Prévia do material em texto
442 | Allergy. 2022;77:442–453.wileyonlinelibrary.com/journal/all 1 | INTRODUC TION Hypersensitivity pneumonitis (HP) is an immune- mediated inter- stitial lung disease (ILD) caused by an inhalational exposure to low- molecular weight compounds, which occurs in susceptible in- dividuals. Hypersensitivity pneumonitis may present in two forms: acute, predominantly inflammatory HP (non- fibrotic HP), and chronic or fibrotic HP (fibrotic HP).1 Some patients will experience a self- limiting acute HP course, whereas others, given sufficient time and exposure, will progress from non- fibrotic to fibrotic dis- ease, and still other patients may present with established fibrotic HP without a clear acute episode of exposure. This suggests that the pathophysiology of HP is contributed by complex pathways of antigen exposure, aberrant immunological mechanisms, and genetic predispositions. 1.1 | Epidemiology Reported estimates of the incidence of HP vary across popula- tions, with 1– 3 per 100,000 people per year (including children) in Denmark,2 11.5 per 100,000 people over 65 years per year in the USA,3 and up to 30 per 100,000 people per year in New Mexico.4 HP is the third most common ILD after idiopathic pulmonary fibro- sis (IPF) and connective tissue disease- related interstitial lung dis- ease (CTD- ILD).5 The estimated prevalence of HP is higher in at- risk Received: 5 June 2021 | Accepted: 21 July 2021 DOI: 10.1111/all.15017 R E V I E W A R T I C L E Hypersensitivity pneumonitis: Current concepts in pathogenesis, diagnosis, and treatment Hayley Barnes1,2 | Lauren Troy3,4 | Cathryn T. Lee5 | Anne Sperling5 | Mary Strek5 | Ian Glaspole1,2 © 2021 European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd 1Central Clinical School, Monash University, Melbourne, VIC, Australia 2Alfred Hospital, Melbourne, VIC, Australia 3Royal Prince Alfred Hospital, Sydney, NSW, Australia 4University of Sydney, Sydney, NSW, Australia 5Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA Correspondence Hayley Barnes, Department of Respiratory Medicine, Alfred Health, 34 Commercial Rd, Melbourne, Australia. Email: Hayley.Barnes@monash.edu Abstract Hypersensitivity pneumonitis is an immune- mediated interstitial lung disease caused by an aberrant response to an inhaled exposure, which results in mostly T cell– mediated inflammation, granuloma formation, and fibrosis in some cases. HP is diagnosed by exposure identification, HRCT findings of ground- glass opacities, cen- trilobular nodules, and mosaic attenuation, with traction bronchiectasis and hon- eycombing in fibrotic cases. Additional testing including serum IgG testing for the presence of antigen exposure, bronchoalveolar lavage lymphocytosis, and lung biopsy demonstrating granulomas, inflammation, and fibrosis, increases the diagnostic con- fidence. Treatment for HP includes avoidance of the implicated exposure, immuno- suppression, and anti- fibrotic therapy in select cases. This narrative review presents the recent literature in the understanding of the immunopathological mechanisms, diagnosis, and treatment of HP. K E Y W O R D S extrinsic allergic alveolitis, interstitial pneumonia, occupational lung disease www.wileyonlinelibrary.com/journal/all mailto: https://orcid.org/0000-0002-7615-4191 mailto:Hayley.Barnes@monash.edu http://crossmark.crossref.org/dialog/?doi=10.1111%2Fall.15017&domain=pdf&date_stamp=2021-07-29 | 443BARNES Et Al. populations, with cross- sectional studies estimating the prevalence of HP in farmers at 1.3– 12.9%,6,7 in bird breeders at 3.7– 10.4%,8,9 and in mushroom workers at 3.5– 29%.10- 13 1.2 | Exposures Certain low- molecular weight compounds have a predilection for causing HP. The first reports of HP were in farmers working with hay containing Saccharomycetes spp. growth,14,15 followed by similar cases in those exposed to maple bark,16 pigeons,17 and mushrooms.18 Since these reports, a multitude of exposures have been reported in association with HP (Table 1). The most commonly implicated expo- sures are birds (up to 30% of HP cohorts) from feather dust (bloom), droppings, and serum, most commonly derived from pigeons, budg- erigars (parakeets), cockatiels, parrots and canaries, and duck and goose down products. Mold due to water damage and microbial and bacterial contamination of air conditioning units and ventilation sys- tems are also common.19 It is estimated that 12– 28% of HP cases are directly attributable to workplace exposures,20 the most common of which is farming. Exposure to other agricultural environments in- cluding mushroom cultivation, organic waste, and greenhouses may also increase the risk of HP. Occupationally or recreationally derived low- molecular weight chemicals including isocyanates, although themselves not immunogenic, can combine with human proteins to form haptens and trigger HP.21 Over time, and due to evolving workplace practices, some exposures have become less frequently seen (paprika splitters, pituitary snuff), and some more frequently reported (metalworking fluids, musical instruments).19,22 Though a multitude of antigens have been reported in association with HP, many share similar properties; small (<3 µm) enough to be inhaled into the distal bronchi and alveoli, clearance via local lymphatic drainage inducing an IgG antibody response, capacity to activate complement pathways (eg, cell walls of molds containing β- (1– 3)- d- glucan) and high molecular weight glycoproteins resistant to degra- dation (eg, pigeon intestinal mucin and Trichosporon cutaneum).23 Few studies have examined the exposure duration and intensity required to cause HP, which may vary depending on the implicated antigen. Greater intensity of exposure and persistent exposure may be more likely to lead to progression of disease.24 Few studies have TA B L E 1 HP exposures Name Exposure source Antigen Birds Pigeons, budgerigars, cockatiels, chickens Proteins from bloom, feather, droppings, serum, or down products Mold Previous water damage, rising damp Aspergillus spp., cladosporium spp., Penicillium spp. Farming Moldy hay or silage Saccharomycetes spp., Aspergillus spp. Air conditioner/ humidifier/ swamp cooler Contaminated water Thermoactinomyces spp., Aspergillus spp., Penicillium spp. Hot tub Contaminated water Mycobacterium avium complex, Aspergillus fumigatus, Mycobacterium abscessus, Cladosporium spp. Machine operators/ metalworking fluid Contaminated water- based metalworking fluid Mycobacterium avium complex, Mycobacterium immunogenum Summer- type HP Damp wooden structures Common in Japan Trichosporon spp. Wood Moldy wood- cedar, mahogany, pine, redwood, spruce, cork (suberosis) Alternaria spp., Bacillus subtilis, Mucor spp., Rhizopus spp. Isocyanates Glue, polyurethane foam, paint, plastic, resins, varnishes Isocyanate acid anhydrides Mushroom worker's lung Moldy compost and mushrooms Shitake, bunashimeji, himeji, thermophilic Actinomycetes Gardening/organic waste Soil/ organic waste Aspergillus fumigatus, Saccharopolyspora rectivirgula, thermophilic Actinomycetes Musical instrument HP Wind instruments - saxophones, trombones, bagpipes Mycobacterium chelonae/abscessus, Fusarium spp. Salami workers/ chacineros’ lung Mold dust Aspergillus spp., Cladosporium spp., Penicillium spp. Cheese- washer's lung Mold dust Penicillium spp. Cane sugar (bagassosis) Moldy sugar cane Thermoactinomyces Sacchari Dental products Dental technicians Methyl acrylates Lifeguard lung Contaminated water jets and sprays – lifeguards, pool workers Pseudomonas spp. CPAP machine Contaminated water Molds and fungi 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions)on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense 444 | BARNES Et Al. considered the contribution of multiple exposures and polysensitiza- tion in HP. Where reported, multiple exposures may be present in up to 8% of HP cases.25,26 The inhalational exposure in many HP cases comprises a mixture of antigens with inert dusts, other chemicals, etc., which although associated with an increased risk of fibrosis in other fibrotic lung diseases,27,28 their contribution in HP is poorly elucidated. 2 | IMMUNOPATHOGENESIS The immunological mechanisms resulting in the development of HP are incompletely understood, though believed to result from an aberrant immune response following inhalational exposure to low- molecular weight compounds in susceptible individuals, resulting in inflammation, granuloma formation, and in some cases perpetual damage and fibrosis (Figure 1). Following inhalation of antigenic particles, antigen- presenting cells including activated macrophages and dendritic cells are stim- ulated. The reasons for this are not completely known, but may be due to repeat exposure,21 genetically based differences in structures involved in antigen processing, and adjuvant modulatory factors. Pattern recognition receptors, including toll- like receptors (TLRs), are critical elements in initiating this immune response. TLR- 2 and −6 recognize diacyl lipopeptides and lipoteichoic acid on bacterial cell walls and are upregulated in murine mouse models of HP.29 TLR- 6 is important in granuloma formation (macrophages which develop into epithelioid cells and formation of multinucleated giant cells).30 Other innate receptors including TLR- 9 and dectin- 1 have also been found to contribute to HP in murine models and promote Th17 differentia- tion, which drives further inflammation and fibrosis.31 Dendritic cells are involved in the initiation of the adaptive immune response by mi- grating to the lymph nodes and priming the T- cell immune response. Those with HP have increased levels of dendritic cells in their lung parenchyma after antigen challenge.32 In parallel, inhaled antigen also binds to IgG antibodies which ini- tiates the complement cascade and produces byproducts including C5, which further stimulates macrophages.33 Serum IgG- specific an- tibodies can be used as part of the diagnostic work- up for HP. Activated macrophages, when presented with foreign antigen, release pro- inflammatory, and chemotactic factors, including CCL- 18 which attracts lymphocytes, and IL- 18, MCP- 1, TNF- α, IL- 1, and IL- 6 which upregulates B7 expression and greatly enhances the antigen- presenting capacity of macrophages in response to inhaled antigens.34,35 Production of TNF- α and IL- 1 are responsible for the development of fever and other acute- phase symptoms present in acute HP.35,36 Presentation of foreign antigen by APCs to CD4+ T cells stimu- lates a predominantly CD4+ Th1 immune response. Correspondingly, presence of lymphocytosis in bronchoalveolar lavage fluid is a useful tool in differentiating HP from other ILDs.37 This response, medi- ated by IL- 12 and interferon- γ, produces CXCL10 to recruit T lym- phocytes to the alveolar space, and promote granuloma formation by inducing the functional differentiation and survival of activated macrophages and dendritic cells.38 The immunologically mediated pathways leading to HP are shared with other granulomatous lung diseases, including chronic beryllium disease and sarcoidosis. These common mechanisms include a response to an inhaled antigen, lym- phocytic alveolitis, granuloma formation, and fibrosis. Differences in the nature of the exposure and to the types of causative antigens, genetic predispositions, and signaling pathways within the inflam- matory response may explain the subsequent histopathological and radiological differences in HP and other granulomatous diseases (characterized by poorly formed granulomas accompanied by cen- trilobular, bronchiolocentric, and peribronchial interstitial inflam- mation in HP, compared with well- formed granulomas involving the perilymphatic and peribronchovascular interstitium in sarcoidosis.39 The transition from an acute, inflammatory process to a chronic, fibrotic process in HP is modulated in part by a switch from a Th1 to Th2 inflammatory response, inhibition of regulatory T cells, and upregulation of NKT cells. It also remains to be elucidated whether an inflammatory phase always precedes fibrosis, or whether sep- arate pathways shared with other fibrotic lung diseases are more prominent in fibrotic HP. Regulatory T- cell inhibition contributes to both a proliferation of inflammation and a switch to a Th2 response via a loss of Foxp3 expression.40 Later in the course of the disease, IL- 6 produced by activated macrophages promotes differentiation of B cells to plasma cells and maturation of cytotoxic CD8+ T cells.41 Although less prominent than CD4+ T cells, CD8+ T cells play an im- portant role in cell lysis, and the development of CD8+CD56+ NKT cells, potent inducers of interferon- γ, promotes differentiation from a Th1 to Th2 response.42 Th2 cytokines including IL- 4, IL- 13, and an increase in CCR4 (a Th2 chemokine receptor) and reduction in CXCR3 (a Th1 chemokine receptor) induce fibroblast proliferation and collagen production. Such changes are found in BAL fluid analysis in those with HP.43 Fibroblast proliferation is also induced by IL- 17, produced by Th17 cells, neutrophils, and expression of CD103+ on dendritic cells.44 Similar to other ILDs including IPF, fibrosis is also perpetuated by persistent inflammation in the interstitial and alveolar spaces, re- sulting in epithelial damage and destruction of the alveolar- capillary basement membrane. Fibroblasts, activated by profibrotic cyto- kines, differentiate into myofibroblasts, and migrate to the alveolar space, predominantly driven by TGF- β. Fibroblasts produce extra- cellular matrix, leading to lung tissue remodeling, resulting in tissue stiffness and hypoxia which in turn upregulates profibrotic pathways and perpetuates fibrogenesis.45 2.1 | Genetic risk factors Common and rare genetic variants have been identified with greater frequency in those with HP and especially fibrotic HP, which aids in determining risk and increases understanding of disease patho- genesis. Increased susceptibility to HP has been associated with gene variants located within the major histocompatibility complex, 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense | 445BARNES Et Al. including the TNF- gene promotor region, correlating with higher TNF production perpetuating inflammation, and HLA- DRB1- related increases in peptide affinity to the HLA- DR4 molecular and T- cell receptor recognition,46 HLA- DR3 (bird- related HP), HLA- DQ3 (summer- type HP), and HLA- A, - B, and - C loci (farmer's lung).46 Similar to IPF, MUC5B gain of function alleles have been detected with greater frequency in chronic HP compared to healthy con- trols.47 Overexpression of MUC5B in the respiratory bronchioles re- sults in mucociliary dysfunction and disruption of the normal repair mechanisms of the lung. Protein- altering telomere- related gene vari- ants including TERT, RTEL, and PARN have been identified in genetic studies of those with HP.48 Telomere dysfunction promotes DNA damage pathways, cellular senescence, inappropriate apoptosis, and stimulation of lung remodeling and fibrosis.49 2.2 | Other risk factors Previous respiratory tract infections (Epstein- Barr virus, human her- pesvirus7 and 8, cytomegalovirus, parvovirus 19)50 are associated with an increased risk of developing HP by increasing MHC II ex- pression on alveolar macrophages, increasing production of TNF, IL- 1, 6, IFN- α, and RANTES, and may promote fibrosis through epithelial cell injury, upregulation of TGF- β, and promotion of epithelial- mesenchymal transition.51 Exposure to organochlorine and carbamate pesticides has been associated with farming- related HP, possibly due to their putative role in upregulation of pro- inflammatory cytokines TNF- α, IL- 2, and 4.6 Smoking paradoxically reduces the risk of developing HP (likely because nicotine attenuates the production of IL- 1, TNF, and macrophage phagocytosis); how- ever, those who have HP who persistently smoke are more likely to develop worse outcomes.52 More recent studies have demonstrated an increase in bacterial burden and altered microbial composition in the lower airways of those with CHP compared to healthy sub- jects,53 which in other fibrotic lung diseases have been found to be associated with risk of progression.54 Further research on the role of the lung microbiome in HP is imperative. Our understanding of the immunopathogenesis of HP has been derived in part from animal models (initially rabbits, guinea pigs, and mice) and later murine knockout models, using intratracheal F I G U R E 1 Immunopathogenesis of HP in the lung. Inhaled antigens interact with antigen- presenting cells (macrophages, dendritic cells), via pattern recognition receptors including toll- like receptor 2, 6, 9. APCs stimulate a Th1 response, enhanced by cytokine and chemokine production. Neutrophils are present in early disease. In parallel, stimulated B cells (plasma cells) produce IgG antibodies which initiates the complement cascade and further stimulates macrophages. Macrophages fuse to multinucleated giant cells and epithelioid cells to form granulomas, mediated by Th1 cytokine production. Chemotactic factors produced by granulomas, a greater Th2 to Th1 response, a decrease in regulatory T- cell response, CD8+ T- cell production, and Th17 differentiation (partly induced by CD103+ on dendritic cells) promotes fibroblast proliferation. Fibroblasts differentiate into myofibroblasts, produce collagen and extracellular matrix 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense 446 | BARNES Et Al. distillation of antigen directly to the animal, or adoptive transfer of T cells to distinguish direct effects of the antigen from indirect ef- fects of the immune response.55 Murine models are useful, however, have limited ability to induce granulomas and may not reflect the complexity of human disease behavior. Human BAL and lung biopsy analysis have confirmed and provided additional understanding of HP mechanisms. More recently, technological advances in genomics, next- generation sequencing, proteomics, and metabolomics56 have and will continue to provide a more nuanced and detailed under- standing of HP immunopathogenesis. 3 | DIAGNOSIS The diagnosis of HP is made through a combination of clinical his- tory (including exposure assessment), suggestive HRCT findings, and where required histopathological material, ideally presented at an interstitial lung disease multi- disciplinary discussion (MDD). The MDD should consider the variable contribution of different diagnos- tic tests dependent on clinical phenotype (ie, BAL lymphocytosis may be less useful in fibrotic HP) and should not only consider what is required for a high confidence diagnosis of HP, but also aim to characterize an inflammatory, fibrotic, or mixed phenotype to aid treatment choices and prognostication.57 3.1 | Clinical assessment A thorough assessment for inhalational exposures is essential in the diagnosis of HP, both to identify an inciting antigen for diagnostic confidence and for exposure mitigation, as lack of exposure identi- fication and ongoing exposure portends a poorer survival.58 A thor- ough clinical history should be undertaken including assessment of known HP exposures. Several HP- specific exposure questionnaires have been proposed59,60; however, none have as yet been clinically validated and should be tailored to consider geographical differ- ences in exposures and climate. For any potential exposure elicited, consideration should be given to the duration, extent, and frequency of exposure, and their relationship to symptoms. Dyspnea and cough are the most common clinical symptoms of HP. Other symptoms include chest tightness, weight loss, and rhi- nitis.8,61,62 Fever and malaise are common symptoms of acute HP. Physical examination most commonly reveals crackles, clubbing (7– 50% patients) and occasionally squawks (inspiratory lung sounds with a mixture of musical and non- musical components), suggestive of small airways disease.63 Patients may also present with acute ex- acerbations (acute respiratory deterioration with new ground- glass opacities on CT, not due to any other cause), with or without re- exposure to the inciting antigen. Similar to other fibrotic ILDs, HP exacerbations are more common in those with a UIP- like pattern and portend poor survival.64,65 Restriction (particularly a reduced forced vital capacity; FVC) is the most common physiological abnormality found on pulmonary lung function tests, followed by a reduction in diffusing capacity for carbon monoxide (DLCO).66- 68 Such parameters may be used to demonstrate response to treatment, or predict progressive fibrosing disease (>10% decline in FVC% predicted over 24 months or 5– 10% decline in FVC% predicted with worsening symptoms and/or pro- gression on CT) which portends poor survival.69 Obstructive pat- terns are also seen in cases with bronchiolitis or emphysema, and airway responsiveness may also be present.70 3.2 | Additional antigen testing Serum- specific IgG tests assess the presence of a specific IgG anti- body which interacts with a pre- formed test antigen (eg, bird drop- pings, specific molds), leading to the production of antigen- antibody complexes. Test performance variation may occur due to differences in the technique used (Ouchterlony or passive double immunodif- fusion technique, electrosyneresis, or ELISA), and the methods by which the antigens are produced (bulk versus local antigen collec- tion versus bespoke personalized antigen derivation).71 Commercial antigen tests (most commonly Aspergillus fumigatus, Aspergillus niger, Aspergillus flavus, Thermoactinomyces vulgaris, Micropolyspora faeni, and parrot, pigeon, parakeet serum) are available in some countries. The test performance characteristics also vary depending on the control population; serum IgG testing in HP patients compared to other ILD patients has a sensitivity of 83% and specificity of 68%; when HP is compared to exposed asymptomatic controls the sensi- tivity is 90% and specificity 91%; and unexposed controls the sensi- tivity is 93% and specificity 100%.72 Serum IgG testing can be most useful when a clinical history reveals more than one active exposure, or where no exposure is revealed73; however, a positive serum IgG does not confirm the diagnosis of HP, nor does a negative serum IgG exclude it. Specific inhalational challenge involves the graded exposure to the causative antigen in a controlled laboratory setting, and a positive test results from a 15% reduction in DLCO% to 20% re- duction in FVC%, relative hypoxemia (decline in oxygen satura- tions by 3%), rise in temperature (by 0.5℃), or clinical symptoms (cough, dyspnea).74 Though few studies have been performed to assess its use, the sensitivityis estimated at 73% and specific- ity 84%.71 Specific inhalational challenge is limited by set up bur- den, lack of test standardization, and potential side effects and is therefore used in few centers. Skin patch testing is not routinely used in HP. In select cases, occupational and environmental assessment may be employed. This involves more detailed history taking, and a home or job site visit to inspect ventilation, water damage, and specific elicited sources of exposure. Surface and air samples may be taken for testing, and bespoke antibody testing may be utilized.71 The ad- ditive benefits are yet to be fully elucidated, though it may identify exposures in antigen- indeterminate disease or distinguish between several potential exposures (eg, bird in the home and hay in the workplace).73 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense | 447BARNES Et Al. 3.3 | Radiology High- resolution computed tomography (HRCT) with inspiratory and expiratory films form an essential component of HP diagnosis and prognosis.66 Compared to other ILDs, those with HP typically exhibit diffuse parenchymal ground- glass opacities, centrilobular nodules, mosaic attenuation, and air trapping on expiratory films, usually in a craniocaudal distribution (Figure 2).66 Airspace consolidation and lung cysts may also be present. In those with fibrotic HP, ad- ditional features of coarse reticulation, traction bronchiectasis, and occasionally honeycombing may be present66 and suggest a poorer prognosis.47 3.4 | Bronchoalveolar lavage (BAL) Limitations in the specificity of HRCT findings and uncertain anti- gen exposure will necessitate further diagnostic evaluation in many ILD patients. Bronchoalveolar lavage (BAL) is commonly performed as an adjunctive investigation to enhance diagnostic confidence.66 Bronchoscopically obtained BAL fluid can be analyzed for nucle- ated immune cells including alveolar macrophages, lymphocytes, neutrophils, and eosinophils. BAL fluid lymphocytosis, defined as lymphocyte percentage >15%, is common in HP.66,75 In non- fibrotic HP, lymphocyte counts may exceed 50%.75,76 In fibrotic HP, more modest BAL fluid lymphocyte elevations are observed and levels may even be normal, particularly in smoking or elderly patients.77,78 In the recently published ATS/JRS/ALAT HP guidelines, an expert panel agreed that 30% BAL fluid lymphocytosis was a reasonable threshold for distinguishing between HP and IPF or sarcoidosis.66 In addition to cytological analysis, BAL is useful for ruling in or out in- fectious pathology of the respiratory tract, including Mycobacterium species. 3.5 | Lung biopsy Histopathologic information may be required when diagnosis re- mains uncertain, following baseline investigations. The three meth- ods for obtaining lung tissue are transbronchial forceps lung biopsy (TBB), transbronchial lung cryobiopsy (TBLC), and surgical lung bi- opsy (SLB). With a diagnostic yield of only 37% (95% CI 32– 42), TBB is less favored than other modalities.66 TBLC has greater diagnostic power than TBB, with an estimated diagnostic yield of 82% (95% CI 78– 86%) for HP.79 TBLC carries increased risk of airway bleeding and pneumothorax compared with TBB, requiring procedural exper- tise and careful patient selection.80,81 SLB is the reference standard for tissue sampling, with a diagnostic yield of 96% (95% CI 90– 100%) in HP, however, also carries a risk of airway bleeding, pneumotho- rax, post- procedural exacerbations, and death.66 The risk is greater in those requiring in- patient biopsy for rapidly progressive disease, male sex, increasing age (over 65 years), multiple comorbidities in- cluding pulmonary hypertension, and low baseline lung function (FVC% predicted <50% and/or DLCO% predicted <35%).66,82 The decision to proceed to biopsy and the choice of technique are neces- sarily determined by available resources and patient suitability, and should be discussed in an MDD prior to undertaking the procedure. 3.6 | Histopathology Non- fibrotic HP is characterized by bronchiolocentric expansion of the interstitium with lymphocytes, occasional plasma cells and eosinophils, and scant lymphoid aggregates without germinal cent- ers.66 Chronic cellular bronchiolitis and peribronchiolar granuloma- tous inflammation are also key findings, the latter typified by loosely formed clusters of epithelioid and multinucleated macrophages, and isolated giant cells with non- specific inclusions such as cholesterol clefts as seen in other interstitial diseases affecting the distal air- ways (Figure 3).83 In fibrotic HP similar features are also observed; however, the in- filtrate is paucicellular and there is associated fibrosis. There is often significant overlap with the usual interstitial pneumonia pattern seen with IPF, including features such as fibroblast foci and subpleu- ral honeycomb cysts.83 Occasionally, other disease patterns, such as F I G U R E 2 A, High- resolution CT expiratory image of inflammatory HP with ground- glass opacities and centrilobular nodules with marked air trapping. B, Fibrotic HP with traction bronchiectasis and reticulation, with mild mosaic attenuation and centrilobular ground- glass nodules (A) (B) 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense 448 | BARNES Et Al. non- specific interstitial pneumonia (NSIP) or organizing pneumonia, are observed in HP. Idiopathic forms of these diseases are important differential diagnoses for HP as are other types of granulomatous ILD such as sarcoidosis, common variable immune deficiency (CVID), and infections due to mycobacteria, fungi, and certain respiratory viruses. 4 | TRE ATMENT 4.1 | Exposure remediation The initial management for those diagnosed with HP includes the identification and remediation of the implicated exposure. Ongoing exposure is associated with further decline in respiratory function, and improvement with exposure remediation may help confirm the diagnosis.69 No evidence- based guidelines specific to HP on how to mitigate exposures currently exist, though case reports suggest removal of the implicated exposure may not be sufficient to clear all antigenic particles from the environment without additional deep cleaning.84,85 4.2 | Immunosuppression While exposure remediation is the most important management strategy, in many patients with HP an antigen is not identified. Persistent respiratory symptoms, lung function decline, and/or ra- diographic progression may occur even with antigen avoidance and prompt the consideration of therapy with anti- inflammatory or anti- fibrotic medications.1,86 The immunosuppressive agents used in HP include systemic corticosteroids (which inhibits the synthesis of inflammatory cytokines through blocking the function of nuclear factor kappa B (ηf- κβ) and activator protein 1 (AP- 1)),87 mycopheno- late mofetil (which inhibits B- and T- cell proliferation and has some anti- fibrotic properties),88 azathioprine (which inhibits B- and T- cell proliferation and inhibits the costimulatory CD28 binding of T cells with APCs)89 and rituximab (which depletes CD 20+ B lymphocytes and inhibits T- cell co- stimulation).90 Demonstrated efficacy of these therapies in rigorous clinical trials, however, is lacking. The anti- fibrotic agent nintedanib(a tyrosine kinase inhibitor which targets multiple growth factor receptors, including vascular endothelial growth factor, fibroblast growth factor, and platelet- derived growth factor) has been shown to be efficacious in a randomized, placebo- controlled trial in patients with progressive fibrosing ILD including HP.91 Acute exacerbations in patients with HP are typically treated with intravenous corticosteroids based on anecdotal evidence of benefit in some cases. Corticosteroids are considered the first- line medical therapy for those in whom the HP does not improve or resolve with successful antigen remediation. In one small randomized study in patients with acute farmer's lung,92 prednisolone compared to placebo resulted in improvement in DLCO at one month but not FVC or DLCO at five years with recurrent farmer's lung noted more commonly in the corticosteroid- treated group. A retrospective study of immunosup- pressive therapy in patients with chronic HP showed that patients treated with prednisone alone had more frequent treatment- related adverse events than those who also received azathioprine or my- cophenolate mofetil.93 This may have been due to the dose reduc- tion in prednisone afforded by the use of these agents as was seen in a recent multicenter retrospective investigation.94 In this study, treatment with either azathioprine or mycophenolate mofetil also resulted in improvement in DLCO with stable FVC after one year of therapy. In patients with chronic HP treated with mycophenolate mofetil, an improved survival was noted in those with longer telo- mere length compared to those with short telomeres.95 The benefit of rituximab in HP has been noted in a case report and uncontrolled studies.96- 98 A recent small retrospective study suggested that rit- uximab was associated with stabilization to modest improvement in lung function in chronic HP.98 In patients with progressive fibros- ing ILD, a subset of whom had HP (n=173), nintedanib was shown to slow the decline in FVC compared to placebo.91 This may be of particular benefit in those with HP and UIP type pulmonary fibro- sis for whom anti- inflammatory therapy may result in little or no improvement. Rigorous trials are urgently needed to confirm the efficacy re- ported in retrospective studies, including when immunosuppression is beneficial and whether it might be harmful in the presence of fi- brosis or known genetic variants. Further studies are also needed to determine the additive benefit of anti- fibrotic therapy and for novel therapies. 4.3 | Non- pharmacological treatments In HP patients, significant physical deconditioning can develop due to avoidance of activity and generalized sarcopenia due to chronic inflammation, altered hormonal profiles, and metabolic F I G U R E 3 High power surgical lung biopsy specimen demonstrating a small aggregate of histiocytes and giant cells, with some cholesterol crystals within the cytoplasm 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense | 449BARNES Et Al. derangement.99 Furthermore, corticosteroid use can lead to proxi- mal myopathy. Many patients will benefit from outpatient pul- monary rehabilitation100,101; shown to improve six- minute walk distance (6MWD), dyspnea scores, and quality of life in mixed ILD populations.102 Supplemental oxygen can improve endur- ance and is advised for all ILD patients with resting hypoxemia, and for those with exercise- induced hypoxemia during pulmonary rehabilitation.101,103 A select group of HP patients with progressive disease may be suitable for lung transplantation. Transplanted HP patients appear to have better outcomes than those with IPF, with lower rates of acute rejection in the HP group in one retrospective series.1,104 Notably, HP may recur in the allograft in a small proportion of patients if the exposure is not removed.104 For patients with progressive disease who are not transplant- eligible, palliative care is an important but currently underutilized service for management of complex needs.105,106 A large burden of respiratory symptoms can be reduced with timely introduction of palliative medications, behavioral strategies, specialist nursing, and psychological support delivered by community- based expert clinicians.107 5 | NATUR AL HISTORY The natural history of HP is dependent on the underlying disease phenotype. Markers of inflammatory HP including ground- glass opacities on HRCT are associated with a better prognosis (reduced hazard ratio for mortality of 0.31108). Conversely, those with fibro- sis on CT have a poorer survival (7.95 years; honeycomb fibrosis 2.76 years; no fibrosis >14.73 years).25,108-110 Similar findings have been demonstrated using automated CT analysis, as well as demonstrating in one study that pulmonary ves- sel volume was a separate predictor of survival, and in another that automated analysis using CALIPER was a stronger predictor of mor- tality than visual CT analysis.111,112 Older age, male gender, lower baseline pulmonary physiology, and a decline in FVC >10% are poor prognostic indicators.3,69 Pulmonary hypertension also portends a poorer survival (23 months when present compared to 98 months when absent).113 Patients with HP experience greater complexity of care than other forms of ILDs and are more likely than their ILD counter- parts to undergo invasive surgical lung biopsy and other additional tests. Conversely, clinical trial access is less commonly available for HP than compared with IPF or other ILDs.114 Health- related quality of life is worse in those with HP, even when controlling for age and pulmonary impairment,115 found to be in part associated with the complex psychosocial problems associated with antigen identification and avoidance.116 Mood disorders may be present in up to 50% of those with HP, and depression in 28%.24 Degree of dyspnea and extent of comorbidities have been shown to con- tribute to mood disorder, while dyspnea and fatigue impact on qual- ity of life.102 6 | BIOMARKERS FOR PROGNOSTIC ATION A number of prognostically significant biomarkers have been de- scribed for HP.117 In addition to CT features of fibrosis as described above, the next most frequently utilized biomarker is BAL lympho- cyte counts, with higher % lymphocytosis being associated with more inflammatory forms of presentation and lower counts carrying a worse prognosis.24 Markers of epithelial and pneumocyte damage (KL- 6, SP- D), inflammatory mediators (CCL17), and protein deposi- tion (periostin) are also associated with mortality.118- 121 CXCL13 (promotes B lymphocyte migration and granuloma formation), ma- trix metalloproteinase- 7 (responsible for extracellular matrix deg- radation and processing of growth factors including FAS ligand, β4 integrin, E- cadherin, plasminogen, transmembrane tumor necrosis factor α (pro- TNF- α), and osteopontin),122 and VCAM- 1 (known to stimulate fibroblast proliferation in IPF)123 are also associated with increased risk of fibrosis and mortality. Genetic predictors of worse outcome include shorter telomere length and the MUC5B rs35705950 single nucleotide polymor- phism being associated with more severe fibrosis on CT, and shorter telomere length with reduced survival.47 Differences in the trigger- ing environmental antigen have not been shown to generally alter outlook, but where the antigen cannot be identified, prognosis is worse.58 Additionally, where ongoing environmental exposure is higher, progression appears more likely.124 A significant minority of those with HP have features or serologic markers consistent with autoimmune disease.These autoimmune features may increase the risk of mortality by fourfold in HP patients when adjusted for other known predictors of survival.125 7 | CONCLUSIONS AND FUTURE RESE ARCH PERSPEC TIVES Hypersensitivity pneumonitis is a commonly encountered ILD, and its recognition and prevalence have increased over time. Large, mul- ticenter cohort registry- based studies have contributed to identi- fying clinical, radiological, and pathological features leading to the development of common key diagnostic criteria, and to the evolution of more useful HP phenotypes (non- fibrotic and fibrotic, versus the previous classification of acute; subacute; chronic) which aid in prog- nostication and treatment making decisions. Our understanding of the complex immunological drivers of dis- ease have been greatly enhanced by the new developments in tech- nology (including RNA sequencing and SNP analysis) and revealed pathways unique to HP and shared pathways with other ILDs lead- ing to fibrosis. In turn, non- invasive biomarkers of disease are being identified, with potential for enhanced diagnostic and prognostic accuracy in the future. The disease trajectory for many patients with HP remains poor, despite treatment. Given that immunosuppression may be dele- terious in certain types of ILDs,126 rigorous trials are needed to 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense 450 | BARNES Et Al. determine the role of immunosuppression in treating the various phenotypes of HP. Anti- fibrotic therapy in HP appears likely to have a role for fibrotic HP, but further confirmation and studies of anti- fibrotics are needed. While antigen identification and remediation are vital, antigen identification remains poor,58 providing a further need for future research. ACKNOWLEDG MENTS We thank Dr Kirk Jones (UCSF) for the provision of histopathology slides, and Dr Brett Elicker (UCSF) for the provision of HRCT images. CONFLIC T OF INTERE S T Dr. Strek conducts clinical research studies for Boehringer- Ingelheim and Galapagos, and has received honoraria for consulting and medi- cal writing from Boehringer- Ingelheim and honoraria for serving on an adjudication committee for FibroGen. AUTHOR CONTRIBUTIONS HB and IG conceived the article. HB, CL, LT, AS, MS, and IG contrib- uted to the draft and approved the final manuscript. ORCID Hayley Barnes https://orcid.org/0000-0002-7615-4191 R E FE R E N C E S 1. Vasakova M, Morell F, Walsh S, Leslie K, Raghu G. Hypersensitivity Pneumonitis: Perspectives in Diagnosis and Management. Am J Respir Crit Care Med 2017;196(6):680- 689. 2. Rittig AH, Hilberg O, Ibsen R, Lokke A. Incidence, comorbid- ity and survival rate of hypersensitivity pneumonitis: a national population- based study. ERJ Open Res. 2019;5(4):00259- 02018. 3. Fernandez Perez ER, Kong AM, Raimundo K, Koelsch TL, Kulkarni R, Cole AL. Epidemiology of Hypersensitivity Pneumonitis among an Insured Population in the United States: A Claims- based Cohort Analysis. Ann Am Thorac Soc 2018;15(4):460- 469. 4. Coultas DB, Zumwalt RE, Black WC, Sobonya RE. The epide- miology of interstitial lung diseases. Am J Respir Crit Care Med 1994;150(4):967- 972. 5. Cottin V, Hirani NA, Hotchkin DL, et al. Presentation, diagnosis and clinical course of the spectrum of progressive- fibrosing inter- stitial lung diseases. Eur Respir Rev 2018;27(150). 6. 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. 7. Stanford CF, Hall G, Chivers A, Martin B, Nicholls DP, Evans J. Farmer's lung in Northern Ireland. Br J Ind Med 1990;47(5):314- 316. 8. Charitopoulos K, Gioulekas D, Sichletidis L, Chloros D, Vamvakopoulou V, Zarogoulidis K. Hypoxemia: an early indica- tion of pigeon breeders’ disease. Clinical and laboratory findings among pigeon breeders in the Salonica area. J Investig Allergol Clin Immunol 2005;15(3):211- 215. 9. Banham SW, McSharry C, Lynch PP, Boyd G. Relationships between avian exposure, humoral immune response, and pi- geon breeders’ disease among Scottish pigeon fanciers. Thorax 1986;41(4):274- 278. 10. Betz B. Diseases of the airways in oyster mushroom cultivators. Pneumologie, Sonderheft. 1990;44(1):339- 340. 11. Konishi K, Mouri T, Chida E, Kubota K, Nakadate T, Tamura M. Chronic type hypersensitivity pneumonitis. Nihon Kyobu Shikkan Gakkai Zasshi. 1993;31(Suppl):62- 66. 12. Tanaka H, Saikai T, Sugawara H, et al. Three- year follow- up study of allergy in workers in a mushroom factory. Respir Med 2001;95(12):943- 948. 13. Tsushima K, Fujimoto K, Yoshikawa S, Kawakami S, Koizumi T, Kubo K. Hypersensitivity pneumonitis due to Bunashimeji mushrooms in the mushroom industry. Int Arch Allergy Immunol 2005;137(3):241- 248. 14. Campbell JM. Acute Symptoms Following Work with Hay. BMJ 1932;2:1143- 1144. 15. Fawcitt R. Fungoid Conditions of the Lung— Part I. Brit J Radiol. 1936;9(99):172- 195. 16. Towey JW, Sweany HC, Huron WH. Severe bronchial asthma apparently due to fungus spores found in maple bark. JAMA 1932;99(6):453- 459. 17. Feldman HA, Sabin AB. Pneumonitis of unknown aetiology in a group of men exposed to pigeon excreta. J Clin Invest 1948;27:533. 18. Bringhurst LS, Byrne RN, Gershon- Cohen J. Respiratory disease of mushroom workers; farmer's lung. J Am Med Assoc 1959;171:15- 18. 19. Barnes H, Lu J, Glaspole I, Collard HR, Johannson KA. Exposures and Associations with Clinical Phenotypes in Hypersensitivity Pneumonitis: A Scoping Review. Respir Med 2021;184:106444. 20. Blanc PD, Annesi- Maesano I, Balmes JR, et al. The occupational burden of nonmalignant respiratory diseases. an official american thoracic society and european respiratory society statement. Am J Respir Crit Care Med 2019;199(11):1312- 1334. 21. Selman M, Pardo A, King TE. Hypersensitivity Pneumonitis. Am J Respir Crit Care Med 2012;186(4):314- 324. 22. Quirce S, Vandenplas O, Campo P, et al. Occupational hy- persensitivity pneumonitis: an EAACI position paper. Allergy 2016;71(6):765- 779. 23. Bourke SJ, Dalphin JC, Boyd G, McSharry C, Baldwin CI, Calvert JE. Hypersensitivity pneumonitis: current concepts. Eur Respir J 2001;18(32 suppl):81s. 24. Ojanguren I, Morell F, Ramon MA, et al. Long- term outcomes in chronic hypersensitivity pneumonitis. Allergy 2019;74(5):944- 952. 25. Salisbury ML, Gu T, Murray S, et al. Hypersensitivity pneumonitis: radiologic phenotypes are associated with distinct survival time and pulmonary function trajectory. Chest 2019;155(4):699- 711. 26. Barnes H, Olin AC, Toren K, et al. Occupation versus environmen- tal factors in hypersensitivity pneumonitis: population attribut- able fraction. ERJ Open Res. 2020;6(4):00374- 02020. 27. Paolocci G, Folletti I, Torén K, et al. Occupational risk factors for idiopathic pulmonary fibrosis in Southern Europe: a case- control study. BMC Pulmonary Medicine. 2018;18(1):75. 28. Abramson MJ, Murambadoro T, Alif SM, et al. Occupational and environmental risk factors for idiopathic pulmonary fibrosis in Australia: case– control study. Thorax 2020;75(10):864. 29. Joshi AD, Fong DJ, Oak SR, et al. Interleukin- 17- mediated immu- nopathogenesis in experimental hypersensitivity pneumonitis. Am J Respir Crit Care Med 2009;179(8):705- 716. 30. Suga M, Yamasaki H, Nakagawa K, Kohrogi H, Ando M. Mechanisms accounting for granulomatous responses in hypersensitivity pneu- monitis. Sarcoidosis Vasc Diffuse Lung Dis. 1997;14(2):131- 138. 31. Simonian PL, Roark CL, Wehrmann F, et al. Th17- polarizedimmune response in a murine model of hypersensitivity pneumonitis and lung fibrosis. J Immunol 2009;182(1):657- 665. 32. Greer AM, Matthay MA, Kukreja J, et al. Accumulation of BDCA1(+) dendritic cells in interstitial fibrotic lung diseases and Th2- high asthma. PLoS One 2014;9(6):e99084. 33. Shanley TP, Peters JL, Jones ML, Chensue SW, Kunkel SL, Ward PA. Regulatory effects of endogenous interleukin- 1 receptor an- tagonist protein in immunoglobulin G immune complex- induced lung injury. J Clin Invest 1996;97(4):963- 970. 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense https://orcid.org/0000-0002-7615-4191 https://orcid.org/0000-0002-7615-4191 | 451BARNES Et Al. 34. Israel- Assayag E, Dakhama A, Lavigne S, Laviolette M, Cormier Y. Expression of costimulatory molecules on alveolar macro- phages in hypersensitivity pneumonitis. Am J Respir Crit Care Med 1999;159(6):1830- 1834. 35. Patel AM, Ryu JH, Reed CE. Hypersensitivity pneumonitis: current concepts and future questions. J Allergy Clin Immunol 2001;108(5):661- 670. 36. Gudmundsson G, Hunninghake GW. Interferon- gamma is nec- essary for the expression of hypersensitivity pneumonitis. J Clin Invest 1997;99(10):2386- 2390. 37. Yamasaki H, Ando M, Brazer W, Center DM, Cruikshank WW. Polarized type 1 cytokine profile in bronchoalveolar lavage T cells of patients with hypersensitivity pneumonitis. J Immunol 1999;163(6):3516- 3523. 38. Agostini C, Calabrese F, Poletti V, et al. CXCR3/CXCL10 interac- tions in the development of hypersensitivity pneumonitis. Respir Res 2005;6(1):20. 39. Suchankova M, Urban J, Ganovska M, et al. TREM- 1 and TREM- 2 Expression on CD14(+) Cells in Bronchoalveolar Lavage Fluid in Pulmonary Sarcoidosis and Hypersensitivity Pneumonitis in the Context of T Cell Immune Response. Mediators Inflamm 2020;2020:9501617. 40. Girard M, Israel- Assayag E, Cormier Y. Impaired function of regulatory T- cells in hypersensitivity pneumonitis. Eur Respir J 2011;37(3):632- 639. 41. Drent MGJ, Mulder PG, et al. Is the different T helper cell activ- ity in sarcoidosis and extrinsic allergic alveolitis also reflected by the cellular bronchoalveolar lavage fluid profile? Sarcoidosis Vasc Diffuse Lung Dis. 1997;14(1):31- 38. 42. Korosec P, Osolnik K, Kern I, Silar M, Mohorcic K, Kosnik M. Expansion of pulmonary CD8+CD56+ natural killer T- cells in hy- persensitivity pneumonitis. Chest 2007;132(4):1291- 1297. 43. Barrera L, Mendoza F, Zuniga J, et al. Functional diversity of T- cell subpopulations in subacute and chronic hypersensitivity pneumo- nitis. Am J Respir Crit Care Med 2008;177(1):44- 55. 44. Bernatchez E, Langlois A, Brassard J, Flamand N, Marsolais D, Blanchet M- R. Hypersensitivity pneumonitis onset and sever- ity is regulated by CD103 dendritic cell expression. PLoS One 2017;12(6):e0179678. 45. Girard M, Lacasse Y, Cormier Y. Hypersensitivity pneumonitis. Allergy 2009;64(3):322- 334. 46. Falfan- Valencia R, Camarena A, Pineda CL, et al. Genetic sus- ceptibility to multicase hypersensitivity pneumonitis is as- sociated with the TNF- 238 GG genotype of the promoter region and HLA- DRB1*04 bearing HLA haplotypes. Respir Med 2014;108(1):211- 217. 47. Ley B, Newton CA, Arnould I, et al. The MUC5B promoter poly- morphism and telomere length in patients with chronic hypersen- sitivity pneumonitis: an observational cohort- control study. Lancet Respir Med. 2017;5(8):639- 647. 48. Ley B, Torgerson DG, Oldham JM, et al. Rare Protein- Altering Telomere- related Gene Variants in Patients with Chronic Hypersensitivity Pneumonitis. Am J Respir Crit Care Med 2019;200(9):1154- 1163. 49. Mathai SK, Yang IV, Schwarz MI, Schwartz DA. Incorporating genetics into the identification and treatment of Idiopathic Pulmonary Fibrosis. BMC Med 2015;13(1):191. 50. Wuyts WA, Agostini C, Antoniou KM, et al. The patho- genesis of pulmonary fibrosis: a moving target. Eur Respir J 2013;41(5):1207- 1218. 51. Cormier Y, Israël- Assayag E. The role of viruses in the patho- genesis of hypersensitivity pneumonitis. Curr Opin Pulm Med. 2000;6(5):420- 423. 52. Blanchet MR, Israel- Assayag E, Cormier Y. Inhibitory effect of nic- otine on experimental hypersensitivity pneumonitis in vivo and in vitro. Am J Respir Crit Care Med 2004;169(8):903- 909. 53. Invernizzi R, Wu BG, Barnett J, et al. The Respiratory Microbiome in Chronic Hypersensitivity Pneumonitis Is Distinct from That of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2020;203(3):339- 347. 54. Molyneaux PL, Cox MJ, Willis- Owen SAG, et al. The Role of Bacteria in the Pathogenesis and Progression of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2014;190(8):906- 913. 55. Fink JN, Ortega HG, Reynolds HY, et al. Needs and opportunities for research in hypersensitivity pneumonitis. Am J Respir Crit Care Med 2005;171(7):792- 798. 56. Chambers DC, Carew AM, Lukowski SW, Powell JE. Transcriptomics and single- cell RNA- sequencing. Respirology 2019;24(1):29- 36. 57. Fernandez Perez ER, Travis WD, Lynch DA, et al. Diagnosis and Evaluation of Hypersensitivity Pneumonitis: CHEST Guideline and Expert Panel Report. Chest 2021. https://doi.org/10.1016/j. chest.2021.03.066 58. Fernandez Perez ER, Swigris JJ, Forssen AV, et al. Identifying an inciting antigen is associated with improved survival in patients with chronic hypersensitivity pneumonitis. Chest 2013;144(5):1644- 1651. 59. Petnak T, Moua T. Exposure assessment in hypersensitivity pneu- monitis: a comprehensive review and proposed screening ques- tionnaire. ERJ Open Res. 2020;6(3). 60. Barnes H, Morisset J, Molyneaux P, et al. A Systematically Derived Exposure Assessment Instrument for Chronic Hypersensitivity Pneumonitis. Chest 2020;157(6):1506- 1512. 61. Walters GI, Mokhlis JM, Moore VC, et al. Characteristics of hy- persensitivity pneumonitis diagnosed by interstitial and occupa- tional lung disease multi- disciplinary team consensus. Respir Med 2019;155:19- 25. 62. Morell F, Roger A, Reyes L, Cruz MJ, Murio C, Munoz X. Bird fancier's lung: a series of 86 patients. Medicine (Baltimore). 2008;87(2):110- 130. 63. Pereira CAC, Soares MR, Boaventura R, et al. Squawks in intersti- tial lung disease prevalence and causes in a cohort of one thou- sand patients. Medicine (Baltimore). 2019;98(29):e16419. 64. Kang J, Kim YJ, Choe J, Chae EJ, Song JW. Acute exacerbation of fibrotic hypersensitivity pneumonitis: incidence and outcomes. Respir Res 2021;22(1):152. 65. Miyazaki Y, Tateishi T, Akashi T, Ohtani Y, Inase N, Yoshizawa Y. Clinical predictors and histologic appearance of acute ex- acerbations in chronic hypersensitivity pneumonitis. Chest 2008;134(6):1265- 1270. 66. Raghu G, Remy- Jardin M, Ryerson CJ, et al. Diagnosis of Hypersensitivity Pneumonitis in Adults. An Official ATS/JRS/ ALAT Clinical Practice Guideline. Am J Respir Crit Care Med 2020;202(3):e36- e69. 67. Gu JP, Tsai CL, Wysham NG, Huang YT. Chronic hypersensitiv- ity pneumonitis in the southeastern United States: an assess- ment of how clinicians reached the diagnosis. BMC Pulm Med. 2020;20(1):32. 68. Morell F, Roger A, Reyes L, Cruz MJ, Murio C, Muñoz X. Bird fancier's lung: a series of 86 patients. Medicine (Baltimore). 2008;87(2):110- 130. 69. Gimenez A, Storrer K, Kuranishi L, Soares MR, Ferreira RG, Pereira CAC. Change in FVC and survival in chronic fibrotic hypersensitiv- ity pneumonitis. Thorax 2018;73(4):391- 392. 70. Erkinjuntti- Pekkanen R, Kokkarinen JI, Tukiainen HO, Pekkanen J, Husman K, Terho EO. Long- term outcome of pulmonary function in farmer's lung: a 14 year follow- up with matched controls. Eur RespirJ 1997;10(9):2046- 2050. 71. Johannson KA, Barnes H, Bellanger AP, et al. Exposure Assessment Tools for Hypersensitivity Pneumonitis. An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc 2020;17(12):1501- 1509. 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense https://doi.org/10.1016/j.chest.2021.03.066 https://doi.org/10.1016/j.chest.2021.03.066 452 | BARNES Et Al. 72. Jenkins AR, Chua A, Chami H, et al. Questionnaires or Serum Immunoglobulin G Testing in the Diagnosis of Hypersensitivity Pneumonitis among Patients with Interstitial Lung Disease. Ann Am Thorac Soc 2021;18(1):130- 147. 73. Millerick- May ML, Mulks MH, Gerlach J, et al. Hypersensitivity pneumonitis and antigen identification– An alternate approach. Respir Med 2016;112:97- 105. 74. Munoz X, Sanchez- Ortiz M, Torres F, Villar A, Morell F, Cruz MJ. Diagnostic yield of specific inhalation challenge in hypersensitivity pneumonitis. Eur Respir J 2014;44(6):1658- 1665. 75. Adams TN, Newton CA, Batra K, et al. Utility of Bronchoalveolar Lavage and Transbronchial Biopsy in Patients with Hypersensitivity Pneumonitis. Lung 2018;196(5):617- 622. 76. Welker L, Jorres RA, Costabel U, Magnussen H. Predictive value of BAL cell differentials in the diagnosis of interstitial lung diseases. Eur Respir J 2004;24(6):1000- 1006. 77. Patolia S, Tamae Kakazu M, Chami HA, et al. Bronchoalveolar Lavage Lymphocytes in the Diagnosis of Hypersensitivity Pneumonitis among Patients with Interstitial Lung Disease. Ann Am Thorac Soc 2020;17(11):1455- 1467. 78. Adderley N, Humphreys CJ, Barnes H, Ley B, Premji ZA, Johannson KA. Bronchoalveolar lavage fluid lymphocytosis in chronic hyper- sensitivity pneumonitis: a systematic review and meta- analysis. Eur Respir J 2020;56(2). 79. Chami HA, Diaz- Mendoza J, Chua A, et al. Transbronchial Biopsy and Cryobiopsy in the Diagnosis of Hypersensitivity Pneumonitis among Patients with Interstitial Lung Disease. Ann Am Thorac Soc 2021;18(1):148- 161. 80. Johannson KA, Marcoux VS, Ronksley PE, Ryerson CJ. Diagnostic Yield and Complications of Transbronchial Lung Cryobiopsy for Interstitial Lung Disease. A Systematic Review and Metaanalysis. Ann Am Thorac Soc 2016;13(10):1828- 1838. 81. Troy LK, Grainge C, Corte TJ, et al. Diagnostic accuracy of trans- bronchial lung cryobiopsy for interstitial lung disease diagno- sis (COLDICE): a prospective, comparative study. The Lancet Respiratory medicine. 2020;8(2):171- 181. 82. Hutchinson JP, Fogarty AW, McKeever TM, Hubbard RB. In- hospital mortality after surgical lung biopsy for intersti- tial lung disease in the United States. Am J Respir Crit Care Med 2016;193(10):1161- 1167. 83. Barrios RJ. Hypersensitivity pneumonitis: histopathology. Arch Pathol Lab Med 2008;132(2):199- 203. 84. Craig TJ, Hershey J, Engler RJ, Davis W, Carpenter GB, Salata K. Bird antigen persistence in the home environment after removal of the bird. Ann Allergy 1992;69(6):510- 512. 85. Greinert U, Lepp U, Becker W. Bird Keeper's lung without bird kee- pinge. Eur J Med Res 2000;5(3):124. 86. Salisbury ML, Myers JL, Belloli EA, Kazerooni EA, Martinez FJ, Flaherty KR. Diagnosis and Treatment of Fibrotic Hypersensitivity Pneumonia. Where We Stand and Where We Need to Go. Am J Respir Crit Care Med 2017;196(6):690- 699. 87. Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids– new mechanisms for old drugs. N Engl J Med 2005;353(16):1711- 1723. 88. Morath C, Schwenger V, Beimler J, et al. Antifibrotic actions of mycophenolic acid. Clin Transplant 2006;20(s17):25- 29. 89. Tiede I, Fritz G, Strand S, et al. CD28- dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. J Clin Invest. 2003;111(8):1133- 1145. 90. Weiner GJ. Rituximab: mechanism of action. Semin Hematol 2010;47(2):115- 123. 91. Flaherty KR, Wells AU, Cottin V, et al. Nintedanib in Progressive Fibrosing Interstitial Lung Diseases. N Engl J Med 2019;381(18):1718- 1727. 92. Kokkarinen JI, Tukiainen HO, Terho EO. Effect of corticosteroid treatment on the recovery of pulmonary function in farmer's lung. Am Rev Respir Dis 1992;145(1):3- 5. 93. Adegunsoye A, Oldham JM, Fernandez Perez ER, et al. Outcomes of immunosuppressive therapy in chronic hypersensitivity pneu- monitis. ERJ Open Res. 2017;3(3):00016- 02017. 94. Morisset J, Johannson KA, Vittinghoff E, et al. Use of Mycophenolate Mofetil or Azathioprine for the Management of Chronic Hypersensitivity Pneumonitis. Chest 2017;151(3):619- 625. 95. Adegunsoye A, Morisset J, Newton CA, et al. Leukocyte Telomere Length and Mycophenolate Therapy in Chronic Hypersensitivity Pneumonitis. Eur Respir J. 2021;57(3):2002872. 96. Lota HK, Keir GJ, Hansell DM, et al. Novel use of rituximab in hy- persensitivity pneumonitis refractory to conventional treatment. Thorax 2013;68(8):780- 781. 97. Keir GJ, Maher TM, Ming D, et al. Rituximab in severe, treatment- refractory interstitial lung disease. Respirology 2014;19(3):353- 359. 98. Ferreira M, Borie R, Crestani B, et al. Efficacy and safety of ritux- imab in patients with chronic hypersensitivity pneumonitis (cHP): A retrospective, multicentric, observational study. Respir Med 2020;172:106146. 99. Guler SA, Hur SA, Lear SA, Camp PG, Ryerson CJ. Body com- position, muscle function, and physical performance in fibrotic interstitial lung disease: a prospective cohort study. Respir Res 2019;20(1):56. 100. Tonelli R, Cocconcelli E, Lanini B, et al. Effectiveness of pulmo- nary rehabilitation in patients with interstitial lung disease of dif- ferent etiology: a multicenter prospective study. BMC Pulm Med. 2017;17(1):130. 101. Spruit MA, Singh SJ, Garvey C, et al. An official American Thoracic Society/European Respiratory Society statement: key concepts and advances in pulmonary rehabilitation. Am J Respir Crit Care Med 2013;188(8):e13- 64. 102. Dowman L, Hill CJ, Holland AE. Pulmonary rehabilita- tion for interstitial lung disease. Cochrane Database Syst Rev. 2014;10:CD006322. 103. Troy LK, Young IH, Lau EM, Corte TJ. Exercise pathophysiology and the role of oxygen therapy in idiopathic interstitial pneumonia. Respirology. 2016;21(6):1005- 1014. 104. Kern RM, Singer JP, Koth L, et al. Lung transplantation for hyper- sensitivity pneumonitis. Chest 2015;147(6):1558- 1565. 105. Maher TM, Molina- Molina M, Russell AM, et al. Unmet needs in the treatment of idiopathic pulmonary fibrosis- insights from pa- tient chart review in five European countries. BMC Pulm Med. 2017;17(1):124. 106. Troy LK, Chapman SA, Lake F, et al. Current Australasian practice for diagnosis and management of idiopathic pulmonary fibrosis: Where are we now? Respirology 2015;20(4):647- 653. 107. Kreuter M, Bendstrup E, Russell AM, et al. Palliative care in in- terstitial lung disease: living well. The Lancet Respiratory medicine. 2017;5(12):968- 980. 108. Chung JH, Montner SM, Adegunsoye A, et al. CT findings asso- ciated with survival in chronic hypersensitivity pneumonitis. Eur Radiol 2017;27(12):5127- 5135. 109. Wang P, Jones KD, Urisman A, et al. Pathologic findings and prog- nosis in a large prospective cohort of chronic hypersensitivity pneumonitis. Chest 2017;152(3):502- 509. 110. Adegunsoye A, Oldham JM, Bellam SK, et al. Computed tomog- raphy honeycombing identifies a progressive fibrotic phenotype with increased mortality across diverse interstitial lung diseases. Ann Am Thorac Soc 2019;16(5):580- 588. 111. Jacob J, Bartholmai BJ, Egashira R, et al. Chronic hypersensitivity pneumonitis: identification of key prognostic determinants using automated CTanalysis. BMC Pulm Med. 2017;17(1):81. 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense | 453BARNES Et Al. 112. Jacob J, Bartholmai BJ, Rajagopalan S, et al. Automated computer- based CT stratification as a predictor of outcome in hypersensitiv- ity pneumonitis. Eur Radiol 2017;27(9):3635- 3646. 113. Koschel DS, Cardoso C, Wiedemann B, Hoffken G, Halank M. Pulmonary hypertension in chronic hypersensitivity pneumonitis. Lung 2012;190(3):295- 302. 114. Wang BR, Edwards R, Freiheit EA, et al. The Pulmonary Fibrosis Foundation Patient Registry. Rationale, Design, and Methods. Ann Am Thorac Soc 2020;17(12):1620- 1628. 115. Lubin M, Chen H, Elicker B, Jones KD, Collard HR, Lee JS. A comparison of health- related quality of life in idiopathic pulmo- nary fibrosis and chronic hypersensitivity pneumonitis. Chest 2014;145(6):1333- 1338. 116. Aronson KI, Hayward BJ, Robbins L, Kaner RJ, Martinez FJ, Safford MM. ‘It’s difficult, it’s life changing what happens to you’ patient perspective on life with chronic hypersensitivity pneumonitis: a qualitative study. BMJ Open Respiratory Research. 2019;6(1):e000522. 117. Alqalyoobi S, Adegunsoye A, Linderholm A, et al. Circulating Plasma Biomarkers of Progressive Interstitial Lung Disease. Am J Respir Crit Care Med. 2020;201(2):250- 253. 118. Janssen R, Grutters JC, Sato H, et al. Analysis of KL- 6 and SP- D as disease markers in bird fancier's lung. Sarcoidosis Vasc Diffuse Lung Dis. 2005;22(1):51- 57. 119. Long X, He X, Ohshimo S, et al. Serum YKL- 40 as predic- tor of outcome in hypersensitivity pneumonitis. Eur Respir J 2017;49(2):1501924. 120. Nukui Y, Miyazaki Y, Masuo M, et al. Periostin as a predictor of prognosis in chronic bird- related hypersensitivity pneumonitis. Allergology International. 2019;68(3):363- 369. 121. Nukui Y, Yamana T, Masuo M, et al. Serum CXCL9 and CCL17 as biomarkers of declining pulmonary function in chronic bird- related hypersensitivity pneumonitis. PLoS One 2019;14(8):e0220462. 122. Pardo A, Cabrera S, Maldonado M, Selman M. Role of matrix metalloproteinases in the pathogenesis of idiopathic pulmonary fibrosis. Respir Res 2016;17(1):23. 123. Agassandian M, Tedrow JR, Sembrat J, et al. VCAM- 1 is a TGF- beta1 inducible gene upregulated in idiopathic pulmonary fibrosis. Cell Signal 2015;27(12):2467- 2473. 124. Tsutsui T, Miyazaki Y, Kuramochi J, Uchida K, Eishi Y, Inase N. The amount of avian antigen in household dust predicts the progno- sis of chronic bird- related hypersensitivity pneumonitis. Ann Am Thorac Soc 2015;12(7):1013- 1021. 125. Adegunsoye A, Oldham JM, Demchuk C, Montner S, Vij R, Strek ME. Predictors of survival in coexistent hypersensitivity pneumo- nitis with autoimmune features. Respir Med 2016;114:53- 60. 126. Idiopathic Pulmonary Fibrosis Clinical Research Network, Raghu G, Anstrom KJ et al. Prednisone, azathioprine, and N- acetylcysteine for pulmonary fibrosis. N Engl J Med. 2012;366(21):1968- 1977. How to cite this article: Barnes H, Troy L, Lee CT, Sperling A, Strek M, Glaspole I. Hypersensitivity pneumonitis: Current concepts in pathogenesis, diagnosis, and treatment. Allergy. 2022;77:442– 453. https://doi.org/10.1111/all.15017 13989995, 2022, 2, D ow nloaded from https://onlinelibrary.w iley.com /doi/10.1111/all.15017 by C A PE S, W iley O nline L ibrary on [29/07/2024]. See the T erm s and C onditions (https://onlinelibrary.w iley.com /term s-and-conditions) on W iley O nline L ibrary for rules of use; O A articles are governed by the applicable C reative C om m ons L icense https://doi.org/10.1111/all.15017