members and inamma-

    tion. Immunity 2004;21(4):467–476

    39   Simonian PL, Roark CL, Born WK, O’Brien RL, Fontenot AP.

    Gammadelta T cells and Th17 cytokines in hypersensitivity

    pneumonitis and lung   brosis. Transl Res 2009;154(5):

    222–

    22740  Dakhama A, Hegele RG, Laamme G, Israël-Assayag E, Cormier Y.

    Common respiratory viruses in lower airways of patients with

    acute hypersensitivity pneumonitis. Am J Respir Crit Care Med

    1999;159(4 Pt 1):1316–1322

    41   Udwadia ZF, Wright MJ, McIntosh LG, Leitch AG. Confusing

    serological abnormalities in bird fancier’s lung. BMJ 1990;

    300(6738):1519–1520

    42   Seifert SA, Von Essen S, Jacobitz K, Crouch R, Lintner CP. Organic

    dust toxic syndrome: a review. J Toxicol Clin Toxicol 2003;41

    (2):185–193

    43   Churg A, Muller NL, Flint J, Wright JL. Chronic hypersensitivity

    pneumonitis. Am J Surg Pathol 2006;30(2):201–208

    44   Lacasse Y, Selman M, Costabel U, et al; HP Study Group. Classica-

    tion of hypersensitivity pneumonitis: a hypothesis. Int Arch

    Allergy Immunol 2009;149(2):161–16645   Miyazaki Y, Tateishi T, Akashi T, Ohtani Y, Inase N, Yoshizawa Y.

    Clinical predictors and histologic appearance of acute exacerba-

    tions in chronic hypersensitivity pneumonitis. Chest 2008;

    134(6):1265–1270

    46   Olson AL, Huie TJ, Groshong SD, et al. Acute exacerbations of 

    brotic hypersensitivity pneumonitis: a case series. Chest

    2008;134(4):844–850

    47   Bourke SJ, Carter R, Anderson K, et al. Obstructive airways disease

    in non-smoking subjects with pigeon fanciers’   lung. Clin Exp

    Allergy 1989;19(6):629–632

    48  Selman M, Vargas MH. Airway involvement in hypersensitivity

    pneumonitis. Curr Opin Pulm Med 1998;4(1):9–15

    49  Silva CI, Churg A, Müller NL. Hypersensitivity pneumonitis: spec-

    trum of high-resolution CT and pathologic   ndings. AJR Am J

    Roentgenol 2007;188(2):334–344

    50  Hansell DM, Wells AU, Padley SPG, Müller NL. Hypersensitivity

    pneumonitis: correlation of individual CT patterns with functional

    abnormalities. Radiology 1996;199(1):123–128

    51   Chung MH,EdinburghKJ, Webb EM,McCowin M, Webb WR.Mixed

    inltrative and obstructive disease on high-resolution CT: differ-

    ential diagnosis and functional correlates in a consecutive series. J

    Thorac Imaging 2001;16(2):69–75

    52  Franquet T, Hansell DM, Senbanjo T, Remy-Jardin M, Müller NL.

    Lung cysts in subacute hypersensitivity pneumonitis. J Comput

    Assist Tomogr 2003;27(4):475–478

    53   Silva CI, Müller NL, Lynch DA, et al. Chronic hypersensitivity

    pneumonitis: differentiation from idiopathic pulmonary  brosis

    and nonspecic interstitial pneumonia by using thin-section CT.

    Radiology 2008;246(1):288–297

    54   Nakata H, Kimoto T, Nakayama T, Kido M, Miyazaki N, Harada S.

    Diffuse peripheral lung disease: evaluation by high-resolution

    computed tomography. Radiology 1985;157(1):181–185

    55   Buschman DL, Gamsu G, Waldron JA Jr, Klein JS, KingTE Jr. Chronic

    hypersensitivity pneumonitis: use of CT in diagnosis. AJR Am J

    Roentgenol 1992;159(5):957–960

    56   Sahin H, Brown KK, Curran-Everett D, et al. Chronic hypersensi-

    tivity pneumonitis: CT features comparison with pathologic evi-

    dence of  brosis and survival. Radiology 2007;244(2):591–598

    57   Cormier Y, Brown M, Worthy S, Racine G, Müller NL. High-

    resolution computed tomographic characteristics in acute farm-

    er’s lung and in its follow-up. Eur Respir J 2000;16(1):56–60

    58  Malinen AP, Erkinjuntti-Pekkanen RA, Partanen PL, Rytkönen HT,

    Vanninen RL. Long-term sequelae of Farmer’s lung disease in

    HRCT: a 14-year follow-up study of 88 patients and 83 matched

    control farmers. Eur Radiol 2003;13(9):2212–2221

    59   Bourke SJ, Banham SW, McKillop JH, Boyd G. Clearance of 99mTc-

    DTPA in pigeon fancier’s hypersensitivity pneumonitis. Am Rev

    Respir Dis 1990;142(5):1168–1171

    60  McSharry C, Banham SW, Lynch PP, Boyd G. Antibody measure-

    ment in extrinsic allergic alveolitis. Eur J Respir Dis 1984;

    65(4):259–265

    61  Cormier Y, Bélanger J, Durand P. Factors inuencing the develop-

    ment of serum precipitinsto farmer’s lung antigenin Quebec dairy

    farmers. Thorax 1985;40(2):138–142

    62  MendozaF, Baltazares M, RamírezA, etal. Detection of salivary and

    seric IgGand IgA antipooledpigeon sera activities in patients with

    pigeon breeder’s disease. J Clin Lab Anal 1996;10(3):149–154

    63   McSharry C, MacLeod K, McGregor S, et al. Mucosal immunity in

    extrinsic allergic alveolitis: salivary immunoglobulins and anti-

    body against inhaled avian antigens among pigeon breeders. Clin

    Exp Allergy 1999;29(7):957–964

    64   Cormier Y, Létourneau L, Racine G. Signicance of precipitins and

    asymptomatic lymphocytic alveolitis: a 20-yr follow-up. Eur Re-

    spir J 2004;23(4):523–525

    65   Ando M, Konishi K, Yoneda R, Tamura M. Difference in the

    phenotypes of bronchoalveolar lavage lymphocytes in patients

    with summer-type hypersensitivity pneumonitis, farmer’s lung,

    ventilation pneumonitis, and bird fancier’s lung: report of anationwide epidemiologic study in Japan. J Allergy Clin Immunol

    1991;87(5):1002–1009

    66  Drent M, van Velzen-Blad H, Diamant M, Wagenaar SS, Donck-

    erwolck-Bogaert M, van den Bosch JM. Differential diagnostic

    value of plasma cells in bronchoalveolar lavage   uid. Chest

    1993;103(6):1720–1724

    67   Drent M, WagenaarSS, vanVelzen-BladH, Mulder PG, Hoogsteden

    HC,van denBosch JM.Relationship betweenplasma cell levelsand

    prole of bronchoalveolar lavage  uid in patients with subacute

    extrinsic allergic alveolitis. Thorax 1993;48(8):835–839

    68  Haslam PL, Dewar A, Butchers P, Primett ZS, Newman-Taylor A,

    Turner-Warwick M. Mast cells, atypical lymphocytes, and neutro-

    phils in bronchoalveolar lavage in extrinsic allergic alveolitis.

    Comparison with other interstitial lung diseases. Am Rev Respir

    Dis 1987;135(1):35–47

    69  Drent M, van Velzen-Blad H, Diamant M, Wagenaar SS, Hoogste-

    den HC, van den Bosch JM. Bronchoalveolar lavage in extrinsic

    allergic alveolitis: effect of time elapsed since antigen exposure.

    Eur Respir J 1993;6(9):1276–1281

    70 Schildge J, KlarB, Hardung-BackesM. Mast cells in bronchoalveolar

    lavage uid of patients with interstitial lung diseases [in German].

    Pneumologie 2003;57(4):202–207

    71   Drent M, Mulder PGH, Wagenaar SS, Hoogsteden HC, van Velzen-

    Blad H, van den Bosch JM. Differences in BAL   uid variables in

    interstitial lung diseases evaluated by discriminant analysis. Eur

    Respir J 1993;6(6):803–810

    72   Hariri LP, Mino-Kenudson M, Shea B, et al. Distinct histopathology

    of acute onset or abrupt exacerbation of hypersensitivity pneu-

    monitis. Hum Pathol 2012;43(5):660–668

    Seminars in Respiratory and Critical Care Medicine Vol. 33 No. 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán   553

  • 8/8/2019 4 neumonitis por hipersensibilidad 2012 dr. Selman YA LEIDO MUY BUENO.pdf

    12/12

    73  Barrios RJ. Hypersensitivity pneumonitis: histopathology. Arch

    Pathol Lab Med 2008;132(2):199–203

    74   Gaxiola M, Buendía-Roldán I, Mejía M, et al. Morphologicdiversity

    of chronic pigeon breeder’s disease: clinical features and survival.

    Respir Med 2011;105(4):608–614

    75   Reyes CN, Wenzel FJ, Lawton BR, Emanuel DA. The pulmonary

    pathology of farmer’s lung disease. Chest 1982;81(2):142–146

    76   Pérez-Padilla R, Gaxiola MN, Salas J, Mejía M, Ramos C, Selman M.

    Bronchiolitis in chronic pigeon breeder’s disease: morphologic

    evidence of a spectrum of small airway lesions in hypersensitivity

    pneumonitis induced by avian antigens. Chest 1996;110(2):

    371–377

    77   Fukuoka J, Franks TJ, Colby TV, et al. Peribronchiolar metaplasia: a

    common histologic lesion in diffuse lung disease and a rare cause

    of interstitial lung disease: clinicopathologic features of 15 cases.

    Am J Surg Pathol 2005;29(7):948–954

    78   Katzenstein AL, Mukhopadhyay S, Myers JL. Diagnosis of usual

    interstitial pneumonia and distinction from other  brosing inter-

    stitial lung diseases. Hum Pathol 2008;39(9):1275–1294

    79   Churg A, Muller NL, Flint J, Wright JL. Chronic hypersensitivity

    pneumonitis. Am J Surg Pathol 2006;30(2):201–208

    80  Vourlekis JS, Schwarz MI, Cool CD, Tuder RM, King TE, Brown KK.

    Nonspecic interstitial pneumonitis as the sole histologic expres-

    sion of hypersensitivity pneumonitis. Am J Med 2002;112(6):490–493

    81   Reghellin D, Poletti V, Tomassett S, et al. Cathepsin-K is a sensitive

    immunohistochemical marker for detection of micro-granulomas

    in hypersensitivity pneumonitis. Sarcoidosis Vasc Diffuse Lung Dis

    2010;27(1):57–63

    82  Verma H, Nicholson AG, Kerr KM, et al. Alveolar proteinosis with

    hypersensitivity pneumonitis: a new clinical phenotype. Respir-

    ology 2010;15(8):1197–1202

    83   Kokkarinen JI, Tukiainen HO, Terho EO. Effect of corticosteroid

    treatment on the recoveryof pulmonary functionin farmer’s lung.

    Am Rev Respir Dis 1992;145(1):3–5

    84   Ramírez A, Sansores R, Chapela R, et al. Inhaled beclomethasone

    versus oral prednisone. A clinical trial in patients with hypersen-

    sitivity pneumonitis [abstract]. Am J Respir Crit Care Med

    1995;151:A605

    85   Carlsen KH,LeegaardJ, Lund OD,Skjaervik H.Allergic alveolitisin a

    12-year-old boy: treatment with budesonide nebulizing solution.

    Pediatr Pulmonol 1992;12(4):257–259

    86   Tanaka H, Tsunematsu K, Nakamura N, et al. Successful treatment

    of hypersensitivity pneumonitis caused by Grifola frondosa (Mai-

    take) mushroom using a HFA-BDP extra-ne aerosol. Intern Med

    2004;43(8):737–740

    87  Khoor A, Leslie KO, Tazelaar HD, Helmers RA, Colby TV. Diffuse

    pulmonary disease caused by nontuberculous mycobacteria in

    immunocompetent people (hot tub lung). Am J Clin Pathol

    2001;115(5):755–762

    88   Hanak V, Kalra S, AksamitTR, Hartman TE,TazelaarHD, RyuJH. Hot

    tub lung: presenting features and clinical course of 21 patients.

    Respir Med 2006;100(4):610–615

    89   Baughman RP, Judson MA, Teirstein AS, Moller DR, Lower EE.

    Thalidomide for chronic sarcoidosis. Chest 2002;122(1):227–232

    90  Ye Q, Chen B, Tong Z, et al. Thalidomide reduces IL-18, IL-8 and

    TNF-alpha release from alveolar macrophages in interstitial lung

    disease. Eur Respir J 2006;28(4):824–831

    91   Pérez-PadillaR, Salas J, Chapela R, et al. Mortality in Mexicanpatients

    with chronic pigeon breeder’s lung compared with those with usual

    interstitial pneumonia. Am Rev Respir Dis 1993;148(1):49–

    5392   Vourlekis JS, Schwarz MI, Cherniack RM, et al. The effect of 

    pulmonary  brosis on survival in patients with hypersensitivity

    pneumonitis. Am J Med 2004;116(10):662–668

    93   Hanak V, Golbin JM, Hartman TE, Ryu JH. High-resolution CT

    ndings of parenchymal   brosis correlate with prognosis in

    hypersensitivity pneumonitis. Chest 2008;134(1):133–138

    94  Lima MS, Coletta EN, Ferreira RG, et al. Subacute and chronic

    hypersensitivity pneumonitis: histopathological patterns and sur-

    vival. Respir Med 2009;103(4):508–515

    95   BangKM, WeissmanDN, PinheiroGA, Antao VC, Wood JM,Syamlal

    G. Twenty-three years of hypersensitivity pneumonitis mortality

    surveillance in the United States. Am J Ind Med 2006;49(12):

    997–1004

    Seminars in Respiratory and Critical Care Medicine Vol 33 No 5/2012

    Immunopathology, Diagnosis, and Management of Hypersensitivity Pneumonitis   Selman, Buendía-Roldán554