Buscar en
Revista Colombiana de Reumatología
Toda la web
Inicio Revista Colombiana de Reumatología Interstitial lung disease in patients with idiopathic inflammatory myopathy (IIM...
Información de la revista
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
754
Review Article
Acceso a texto completo
Disponible online el 17 de Octubre de 2023
Interstitial lung disease in patients with idiopathic inflammatory myopathy (IIM-ILD): Definitions, epidemiology, pathophysiology, clinical manifestations, complications, risk, and mortality factors (narrative review)
Enfermedad pulmonar intersticial en pacientes con miopatía idiopática inflamatoria (EPI-MII): definiciones, epidemiología, fisiopatología, manifestaciones clínicas, complicaciones, factores de riesgo y mortalidad (revisión narrativa)
Visitas
754
Andrés Hormaza-Jaramilloa,b,
Autor para correspondencia
andres.hormaza@fvl.org.co

Corresponding author.
, Liliana Fernandez-Trujillob,c, Tatiana Delgado-Morad, Carlos Julio Vargas-Potesb,e
a Department of Internal Medicine, Rheumatology Service, Fundación Valle del Lili, Cali, Colombia
b Faculty of Health Sciences, Universidad Icesi, Cali, Colombia
c Department of Internal Medicine, Pneumology Service, Fundación Valle del Lili, Cali, Colombia
d Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
e Department of Internal Medicine, Internal Medicine Service, Fundación Valle del Lili, Cali, Colombia
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (1)
Tablas (1)
Table 1. Autoantibodies and their frequency in interstitial lung disease, myositis, mortality, clinical features and radiological patterns.
Abstract

Interstitial lung disease refers to a group of disorders generally characterized by progressive scarring of lung tissue due to a wide variety of causes and associated with a plethora of symptoms. Patients with this diagnosis can be asymptomatic or present severe symptoms that could lead to death. Its signs and symptoms are the same in patients with concomitant connective tissue disease and those without. Genetics and immunity play essential roles in patients with interstitial lung disease and idiopathic inflammatory myopathy. Alterations in genes and excessive production of specific cytokines can lead to the development of interstitial lung disease. Interstitial lung disease can have several complications, including chronic respiratory distress and infections, and can worsen the prognosis of patients with idiopathic inflammatory myopathy. Here, we present a narrative review describing the epidemiology, pathophysiology, clinical manifestations, risk factors, and complications of the population with interstitial lung disease and idiopathic inflammatory myopathy.

Keywords:
Interstitial lung disease
Idiopathic inflammatory myopathy
Autoantibodies
Epidemiology
Clinical manifestations
Mortality
Resumen

La enfermedad pulmonar intersticial se refiere a un grupo de trastornos generalmente caracterizados por cicatrización progresiva del tejido pulmonar, debidos a una amplia variedad de causas y asociados con múltiples síntomas. Los pacientes con este diagnóstico pueden estar asintomáticos o presentar síntomas severos que podrían conducir a la muerte. Los signos y los síntomas de esta enfermedad son iguales en pacientes que tienen una enfermedad del tejido conectivo concomitante y en aquellos que no la tienen. La genética y la inmunidad desempeñan papeles esenciales en pacientes con enfermedad pulmonar intersticial y miopatía idiopática inflamatoria. Las alteraciones en los genes y la producción excesiva de citoquinas específicas pueden conducir al desarrollo de la enfermedad. A su vez, la enfermedad pulmonar intersticial puede tener varias complicaciones, como dificultad respiratoria crónica e infecciones, y puede empeorar el pronóstico de los pacientes con miopatía idiopática inflamatoria. Presentamos una revisión narrativa que describe la epidemiología, la fisiopatología, las manifestaciones clínicas, los factores de riesgo y las complicaciones de la población con enfermedad pulmonar intersticial y miopatía idiopática inflamatoria.

Palabras clave:
Enfermedad pulmonar intersticial
Miopatía idiopática inflamatoria
Autoanticuerpos
Epidemiología
Manifestaciones clínicas
Mortalidad
Texto completo
Introduction

Interstitial lung disease (ILD) encompass a heterogeneous group of conditions that affect the lungs, characterized by varying degrees of inflammatory and/or fibrotic involvement with specific radiological patterns on high-resolution computed tomography and in histopathology in lung biopsy samples, leading to deterioration of lung function and increased morbidity/mortality in patients.1,2 The respiratory system, especially the lung parenchyma, is a frequent target of autoimmune-mediated injury. Rheumatological diseases can present different forms of pulmonary involvement, one of which is interstitial lung disease associated with connective tissue disease (CTD-ILD).3

CTD-ILD is a significant risk factor for morbidity and mortality and represents a diagnostic challenge in the earliest stages,4 which warrants a multidisciplinary approach. As in other autoimmune diseases, ILD is a complication in patients with idiopathic inflammatory myopathies (IIMs), disorders of unknown etiology that involve different degrees of muscle inflammation. In this review of IIM-ILD, we summarize the essential disease features of this condition and its evolving epidemiology.

Based on the clinical, immunological, and histological characteristics, five groups that make up the IIM complex can be distinguished: overlap myositis (which includes antisynthetase syndrome, aSyS), dermatomyositis (DM), polymyositis (PM), inclusion body myositis, and immune-mediated necrotizing myopathy.5 We can also include the spectrum of clinically amyopathic dermatomyositis (CADM) or “dermatomyositis sine myositis,” characterized by the same skin findings as in DM but without muscle weakness or elevation of muscle enzymes, which confer a higher risk of ILD, the rapid progressive form (RP-ILD).6 Of the aforementioned subtypes, DM, PM, aSyS, and CADM are frequently associated with ILD.7

Methodology

A non-systematic narrative review of the literature was carried based upon available Spanish and English language literature in the PubMed database.

ResultsEpidemiology

IIM-ILD was first described in a patient with DM in 1956.8 By 1974, the prevalence of ILD in DM/PM was estimated at 5%.9 Since then, the association of IIM with ILD has been established through multiple case series, multicenter studies with highly variable prevalences (from 19.9 to 86%),10–12 and later meta-analyses and systematic reviews of the literature. The overall prevalence of IIM has been estimated to range from 14.0 to 21.4 cases per 100,000 population in the United States.13 Sun et al. detected a global prevalence of 41% of ILD presentation in the composite of patients with DM/PM in the first meta-analysis and systematic review, with a predominance in the Asian population (twice that in Europe and America). The prevalences of ILD in the DM, PM, and CADM subtypes, were 42%, 35%, and 53%, respectively, with high heterogeneity between the studies. Based on the data available, ILD has a rising prevalence, probably attributed to a more active search and the diagnostic tools available.14 In the most recent meta-analysis by Joy et al., an overall prevalence of IIM-ILD of 41% (95% CI 33–50%) was reported, without specifying prevalences in the clinical subtypes.15 However, when associated with antibodies against aminoacyl-tRNA synthetase (ARS), the majority exceeds 70%.16

Pathophysiology

Lung injury begins with damage to the alveolar epithelial or endothelial cells, allowing an increase in permeability and destruction of the alveolar-capillary membrane. Next, inflammatory cell infiltration at the interstitial space, promoting the infiltration of fibroblasts and the formation of fibrotic foci from the production of extracellular matrix proteins by myofibroblasts.17,18 Endothelial damage has been proposed as the initial pathway for events in CTD-ILD, potentially through the early growth response protein 1 (EGR1), lysophosphatidic acid receptor 1 (LPAR1), and Wnt/catenin pathways. Oxidative stress and the response to proteins unfolded by cellular stress are related to the endoplasmic reticulum.19 This damage induces the release of proteins such as surfactant protein D (SP-D) and Krebs von den Lungen protein-6 (KL-6), which have key roles in the prognosis of patients, which will be explained later.

Genetics

Immunogenetics plays an essential role in developing IIM-ILD, and specific subtypes of HLA-DRB1 and tumor necrosis factor alpha have been reported in patients with IIM-ILD. HLA-DRB1*03–DQA1*05–DQB1*02 showed an association with the expression of ILD in patients with DM and patients with PM, in whom it has been associated with the positivity of an antisynthetase antibody.20 In turn, HLA-B*08.01 favors the cascade of events and includes the presentation of antigens, the priming of CD8+ T cells, and the crosstalk of CD4+ T cells with B cells.21

Telomere shortening can hinder the healing and/or turnover of alveolar epithelial cells after an initial injury, and mutations in genes related to leukocyte telomeres have been related to cellular senescence and alteration of the reparative response. Among them is telomerase reverse transcriptase (TERT), telomerase RNA component (TERC), dyskerin pseudouridine synthase 1 (DKC1), telomere elongation regulator helicase 1 (RTEL1), poly-A-specific ribonuclease (PARN), and TERF1-interacting nuclear factor 2 (TINF2).22 In the case of DM-/PM-ILD, specific susceptibility gene loci have been detected (DQB1*06:02, DRB1*03, DRB1*01:01, DRB1*04:05).23–25

Innate immunity

Among the environmental factors related to inflammatory lung injury are gastroesophageal reflux and infections (mainly Epstein–Barr virus, retroviruses, parvoviruses, mycobacteria, Mycoplasma spp., and Borrelia spp.).26 In ILD, macrophages can be polarized to become classic proinflammatory M1 macrophages, which secrete proinflammatory and/or profibrotic cytokines (IL-1β, IL-8, IL-10, and CXCL13), or alternative profibrotic M2a macrophages, which secrete profibrotic cytokines (CCL22, PDGF-BB, and IL-6).18 Ye et al. used a single-sample gene set analysis of variation. They found the type I interferon signaling pathway in the lungs of DM patients with MDA5+ antibodies and a higher score of interferon-stimulated genes (ISG) and fibrosis. Additionally, they observed that fibroblasts had strong interactions of the type I interferon signaling pathway with antibody-secreting cells (ASCs), subsets of CD8+ T cells, and macrophages in the lungs of the DM MDA5+ patient, but not in healthy controls. These data suggest that, in the context of the overactivation of type I interferon signaling, infiltrated immune cells and fibroblasts potentially form a unique profibrotic microenvironment in the lungs of patients with MDA5+ DM.27

Extracellular neutrophil traps (NETs), which can lead to the formation of autoantibodies, cause direct damage to epithelial cells and result in increased production of proinflammatory cytokines that induce different NET formation, thus perpetuating the harm (found in PM/DM), in addition to tending to activate pulmonary fibroblasts and differentiate into myofibroblasts.22 Proteomics analysis shows a significant increase in galactosylated IgG Fc-glucan has been detected in patients with IIM vs. the general population. This finding was not correlated with other extra muscular manifestations, so overexpression may have a specific role in pulmonary involvement. Overexpression has also been found in gelsolin (related to the degradation of actin filaments released by necrotic cells during inflammation) and calgranulin B in patients with overlap syndrome.28

Adaptive immunityT lymphocytes

Preclinical studies have identified profibrotic profiles (Th2, Th17), antifibrotic profiles (Th1, Th22, and γδ-T), and pleiotropic T lymphocytes (Tregs and Th9) as mediators of fibrosis.29 Lung biopsies of patients with CTD-ILD have shown an increase in T lymphocytes in lung tissue and in lymphoid aggregates. In addition, the bronchoalveolar lavage of patients with IIM has accumulated cytotoxic CD8+ T cells.30 A cohort study in patients with ILD showed a decrease in blood lymphocytes with a higher ratio of CD4:CD8, which suggests an increase in cytotoxic activity with accelerated cell destruction.21 In another study, an increase in CD4+ CXCR4+ T cells was detected in the blood and bronchoalveolar lavage of patients with MDA5+ DM, with the ability to promote the proliferation of pulmonary fibroblasts through IL-21, which suggests a potential pathogenic role in MDA5+ DM.31

B lymphocytes

An exaggerated ASC response has been found in patients with MDA5+ DM. Ye et al. evaluated the peripheral B-cell compartment by single-cell RNA sequencing: three groups were naive B cells (scB1-Transitional, scB2-ISG, and scB3-Naïve), three were memory B cells (scB4-Unswitched Bm, scB5-Switch Bm [changed Bm], and atypical scB6-Bm), and two were ASCs (scB7-pASC [proliferating] and scB8-rASC [resting]). The MDA5+ DM-Active group showed significantly higher proportions of scB2-ISG, scB7-pASC, and scB8-rASC cells. Furthermore, the proportions of the three groups were strongly reduced in the three MDA5+ DM-Remission patients. The frequencies of CD19+CD20−CD38hi were significantly higher in MDA5+ DM-Active patients than in control patients (IIM without ILD) and healthy controls. A high clonal expansion of ASCs was also found in patients with MDA5+ DM-Active.27

B-cell activating factor (BAFF), also known as BLys, is a member of the TNF family of cytokines and an important agent in B-cell survival. It has been implicated in pathogenesis and as a biomarker correlated with disease activity in a variety of autoimmune diseases in which antibodies and B cells are believed to be relevant for pathogenesis. In IIM, elevated levels of BAFF have been reported in patients with PM, anti-Jo-1 antibodies and ILD and in patients with DM, regardless of the presence of anti-Jo-1 positivity ILD.32,33 In Fig. 1, we propose an image illustrating the pathophysiology of ILD in patients with IIM.

Fig. 1.

Pathophysiology of interstitial lung disease in patients with idiopathic inflammatory myopathy.

(0,47MB).
Autoantibodies

In patients with IIM-ILD in Latin American population the most frequently detected autoantibodies are anti-Ro-52 and anti-Jo-1.34 Although myositis-specific autoantibodies (MSAs) and myositis-associated autoantibodies (MAAs) as a whole are not part of the current diagnostic and classification criteria for myositis, it has been recognized that specific autoantibodies confer distinct clinical phenotypes.35 Despite the aforementioned major importance of autoantibodies in the pathophysiology of IIM-ILD, the 2017 classification criteria of the European League Against Rheumatism/American College of Rheumatology (EULAR/ACR) only incorporated the presence of anti-Jo-1.2 Therefore, some patients could be misclassified, especially those who are hypo or amyopathic, or patients with ILD, MSA and hypo or amyopathic disease could be classified as having interstitial pneumonia with autoimmune features (IPAF).36Table 1 presents the frequency of each autoantibody in ILD, in myositis, mortality, clinical features, and radiological patterns.

Table 1.

Autoantibodies and their frequency in interstitial lung disease, myositis, mortality, clinical features and radiological patterns.

Group  Antibody  Antigen  Frequency in ILD  Frequency in myositis  Mortality  Clinical features  Radiological patterns 
Myositis-specific antibodies (MSA)Anti-ARS  Aminoacyl-tRNA synthetase (ARS)  Strong association37All ARS: 70%38Anti-Jo-1: 66%38OR: 11.138Non-Jo-1: 84%38Anti-PL12 OR: 4.9 (0.45–53)39Anti-PL7 OR: 3.04 (0.6–15.2)39Anti EJ: OR: 14.2 (1.69–118.9)39  General: 25–42%40Anti-Jo-1: DM: 11% PM: 21%38Non-Jo-1: DM: 10% PM: 8%38Anti-PL7: 5–10%41Anti-PL12: <5%41Anti EJ: 5–10%41Anti OJ: <5%41Anti KS: <5%41Anti Ho: <1%41  49 months survival. ARS-ILD with myositis: 20%42ARS-ILD without myositis: 18.8%425-Years survivalAnti-Jo-1: 90%43Non-Jo-1: 75%43  Lung: chronic or subacute ILD (more severe in anti-PL7, -KS, -OJ, -PL12)Muscle: myositis (mostly in anti-Jo-1, -PL7, -EJ).Skin: Mechanic's handOthers: Raynaud phenomenon, non-erosive arthritis (especially anti-Jo-1), fever16,44  NSIP (50%)OP (20%)5,16Traction bronchiectasis (75%)5 
Anti-MDA5  Melanoma differentiation-associated protein 5 (MDA5)  Strong association3750–100%16,39,40OR: 3.109 (1.578–6.128)39  ∼10–20% in DM16∼50–70% in CADM16Caucasian: 0–13%45Asian: 11–57%45  6-Months survival: 33–66%465-Year survival: 56%47  Lung: RP-ILD, OR: 25.33. Spontaneous pneumomediastinum or pneumothoraxMuscle: Often amyopathic or mild myositisSkin: Gottron's papules, heliotrope rash, ulceration, palmar pustulesOthers: Raynaud phenomenon16,44–46  Mixture of characteristics of NSIP, OP and UIP, without a typical pattern46 
Anti-Mi-2  Nucleosome remodeling and deacetylase complex (NuRD)  Weak association374%38OR: 0.18 (0.05–0.58)39  4–10% of all myositis41DM: 9% PM: 1%38  44 months survival: 97.1%48  Lung: No known associationMuscle: myositis (generally mild)Skin: classic DM rash44  Not reported 
Anti-SAE  Small ubiquitin-like modifier activating enzyme  Western: 0–18%49Asian: 25–71%49General: 21%50OR: 5.54 (0.192–160.19)51  2–8% of adult DM7,52  5%53Good response to treatment49  Lung: no known associationMuscle: may be amyopathic initiallySkin: classic DM rash, cutaneous ulcers, dark red/violaceous rashOther: increased cancer associated DM prevalence44,45,50  Organizing pneumonia (OP)54 
Anti-NXP2  Nuclear matrix protein 2  Doubtful association37Li et al.: OR 0.26 (0.18–0.38)55Xing et al.: OR: 1.4 (0.43–4.75)51  Adult: 2–17%56Caucasian: 14–25%45Asian: 2–5%45  No connection with poor prognosis55  Lung: no known associationMuscle: myositis (generally severe, distal weakness, dysphagia)Skin: classic DM rash, calcinosisOther: Peripheral edema. Increased cancer association (3.68-fold increase)44,45,57  NSIP, OP37 
Anti-HMGCR  3-Hydroxy-3-methylglutaryl CoA reductase  Weak association371%.OR: 0.05 (0.007–0.451)39  Adult: 6%56  Limited data  Lung: no known associationMuscle: necrotizing myositisSkin: often absent. Statin use association44  Not reported37 
Anti-SRP  Signal recognition particle (SRP)  Doubtful association3715% (7–25)38OR: 2.014 (0.405–10.02)51  DM: 1% (1–2)38PM: 5% (3–7)383–10% of all myositis40Adult: 2%56  10 years survival: 96.4%58Good response to therapy  Lung: association with ILD reported44Muscle: necrotizing myositis severe and rapidly progressive myositis, severe symmetric proximal muscle diseaseSkin: Often absent40  NSIP37 
Anti-TIF1-γ/α  Transcription intermediary factor 1-γ/α (TIF1-γ/α)  Weak association3718%OR: 0.163 (0.080–0.333)39  20–29% of DM40  Limited data  Lung: negatively associatedMuscle: myositis (mild or rarely amyopathic)Skin: classic DM rash, severe skin disease, ‘red on white’ lesionsOther: Strong association with malignancy (up to 75%)44,45  Not reported37 
Myositis-associated antibodies (MAA)Anti-cN1A  Cytosolic 5′-nucleotidase 1A (cN1A)  Not typical. Limited to case reports  33–37% of IBM40Adult: 4–21%56  HR 1.95, 95% CI=1.17–3.27 in IBM59  IBM, inclusion body myositis40  Not reported 
Anti-U1-RNP  U1-ribonucleoprotein (U1-RNP)  7%(1–24)38  DM: 6%(4–8)PM: 5% (3–7)38Adult: 10%56  Limited dataGood response to therapy60  Overlap syndrome, MCTD Raynaud's phenomenon, ILD, PAH40  Not reported 
Anti-PM/Scl  PM/Scl100, PM/Scl75  Strong association3738% (25–52)38  DM: 9%(6–12)38PM: 6% (4–9)383–10% of all myositis40  9.5-Year mortality: 8%61  Lung: late onset, chronicIndolent37PM-SSc overlap syndrome40  NSIP61 
Anti-Ku  Components of DNA-dependent protein kinase (Ku70/Ku80)  Doubtful association3727% (8–55)38OR: 0.379 (0.06–2.39)51  DM: 1 (1–2)38PM: 2 (1–3)382% of all myositis40  18%62  Lung: refractory to therapy, impacts on prognosis37Muscle: overlap syndrome40Skin: Raynaud phenomenon63Other: arthralgia63  Unknown37 
Anti-Ro52  TRIM21 located in cytoplasm and nucleus  Strong association37OR 3.1, 95% CI 1.3–7.651Isolated: 7%2  25–30.9%54,56With con concurrent anti-Jo-1 positive: 56–72%64  All-cause mortality: 28.8% (composite of patients with anti-Ro52 alone vs. anti-Ro52 plus an additional MSA)6524-Month mortality (in patients with anti-MDA5-positive CADM-ILD): 59.9% vs. 85.7%; p=0.05166  Lung: High predictor of ILD, poor outcomes if associated with anti-MDA5 and antisynthetase37RP-ILD: 54.8% vs. 23.8%; p=0.01466  Chronic, insidious, fibrosing processes, less acute/subacute subtypes37 
Biomarkers

Other biomarkers of ILD activity include lactate dehydrogenase (LDH), Krebs von den Lungen protein-6 (KL-6), serum surfactant protein D (SP-D) and ferritin. KL-6 is a high molecular weight mucin-like glycoprotein expressed in type II alveolar pneumocytes and bronchiolar epithelial cells. Fathi et al. detected significantly more KL-6 in PM/DM patients with ILD than in PM/DM patients without ILD, which was correlated with functional residual capacity and an increased risk of developing ground-glass opacities, diffuse fibrosis and honeycombing.67 Ihn et al. reported higher levels of SP-D in ILD-DM/PM, with an inverse relationship with FVC and DLCO.68 Serum ferritin is correlated with the disease activity of CADM-ILD or anti-MDA5+ DM.32 In a small cohort, a threshold ferritin level greater than or equal to 1600ng/ml was the most sensitive indicator of survival.69

Clinical manifestations

The signs and symptoms related to IIM-ILD do not differ from those of other types of ILD, and it has a variable presentation from asymptomatic forms to exertional dyspnea (50%), nonproductive cough (33%), decreased exercise tolerance, clubbing and asthenia.5 As in other autoimmune diseases, respiratory manifestations can appear before the appearance of skin or muscle manifestations in 2.7–37.5% of cases1,70,71 or afterward (up to 40%).3,72,73

Particular attention should be paid to the extrapulmonary characteristics of IIMs to distinguish secondary ILD from primary ILD. The evaluation of these patients includes a careful investigation of constitutional symptoms (weight loss and fever, present in up to 20%), respiratory manifestations (bibasal rales on auscultation and reduction of respiratory movements due to muscle weakness), cutaneous manifestations (“climber's foot”, “mechanic's hands”, Gottron's papules and rash, heliotrope rash, Holster and shawl signs, and Raynaud's phenomenon), articular (seronegative arthritis of the distal joints, with asymmetric and oligoarticular characteristics). Clubbing and hypertrophic osteopathy are rare in IIM-ILD.74,75

In 2021, a multidisciplinary consensus of experts was published aiming at the early diagnosis and follow-up of CTD-ILD. One of the recommendations is to recognize signs and symptoms as “red flags” to suspect ILD, including basal Velcro rales on lung auscultation and dry cough with dyspnea on exertion not related to infectious or active cardiovascular disease.4 Specifically, in IIM-ILD, the clinical presentation can be classified into three patterns based on the respiratory symptoms at presentation: the rapidly progressive form with acute/subacute symptoms (RP-ILD, 18.7–28.8%), chronic with progressive symptoms (51.4–57.7%), and the asymptomatic/subclinical form (11.5–29.9%).17,76 Deterioration of RP-ILD (expected in less than three months) is defined by two or more of the following: symptomatic exacerbation (dyspnea on exertion), increase in the severity of abnormalities in the parenchyma on HRCT, and worsening of lung function parameters (10% in forced vital capacity or ten mmHg in the partial pressure of oxygen).2,77

A clear relationship has been found between CADM and ILD. Mukae et al. compared the clinical presentation of ILD in patients with CADM and DM, finding a much shorter time of respiratory symptoms before hospital admission (4.6 vs. 34.1 months), a greater rate of PR-ILD (64 vs. 19%), and higher mortality (45 vs. 6%, all in the acute compromise subgroup).78 Fewer than 20% of patients with DM and PM have this presentation.3 The chronic form presents with dyspnea of insidious appearance and nonproductive cough with rare constitutional symptoms. Radiographically, it can appear as organized pneumonia (OP) or overlap of OP with nonspecific interstitial pneumonia (NSIP) with an excellent response to glucocorticoids and a chronic fibrosing form, which corresponds to fibrotic NSIP or usual interstitial pneumonia (UIP), which tends to respond poorly to glucocorticoids and other types of immunosuppression.79 The presence of radiological, and physiological defines the asymptomatic or subclinical form, and, in some cases, subtle or histopathological abnormalities in asymptomatic patients with symptoms that have not been attributed to ILD80; it appears in DM and PM in 30% of patients.72,81

Complications

The complications derived from IIM-ILD are many and include chronic respiratory failure, respiratory and gastrointestinal opportunistic infections (with a mortality of 28%), hypoventilation and hypercapnia in the context of muscle dysfunction, microaspirations with risk of aspiration pneumonia/pneumonitis, type 1 pulmonary hypertension (associated with connective tissue disease), and type 3 pulmonary hypertension (mediated by pulmonary involvement). Finally, pneumomediastinum occurs in 15% of IIM-ILD patients and has a high mortality rate.5,79 The appearance of opportunistic infections in IIM-ILD is significant and could at least be associated with the disease itself and its treatments. Thus, preventive therapy of Pneumocystis jirovecii should be prescribed as soon as patients receive glucocorticoids at equivalent doses of prednisolone >20mg/d for >4 weeks, especially for the most severe cases.82,83

Risk factors and prognosis

It is clinically of considerable importance to identify antibodies in patients with PM/DM, because each is closely associated with certain clinical features. Older age of presentation,84,85 arthralgia,15,84,85 fever,85 elevated acute-phase reactants (erythrocyte sedimentation rate, C-reactive protein),15,85 Afro-descendant ethnicity, presence of mechanical hands, lateral erythema of the hip, anti-ARS, anti-MDA5, antinuclear antibodies (ANA), anti-Sjögren syndrome type B (anti-SSB) antibodies, and anti-Ro52 are independent risk factors for developing IIM-ILD.15 ILD was observed more frequently in patients with anti-EJ (OR 14.202, 95% CI 1.696–118.902), anti-Jo-1 (OR 11.111, 95% CI 3.306–37.335), and anti-MDA5 antibodies (OR 3.109, 95% CI 1.578–6.128). Patients with ILD who had anti-HMGCR seemed less likely to develop ILD, which suggests that anti-HMGCR could help protect against ILD in patients with IIM.39

Anti-Mi-2 (OR 0.180, 95% CI 0.055–0.589), anti-TIF1-γ (OR 0.163, 95% CI 0.080–0.333), and anti-HMGCR (OR 0.058, 95% CI 0.007–0.451) were protective factors against the development of ILD.39 ANAs, anti-Ro52, or SSA did not significantly alter mortality.86,87 In the Latin American cohort of Alberti et al., none of the clinical or antibody variables were statistically significant for poor baseline lung function in multivariate analysis.34 The prevalence of anti-MDA5 and anti-Ro-52 was significantly higher in DM/PM with ILD than in DM/PM without ILD (anti-MDA5, 45.57 vs. 0.00%, respectively, p<0.001; anti-Ro-52, 60.76 vs. 26.09%, respectively, p<0.001). In contrast, the prevalence of anti-TIF1-γ and anti-NXP2 was significantly lower in DM/PM with ILD than in DM/PM without ILD (anti-TIF1-γ, 3.80 vs. 19.57%, respectively, p=0.01; anti-NXP2, 1.27 vs. 10.87%, respectively, p=0.047). No significant difference was observed in the prevalence of anti-Mi-2α, anti-Mi-2β, anti-SAE1, anti-SRP, anti-Ku, anti-PM-Scl75, or anti-PM-Scl100 in DM/PM patients with ILD vs. DM/PM patients without ILD (anti-Mi-2α, 6.33 vs. 4.35%; anti-Mi-2β, 7.59 vs. 0.00%; anti-SAE1, 0.00 vs. 4.35%; anti-SRP, 2.53 vs. 2.17%; anti-Ku, 3.80 vs. 2.17%; anti-PM-Scl75, 2.53 vs. 0.00%; PMScl100, 1.27 vs. 0.00%).88

Although the rates of ILD are high in patients positive for anti-ARS, these patients tend to have a better overall prognosis, with a better response to therapy and higher overall survival rates than patients with anti-ARS-negative IIM-ILD.35 Patients positive for non-Jo-1 anti-ARS have reduced overall survival compared to patients positive for anti-Jo-1. In a study of more than 200 patients, Aggarwal et al. reported that patients with anti-Jo-1 had a 5-year survival of 90% vs. 75% in patients with non-anti-Jo-1 anti-ARS.2 Patients positive for anti-PL7 and anti-PL12 have a high prevalence of lung disease (60–90%). They are strongly associated with milder and rapidly resolving myositis but with early and severe ILD.89 Ro52 and Ro60 are part of a ribonucleoprotein complex called SSA/Ro. Still, only antibodies against the Ro52 subunit are considered markers of IIM and are found in up to 56% of patients positive for anti-Jo-1.89 La Corte et al. reported that patients with coexisting anti-Ro52 antibodies have more severe pulmonary symptoms and greater ILD than those without.90

De Lorenzo et al. reported higher FVC readings in patients with anti-PM/Scl antibodies than in those with anti-ARS antibodies, with less decline in lung function over time. Similarly, a good clinical course has been observed in scleroderma ILD associated with anti-PM/Scl antibodies compared to anti-Scl70 antibodies. Although ILD can manifest early in anti-PM/Scl disease, it occurs more frequently in later stages of the natural history and is strongly associated with cutaneous manifestations.2 In a study in China, the ILD frequency was 35.29% vs. 66.67% in patients positive vs. negative for anti-NXP2 antibodies. All the findings of HRCT showed image characteristics of nonspecific interstitial pneumonia and/or organized pneumonia, and no cases of RP-ILD were observed.91 Authors such as Gossez hold that the measurement of anti-NXP2 should be part of assessing ILD in patients with suspected DM.92 Although all such studies have had small sample sizes, a meta-analysis found that the prevalence of ILD in patients positive for anti-Ku ranged from 8% to 55%. ILD is not strongly associated with anti-Mi-2; Lega et al. reported an average ILD prevalence of 4% in 154 seropositive patients. In a review of 226 patients with anti-U1-RNP and IIM antibodies, only 7% had ILD.2,38 The prevalence of specific antibodies for Ks, Ha, Zoα, and cN1A was 1.3%, 2.0%, 1.4%, and 0.9%, respectively, in ILD.93

The prevalence of ILD among patients positive for anti-SAE1 varies from 50 to 71%,49,94–96 being the highest in Asian populations. ILD is usually mild, with few respiratory symptoms, despite abnormalities on imaging. The characteristics of ILD in anti-SAE1 patients on HRCT are predominantly peripheral subpleural ground-glass opacities corresponding to organized pneumonia.94,95 Li et al. sought to distinguish DM/PM with ILD from DM/PM without ILD, and the sensitivity, specificity, and positive predictive value for anti-MDA5 were 45.57, 100.00, and 100.00%, respectively, while those for anti-Ro-52 were 60.76, 73.91, and 80.00%, respectively.88

Mortality

Significant heterogeneity in the literature regarding reported mortality, ranging from 7.5% to 55%.71,87,97–99 The mortality rates for PM-ILD, DM-ILD, and CADM-ILD are reportedly 16.7%, 24.4%, and 37.2%, respectively.12,84,98,99 Despite early mortality in severe forms, the 5-year survival rate in IIM-ILD is >85%. However, some patients may worsen during the first year of treatment; the time until disease progression is usually counted in years. As an example, in a long-term follow-up series, 20% of patients with IIM-ILD (not including patients with anti-MDA5 antibodies) worsened despite immunosuppressive treatments, and the other patients were stable (35–55%) or improved (25–45%).5

With data collected from the Multicenter Retrospective Cohort of Japanese Patients with myositis-Associated ILD (JAMI), Gono et al. created a prognostic prediction model for patients with DM-/PM-ILD. They found that the combination of C-reactive protein >0.8mg/dL and KL-6 >1000U/ml in patients with anti-MDA5 was associated with a mortality risk of more than 50%.100 Regarding mortality, African-descended ethnicity,89 anti-MDA5,39,86,87 age, male sex,86,87 acute/subacute onset,87 amyopathic clinical disease,84,87 dyspnea, fever, elevated C-reactive protein, LDH, ground-glass opacities,86 ulcers,85 ferritin,86,87 albumin,84,86 reduced %TLCO, and %CV,86,87 cardiac involvement84 carry high risk.

Conclusions

Since the first descriptions of IIM-ILD, our understanding of its pathophysiology and clinical phenotypes have grown. Recognizing the risk and prognostic factors has undoubtedly changed the approach to this rare clinical presentation.

Authors’ contributions

AHJ conceived the idea and design of the study. AHJ, LFT, TDM, and CJVP searched for information in databases. All authors contributed substantially to the writing of the manuscript and approved the submitted version.

Funding

This narrative review did not receive financing from any entity.

Conflicts of interest

The authors declare no conflicts of interest.

References
[1]
R. Vij, M.E. Strek.
Diagnosis and treatment of connective tissue disease-associated interstitial lung disease.
Chest, 143 (2013), pp. 814-824
[2]
M. Basuita, L.M. Fidler.
Myositis antibodies and interstitial lung disease.
J Appl Lab Med, 7 (2022), pp. 240-258
[3]
Y.J. Ha, Y.J. Lee, E.H. Kang.
Lung involvements in rheumatic diseases: update on the epidemiology, pathogenesis, clinical features, and treatment.
Biomed Res Int, 2018 (2018), pp. 6930297
[4]
S.L. Bosello, L. Beretta, N. Del Papa, S. Harari, S. Palmucci, A. Pesci, et al.
Interstitial lung disease associated with autoimmune rheumatic diseases: checklists for clinical practice.
Front Med, 8 (2021), pp. 732761
[5]
B. Hervier, Y. Uzunhan.
Inflammatory myopathy-related interstitial lung disease: from pathophysiology to treatment.
Front Med, 6 (2020), pp. 326
[6]
Y.Z. Gan, L.H. Zhang, L. Ma, F. Sun, Y.H. Li, Y. An, et al.
Risk factors of interstitial lung diseases in clinically amyopathic dermatomyositis.
Chin Med J (Engl), 644 (2020),
[7]
Y. Waseda.
Myositis-related interstitial lung disease: a respiratory physician's point of view.
Medicina (Mex), 57 (2021), pp. 599
[8]
E.S. Mills, W.H. Mathews.
Interstitial pneumonitis in dermatomyositis.
J Am Med Assoc, 160 (1956), pp. 1467-1470
[9]
A.R. Frazier, R.D. Miller.
Interstitial pneumonitis in association with polymyositis and dermatomyositis.
Chest, 65 (1974), pp. 403-407
[10]
B. Atienza-Mateo, S. Remuzgo-Martínez, V.M. Mora Cuesta, D. Iturbe-Fernández, S. Fernández-Rozas, D. Prieto-Peña, et al.
The spectrum of interstitial lung disease associated with autoimmune diseases: data of a 3.6-year prospective study from a referral center of interstitial lung disease and lung transplantation.
J Clin Med, 9 (2020), pp. 1606
[11]
J. Morisset, C. Johnson, E. Rich, H.R. Collard, J.S. Lee.
Management of myositis-related interstitial lung disease.
Chest, 150 (2016), pp. 1118-1128
[12]
S.C. Mathai, S.K. Danoff.
Management of interstitial lung disease associated with connective tissue disease.
BMJ, 352 (2016), pp. h6819
[13]
M.M. Hossain, J. Wilkerson, J.A. McGrath, P.N. Farhadi, C. Brokamp, M.T.F. Khan, et al.
The geospatial distribution of myositis and its phenotypes in the United States and associations with roadways: findings from a national myositis patient registry.
Front Med, 9 (2022), pp. 842586
[14]
K.Y. Sun, Y. Fan, Y.X. Wang, Y.J. Zhong, G.F. Wang.
Prevalence of interstitial lung disease in polymyositis and dermatomyositis: a meta-analysis from 2000 to 2020.
Semin Arthritis Rheum, 51 (2021), pp. 175-191
[15]
G.M. Joy, O.A. Arbiv, C.K. Wong, S.D. Lok, N.A. Adderley, K.M. Dobosz, et al.
Prevalence, imaging patterns and risk factors of interstitial lung disease in connective tissue disease: a systematic review and meta-analysis.
Eur Respir Rev, 32 (2023), pp. 220210
[16]
T. Mimori, R. Nakashima, Y. Hosono.
Interstitial lung disease in myositis: clinical subsets, biomarkers, and treatment.
Curr Rheumatol Rep, 14 (2012), pp. 264-274
[17]
P. Panagopoulos, A. Goules, A.M. Hoffmann-Vold, E.L. Matteson, A. Tzioufas.
Natural history and screening of interstitial lung disease in systemic autoimmune rheumatic disorders.
Ther Adv Musculoskelet Dis, 13 (2021),
[18]
P. Spagnolo, O. Distler, C.J. Ryerson, A. Tzouvelekis, J.S. Lee, F. Bonella, et al.
Mechanisms of progressive fibrosis in connective tissue disease (CTD)-associated interstitial lung diseases (ILDs).
Ann Rheum Dis, 80 (2021), pp. 50-143
[19]
L.A. Murray, A. Rubinowitz, E.L. Herzog.
Interstitial lung disease: is interstitial lung disease the same as scleroderma lung disease?.
Curr Opin Rheumatol, 24 (2012), pp. 62-656
[20]
G.R. Connors, L. Christopher-Stine, C.V. Oddis, S.K. Danoff.
Interstitial lung disease associated with the idiopathic inflammatory myopathies: what progress has been made in the past 35 years?.
Chest, 138 (2010), pp. 1464-1474
[21]
S. Rothwell, R.G. Cooper, I.E. Lundberg, F.W. Miller, P.K. Gregersen, J. Bowes, et al.
Dense genotyping of immune-related loci in the idiopathic inflammatory myopathies confirms HLA alleles as strongest genetic risk factor and suggests different genetic background for major clinical subgroups.
Ann Rheum Dis, 75 (2016), pp. 1558-1566
[22]
G. Cerro Chiang, T. Parimon.
Understanding interstitial lung diseases associated with connective tissue disease (CTD-ILD): genetics, cellular pathophysiology, and biologic drivers.
Int J Mol Sci, 24 (2023), pp. 2405
[23]
T.E. Fingerlin, W. Zhang, I.V. Yang, H.C. Ainsworth, P.H. Russell, R.Z. Blumhagen, et al.
Genome-wide imputation study identifies novel HLA locus for pulmonary fibrosis and potential role for auto-immunity in fibrotic idiopathic interstitial pneumonia.
[24]
H. Chinoy, F. Salway, N. Fertig, N. Shephard, B.D. Tait, W. Thomson, et al.
In adult onset myositis, the presence of interstitial lung disease and myositis specific/associated antibodies are governed by HLA class II haplotype, rather than by myositis subtype.
Arthritis Res Ther, 8 (2006), pp. R13
[25]
T. Gono, Y. Kawaguchi, M. Kuwana, T. Sugiura, T. Furuya, K. Takagi, et al.
Brief report: association of HLA-DRB1*0101/*0405 with susceptibility to anti-melanoma differentiation-associated gene 5 antibody-positive dermatomyositis in the Japanese population.
Arthritis Rheum, 64 (2012), pp. 3736-3740
[26]
T. Shao, X. Shi, S. Yang, W. Zhang, X. Li, J. Shu, et al.
Interstitial lung disease in connective tissue disease: a common lesion with heterogeneous mechanisms and treatment considerations.
Front Immunol, 12 (2021), pp. 684699
[27]
Y. Ye, Z. Chen, S. Jiang, F. Jia, T. Li, X. Lu, et al.
Single-cell profiling reveals distinct adaptive immune hallmarks in MDA5+ dermatomyositis with therapeutic implications.
Nat Commun, 13 (2022), pp. 6458
[28]
Y. Wu, Y. Li, Y. Luo, Y. Zhou, X. Liang, L. Cheng, et al.
Proteomics: potential techniques for discovering the pathogenesis of connective tissue diseases-interstitial lung disease.
[29]
S. Kolahian, I.E. Fernandez, O. Eickelberg, D. Hartl.
Immune mechanisms in pulmonary fibrosis.
Am J Respir Cell Mol Biol, 55 (2016), pp. 309-322
[30]
W. Huang, F. Ren, L. Luo, J. Zhou, D. Huang, Z. Pan, et al.
The characteristics of lymphocytes in patients positive for anti-MDA5 antibodies in interstitial lung disease.
Rheumatology, 59 (2020), pp. 3886-3891
[31]
K. Wang, J. Zhao, Z. Chen, T. Li, X. Tan, Y. Zheng, et al.
CD4+CXCR4+ T cells as a novel prognostic biomarker in patients with idiopathic inflammatory myopathy-associated interstitial lung disease.
Rheumatology (Oxford), 58 (2019), pp. 511-521
[32]
P.D. Kiely, F. Chua.
Interstitial lung disease in inflammatory myopathies: clinical phenotypes and prognosis.
Curr Rheumatol Rep, 15 (2013), pp. 359
[33]
O. Krystufková, T. Vallerskog, S.B. Helmers, H. Mann, I. Putová, J. Belácek, et al.
Increased serum levels of B cell activating factor (BAFF) in subsets of patients with idiopathic inflammatory myopathies.
Ann Rheum Dis, 68 (2009), pp. 836-843
[34]
M.L. Alberti, V. Wolff, F. Reyes, E. Juárez-León, L. Fassola, G. Carballo, et al.
Myositis-associated Interstitial lung disease: clinical characteristics and factors related to pulmonary function improvement: a Latin-American multicenter cohort study.
Reumatol Clin (Engl Ed), 18 (2022), pp. 293-298
[35]
K. Long, S.K. Danoff.
Interstitial lung disease in polymyositis and dermatomyositis.
Clin Chest Med, 40 (2019), pp. 561-572
[36]
A. Fischer, K.M. Antoniou, K.K. Brown, J. Cadranel, T.J. Corte, R.M. du Bois, et al.
An official European Respiratory Society/American Thoracic Society research statement: interstitial pneumonia with autoimmune features.
Eur Respir J, 46 (2015), pp. 976-987
[37]
A. Ceribelli, A. Tonutti, N. Isailovic, M. De Santis, C. Selmi.
Interstitial lung disease associated with inflammatory myositis: autoantibodies, clinical phenotypes, and progressive fibrosis.
Front Med (Lausanne), 10 (2023), pp. 1068402
[38]
J.C. Lega, N. Fabien, Q. Reynaud, I. Durieu, S. Durupt, M. Dutertre, et al.
The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome.
Autoimmun Rev, 13 (2014), pp. 883-891
[39]
S. Li, Y. Ge, H. Yang, T. Wang, X. Zheng, Q. Peng, et al.
The spectrum and clinical significance of myositis-specific autoantibodies in Chinese patients with idiopathic inflammatory myopathies.
Clin Rheumatol, 38 (2019), pp. 2171-2179
[40]
H. Yoshifuji.
Biomarkers and autoantibodies of interstitial lung disease with idiopathic inflammatory myopathies.
Clin Med Insights Circ Respir Pulm Med, 9 (2016), pp. 141-146
[41]
T. Mimori, Y. Imura, R. Nakashima, H. Yoshifuji.
Autoantibodies in idiopathic inflammatory myopathy: an update on clinical and pathophysiological significance.
Curr Opin Rheumatol, 19 (2007), pp. 523-529
[42]
K. Tanizawa, T. Handa, R. Nakashima, T. Kubo, Y. Hosono, K. Watanabe, et al.
The long-term outcome of interstitial lung disease with anti-aminoacyl-tRNA synthetase antibodies.
Respir Med, 127 (2017), pp. 57-64
[43]
R. Aggarwal, E. Cassidy, N. Fertig, D.C. Koontz, M. Lucas, D.P. Ascherman, et al.
Patients with non-Jo-1 anti-tRNA-synthetase autoantibodies have worse survival than Jo-1 positive patients.
Ann Rheum Dis, 73 (2014), pp. 227-232
[44]
J. Damoiseaux, J.B. Vulsteke, C.W. Tseng, A.C.M. Platteel, Y. Piette, O. Shovman, et al.
Autoantibodies in idiopathic inflammatory myopathies: clinical associations and laboratory evaluation by mono- and multispecific immunoassays.
Autoimmun Rev, 18 (2019), pp. 293-305
[45]
P.W. Wolstencroft, D.F. Fiorentino.
Dermatomyositis clinical and pathological phenotypes associated with myositis-specific autoantibodies.
Curr Rheumatol Rep, 20 (2018), pp. 28
[46]
W. Wu, L. Guo, Y. Fu, K. Wang, D. Zhang, W. Xu, et al.
Interstitial lung disease in anti-MDA5 positive dermatomyositis.
Clin Rev Allergy Immunol, 60 (2021), pp. 293-304
[47]
Y. Hamaguchi, M. Kuwana, K. Hoshino, M. Hasegawa, K. Kaji, T. Matsushita, et al.
Clinical correlations with dermatomyositis-specific autoantibodies in adult Japanese patients with dermatomyositis: a multicenter cross-sectional study.
Arch Dermatol, 147 (2011), pp. 391-398
[48]
L. Liang, Y.M. Zhang, H. Chen, L.F. Ye, S.S. Li, X. Lu, et al.
Anti-Mi-2 antibodies characterize a distinct clinical subset of dermatomyositis with favourable prognosis.
Eur J Dermatol, 30 (2020), pp. 151-158
[49]
Y. Ge, X. Lu, X. Shu, Q. Peng, G. Wang.
Clinical characteristics of anti-SAE antibodies in Chinese patients with dermatomyositis in comparison with different patient cohorts.
[50]
J. Demortier, M. Vautier, O. Chosidow, L. Gallay, D. Bessis, A. Berezne, et al.
Anti-SAE autoantibody in dermatomyositis: original comparative study and review of the literature.
Rheumatology (Oxford), (2023), pp. kead154
[51]
X. Xing, A. Li, C. Li.
Anti-Ro52 antibody is an independent risk factor for interstitial lung disease in dermatomyositis.
Respir Med, 172 (2020), pp. 106134
[52]
E. Tarricone, A. Ghirardello, M. Rampudda, N. Bassi, L. Punzi, A. Doria.
Anti-SAE antibodies in autoimmune myositis: identification by unlabelled protein immunoprecipitation in an Italian patient cohort.
J Immunol Methods, 384 (2012), pp. 128-134
[53]
J. Albayda, C. Mecoli, L. Casciola-Rosen, S.K. Danoff, C.T. Lin, D. Hines, et al.
A North American cohort of anti-SAE dermatomyositis: clinical phenotype, testing, and review of cases.
ACR Open Rheumatol, 3 (2021), pp. 287-294
[54]
Y. Zuo, L. Ye, M. Liu, S. Li, W. Liu, F. Chen, et al.
Clinical significance of radiological patterns of HRCT and their association with macrophage activation in dermatomyositis.
Rheumatology (Oxford), 59 (2020), pp. 2829-2837
[55]
L. Li, C. Liu, L. Cheng, S. Yan, H. Chen, Y. Li.
Assessment of diagnostic utility, clinical phenotypic associations, and prognostic significance of anti-NXP2 autoantibody in patients with idiopathic inflammatory myopathies: a systematic review and meta-analysis.
Clin Rheumatol, 40 (2021), pp. 819-832
[56]
N.J. McHugh, S.L. Tansley.
Autoantibodies in myositis.
Nat Rev Rheumatol, 14 (2018), pp. 290-302
[57]
M. Marzęcka, A. Niemczyk, L. Rudnicka.
Autoantibody markers of increased risk of malignancy in patients with dermatomyositis.
Clin Rev Allergy Immunol, 63 (2022), pp. 289-296
[58]
W. Jiang, J. Shi, H. Yang, X. Tian, H. Yang, Q. Chen, et al.
Long-term outcomes and prognosis factors in patients with idiopathic inflammatory myopathies based on myositis-specific autoantibodies: a single cohort study.
Arthritis Care Res (Hoboken), 75 (2023), pp. 1175-1182
[59]
J. Lilleker, A. Rietveld, S. Pye, J. Lamb.
Implications of anti-CN1A serotype in inclusion body myositis.
J Neurol Neurosurg Psychiatry, 87 (2016), pp. e1
[60]
P. Coppo, J.P. Clauvel, D. Bengoufa, E. Oksenhendler, C. Lacroix, K. Lassoued.
Inflammatory myositis associated with anti-U1-small nuclear ribonucleoprotein antibodies: a subset of myositis associated with a favourable outcome.
Rheumatology (Oxford), 41 (2002), pp. 1040-1046
[61]
K. Ussavarungsi, K. Nugent, A.K. Gerke, M.D. Krasowski, R.S. Tuetken, P.S. Lenert.
Interstitial lung disease associated with anti-PM-Scl antibody: a single center experience.
Autoimmun Rev, 18 (2019), pp. 102355
[62]
M. Casal-Dominguez, I. Pinal-Fernandez, A. Derfoul, R. Graf, H. Michelle, J. Albayda, et al.
The phenotype of myositis patients with anti-Ku autoantibodies.
Semin Arthritis Rheum, 51 (2021), pp. 728-734
[63]
A. Rigolet, L. Musset, O. Dubourg, T. Maisonobe, P. Grenier, J.L. Charuel, et al.
Inflammatory myopathies with anti-Ku antibodies: a prognosis dependent on associated lung disease.
Medicine (Baltimore), 91 (2012), pp. 95-102
[64]
Z. Betteridge, N. McHugh.
Myositis-specific autoantibodies: an important tool to support diagnosis of myositis.
J Intern Med, 280 (2016), pp. 8-23
[65]
A. Sclafani, K.M. D'Silva, B.P. Little, E.M. Miloslavsky, J.J. Locascio, A. Sharma, et al.
Presentations and outcomes of interstitial lung disease and the anti-Ro52 autoantibody.
Respir Res, 20 (2019), pp. 256
[66]
A. Xu, Y. Ye, Q. Fu, X. Lian, S. Chen, Q. Guo, et al.
Prognostic values of anti-Ro52 antibodies in anti-MDA5-positive clinically amyopathic dermatomyositis associated with interstitial lung disease.
Rheumatology (Oxford), 60 (2021), pp. 3343-3351
[67]
M. Fathi, S. Barbasso Helmers, I.E. Lundberg.
KL-6: a serological biomarker for interstitial lung disease in patients with polymyositis and dermatomyositis.
J Intern Med, 271 (2012), pp. 589-597
[68]
H. Ihn, Y. Asano, M. Kubo, K. Yamane, M. Jinnin, N. Yazawa, et al.
Clinical significance of serum surfactant protein D (SP-D) in patients with polymyositis/dermatomyositis: correlation with interstitial lung disease.
Rheumatology (Oxford), 41 (2002), pp. 1268-1272
[69]
M. Fathi, M. Dastmalchi, E. Rasmussen, I.E. Lundberg, G. Tornling.
Interstitial lung disease, a common manifestation of newly diagnosed polymyositis and dermatomyositis.
Ann Rheum Dis, 63 (2004), pp. 297-301
[70]
R.W. Hallowell, J.J. Paik.
Myositis-associated interstitial lung disease: a comprehensive approach to diagnosis and management.
Clin Exp Rheumatol, 40 (2022), pp. 373-383
[71]
V. Cottin, F. Thivolet-Béjui, M. Reynaud-Gaubert, J. Cadranel, P. Delaval, P.J. Ternamian, et al.
Interstitial lung disease in amyopathic dermatomyositis, dermatomyositis and polymyositis.
Eur Respir J, 22 (2003), pp. 245-250
[72]
I. Marie, E. Hachulla, P. Chérin, S. Dominique, P.Y. Hatron, M.F. Hellot, et al.
Interstitial lung disease in polymyositis and dermatomyositis.
Arthritis Rheum, 47 (2002), pp. 614-622
[73]
H. Yoshifuji, T. Fujii, S. Kobayashi, Y. Imura, Y. Fujita, D. Kawabata, et al.
Anti-aminoacyl-tRNA synthetase antibodies in clinical course prediction of interstitial lung disease complicated with idiopathic inflammatory myopathies.
Autoimmunity, 39 (2006), pp. 233-241
[74]
S. Palmucci, A. Di Mari, G. Cancemi, I. Pennisi, L.A. Mauro, G. Sambataro, et al.
Clinical and radiological features of interstitial lung diseases associated with polymyositis and dermatomyositis.
Medicina (Mex), 58 (2022), pp. 1757
[75]
J.C. Lega, Q. Reynaud, A. Belot, N. Fabien, I. Durieu, V. Cottin.
Idiopathic inflammatory myopathies and the lung.
Eur Respir Rev, 24 (2015), pp. 216-238
[76]
T. Vojinovic, I. Cavazzana, P. Ceruti, M. Fredi, D. Modina, M. Berlendis, et al.
Predictive features and clinical presentation of interstitial lung disease in inflammatory myositis.
Clin Rev Allergy Immunol, 60 (2021), pp. 87-94
[77]
G. Raghu, H.R. Collard, J.J. Egan, F.J. Martinez, J. Behr, K.K. Brown, et al.
An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management.
Am J Respir Crit Care Med, 183 (2011), pp. 788-824
[78]
H. Mukae, H. Ishimoto, N. Sakamoto, S. Hara, T. Kakugawa, S. Nakayama, et al.
Clinical differences between interstitial lung disease associated with clinically amyopathic dermatomyositis and classic dermatomyositis.
Chest, 136 (2009), pp. 1341-1347
[79]
L.A. Saketkoo, D.P. Ascherman, V. Cottin, L. Christopher-Stine, S.K. Danoff, C.V. Oddis.
Interstitial lung disease in idiopathic inflammatory myopathy.
Curr Rheumatol Rev, 6 (2010), pp. 108-119
[80]
T.J. Doyle, G.M. Hunninghake, I.O. Rosas.
Subclinical interstitial lung disease.
Am J Respir Crit Care Med, 185 (2012), pp. 1147-1153
[81]
B.F. Dickey, A.R. Myers.
Pulmonary disease in polymyositis/dermatomyositis.
Semin Arthritis Rheum, 14 (1984), pp. 60-76
[82]
J.W. Park, J.R. Curtis, J. Moon, Y.W. Song, S. Kim, E.B. Lee.
Prophylactic effect of trimethoprim–sulfamethoxazole for pneumocystis pneumonia in patients with rheumatic diseases exposed to prolonged high-dose glucocorticoids.
Ann Rheum Dis, 77 (2018), pp. 644-649
[83]
A. Redondo-Benito, A. Curran, A. Villar-Gomez, E. Trallero-Araguas, A. Fernández-Codina, I. Pinal-Fernandez, et al.
Opportunistic infections in patients with idiopathic inflammatory myopathies.
Int J Rheum Dis, 21 (2018), pp. 487-496
[84]
S.Y. Ji, F.Q. Zeng, Q. Guo, G.Z. Tan, H.F. Tang, Y.J. Luo, et al.
Predictive factors and unfavourable prognostic factors of interstitial lung disease in patients with polymyositis or dermatomyositis: a retrospective study.
Chin Med J (Engl), 123 (2010), pp. 517-522
[85]
L. Zhang, G. Wu, D. Gao, G. Liu, L. Pan, L. Ni, et al.
Factors associated with interstitial lung disease in patients with polymyositis and dermatomyositis: a systematic review and meta-analysis.
PLoS One, 11 (2016), pp. e0155381
[86]
J.R. Hannah, H.E. Law, T. Gordon, M. Rooney, A. Buazon, M. Adas, et al.
A systematic review and metaanalysis of predictors of mortality in idiopathic inflammatory myopathy-associated interstitial lung disease.
J Rheumatol, 50 (2023), pp. 373-383
[87]
T. Fujisawa, H. Hozumi, M. Kono, N. Enomoto, D. Hashimoto, Y. Nakamura, et al.
Prognostic factors for myositis-associated interstitial lung disease.
[88]
L. Li, H. Wang, Q. Wang, C. Wu, C. Liu, Y. Zhang, et al.
Myositis-specific autoantibodies in dermatomyositis/polymyositis with interstitial lung disease.
J Neurol Sci, 397 (2019), pp. 123-128
[89]
M. Gasparotto, M. Gatto, F. Saccon, A. Ghirardello, L. Iaccarino, A. Doria.
Pulmonary involvement in antisynthetase syndrome.
Curr Opin Rheumatol, 31 (2019), pp. 603-610
[90]
R. La Corte, A. Lo Mo Naco, A. Locaputo, F. Dolzani, F. Trotta.
In patients with antisynthetase syndrome the occurrence of anti-Ro/SSA antibodies causes a more severe interstitial lung disease.
Autoimmunity, 39 (2006), pp. 249-253
[91]
T.T. Yan, X. Zhang, H.H. Yang, W.J. Sun, L. Liu, Y. Du, et al.
Association of anti-NXP2 antibody with clinical characteristics and outcomes in adult dermatomyositis: results from clinical applications based on a myositis-specific antibody.
Clin Rheumatol, 40 (2021), pp. 3695-3702
[92]
M. Gossez, M. Levesque, C. Khouatra, V. Cottin, L. Garnier, N. Fabien.
Interstitial lung disease in an adult patient with dermatomyositis and anti-NXP2 autoantibody.
Eur Respir Rev, 24 (2015), pp. 370-372
[93]
S.A. Moll, M.G.J.P. Platenburg, A.C.M. Platteel, A.D.M. Vorselaars, M.J. Bonàs, C. Roodenburg-Benschop, et al.
Prevalence of novel myositis autoantibodies in a large cohort of patients with interstitial lung disease.
J Clin Med, 9 (2020), pp. 2944
[94]
T. Gono, Y. Tanino, A. Nishikawa, T. Kawamata, K. Hirai, Y. Okazaki, et al.
Two cases with autoantibodies to small ubiquitin-like modifier activating enzyme: a potential unique subset of dermatomyositis-associated interstitial lung disease.
Int J Rheum Dis, 22 (2019), pp. 1582-1586
[95]
M. de Vries, M.W.J. Schreurs, E.J.M. Ahsmann, M. Spee-Dropkova, F. Karim.
A case of anti-SAE1 dermatomyositis.
Case Rep Immunol, 2022 (2022), pp. 9000608
[96]
M. Fujimoto, T. Matsushita, Y. Hamaguchi, K. Kaji, Y. Asano, F. Ogawa, et al.
Autoantibodies to small ubiquitin-like modifier activating enzymes in Japanese patients with dermatomyositis: comparison with a UK Caucasian cohort.
Ann Rheum Dis, 72 (2013), pp. 151-153
[97]
I. Marie, P.Y. Hatron, S. Dominique, P. Cherin, L. Mouthon, J.F. Menard.
Short-term and long-term outcomes of interstitial lung disease in polymyositis and dermatomyositis: a series of 107 patients.
Arthritis Rheum, 63 (2011), pp. 3439-3447
[98]
S. Hayashi, M. Tanaka, H. Kobayashi, T. Nakazono, T. Satoh, Y. Fukuno, et al.
High-resolution computed tomography characterization of interstitial lung diseases in polymyositis/dermatomyositis.
J Rheumatol, 35 (2008), pp. 260-269
[99]
K.H. Yu, Y.J.J. Wu, C.F. Kuo, L.C. See, Y.M. Shen, H.C. Chang, et al.
Survival analysis of patients with dermatomyositis and polymyositis.
Clin Rheumatol, 30 (2011), pp. 1595-1601
[100]
T. Gono, K. Masui, N. Nishina, Y. Kawaguchi, A. Kawakami, K. Ikeda, et al.
Risk prediction modeling based on a combination of initial serum biomarker levels in polymyositis/dermatomyositis-associated interstitial lung disease.
Arthritis Rheumatol (Hoboken), 73 (2021), pp. 677-686
Copyright © 2023. Asociación Colombiana de Reumatología
Opciones de artículo
Herramientas
Quizás le interese:
10.1016/j.rcreu.2023.09.003
No mostrar más