Semin Respir Crit Care Med 2024; 45(03): 365-385
DOI: 10.1055/s-0044-1785536
Review Article

Review of Pulmonary Manifestations in Antisynthetase Syndrome

Mohammad I. Ghanbar
1   Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
,
Sonye K. Danoff
1   Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
› Author Affiliations
 

Abstract

Antisynthetase syndrome (ASyS) is now a widely recognized entity within the spectrum of idiopathic inflammatory myopathies. Initially described in patients with a triad of myositis, arthritis, and interstitial lung disease (ILD), its presentation can be diverse. Additional common symptoms experienced by patients with ASyS include Raynaud's phenomenon, mechanic's hand, and fever. Although there is a significant overlap with polymyositis and dermatomyositis, the key distinction lies in the presence of antisynthetase antibodies (ASAs). Up to 10 ASAs have been identified to correlate with a presentation of ASyS, each having manifestations that may slightly differ from others. Despite the proposal of three classification criteria to aid diagnosis, the heterogeneous nature of patient presentations poses challenges. ILD confers a significant burden in patients with ASyS, sometimes manifesting in isolation. Notably, ILD is also often the initial presentation of ASyS, requiring pulmonologists to remain vigilant for an accurate diagnosis. This article will comprehensively review the various aspects of ASyS, including disease presentation, diagnosis, management, and clinical course, with a primary focus on its pulmonary manifestations.


#

Idiopathic inflammatory myopathies (IIMs) constitute a group of autoimmune disorders that encompasses several myopathy forms.[1] Antisynthetase syndrome (ASyS) is a myopathy that is distinguished by the presence of antibodies targeting several aminoacyl-transfer RNA synthetases (ARSs). First described by Hochberg et al who associated anti-Jo antibodies with interstitial lung disease (ILD) in myositis patients,[2] Marguerie et al later emphasized the clinical spectrum and the triad of polymyositis, arthritis, and ILD in the presence of autoantibodies to ARSs.[3] Today, the syndrome has evolved with more identifiable autoantibodies while largely retaining its core characteristics. Although it is not included as a separate entity in the European League Against Rheumatism/American College of Rheumatology's classification of inflammatory myopathies,[1] Connors et al,[4] Solomon et al,[5] and Lega et al[6] have proposed alternative classification criteria to assist in identifying the disease. Many patients afflicted with ASyS experience significant pulmonary involvement either in isolation or in combination with other organs. This review outlines the common features of ASyS focusing on the pulmonary manifestations, their appropriate management, and outcomes.

Epidemiology

IIMs are generally rare,[7] with ASyS considered even more uncommon. But true prevalence of ASyS is affected by the slow discovery of antisynthetase antibodies (ASAs)[8] and the performance limitations between assays[9] resulting in misclassifying some patients with ASyS into dermatomyositis (DM) or polymyositis (PM). Finally, many studies of prevalence depended on muscle biopsies for diagnosis of IIM,[10] [11] [12] excluding in the process patients with amyopathic disease.[13]

Taking all these factors into consideration, a comprehensive international registry highlighted ASyS as the third most common IIM. Seventeen percent of the total 3,002 included individuals were classified as ASyS. Intriguingly, a large number of patients with AsyS were reclassified from other IIM upon retrospective review and application of Connors' proposed criteria. In a more recent update of the same registry, there was a larger proportion of ASyS that was closer to 20%.[14] In another large cohort of 1,113 IIM patients from China, 21% were found to have ASyS,[15] whereas 22% in a Canadian cohort had ASA.[16] A similar 19.4% proportion of ASyS was seen within another cohort from China.[17] A lower 14.1% of IIM had ASyS,[18] in an ethnically diverse study from a large U.S. myositis center. An even lower proportion of 11.1% were observed to have ASyS when exclusively relying on biopsies for the diagnosis of IIM.[19] Orphanet estimates that a quarter of all IIM have ASyS and predicts a prevalence of around 1/25,000–33,000.[20]

ASyS more frequently affects females, with a ratio of approximately 2–3:1.[15] [17] [18] [21] [22] Although anti-Jo-1 antibody remains the most common ASA, regional or ethnic differences may exist. Patients in East Asia and those with African heritage show higher levels of non-anti-Jo-1 positivity.[15] [17] [19] [23] [24] [25] [26] Disease onset is often between the fourth and fifth decades of life. It is unclear whether men may have a later disease onset.[27]


#

Pathogenesis

The precise underlying mechanisms leading to immune-mediated disease in IIMs are yet to be elucidated. However, the distinct mechanism between the different forms of IIM has been supported by the difference in their pathological picture. In ASyS, presence of myonuclear actin aggregation, intranuclear rod formation, and presence of atrophic fibers and perimysial fragmentation[28] are unique. Myxovirus resistance A expression, an interferon-inducible protein recognized for its high specificity as a marker for DM (98% specificity), is not displayed in ASyS.[29] These pathological distinctions have aided in the classification of ASyS as a separate entity[30] and provided insights into a differing pathogenesis.

Our current understanding of ASyS like other autoimmune disorders suggests a dysfunction in the innate and acquired immune system that is probably triggered by an environmental event. This leads to a selection of immune cells that recognize self-antigens leading to autoimmunity. This selection is probably favored under certain conditions (e.g., genetic predisposition, estrogen level, microbiome)[31] [32] [33] leading to auto-antigen production.[34] It is widely postulated that the lungs may be the origin of the disease pathogenesis. Observational studies have noted that there is higher incidence of respiratory tract disease or environmental irritant exposure preceding the manifestation of myositis.[35] [36] The resultant epithelial stress or death imposed by previous exposures is thought to cause microparticulate release that includes cytoplasmic and nuclear materials that likely include cytoplasmic ARSs. These ARSs trigger the immune system leading to the development of ASA.[37] While B cells are responsible for antibody production in these patients, activated CD4+ T cells appear integral in their generation.[38] [39] [40]

The subsequent generation of the disease phenotype is thought to be at least partially mediated by ASA causing the loss of ARS function. Most of the ARSs have pleiotropic functions and can affect both immune and nonimmune processes. For example, histidyl-tRNA-synthetase (the target of anti-Jo-1) has antioxidant, anti-inflammatory, and antisecretory properties while also involved in cellular adhesion mechanisms and activation of various immune cell subtypes.[41] [42] [43] [44] The generation of specific ASA has been shown to subsequently lead to muscle inflammation and lymphocytic predominant lung infiltrates in murine models.[37] These immune changes likely ensue in a genetically predisposed host. Single nucleotide polymorphisms in HLA-B and HLA-DR have been shown to have the strongest association with ASyS.[45] [46] [47] [48] Despite the ubiquitous presence of ARS, the tropism of ASA to certain organs is poorly understood. Since immune dysfunction is thought to be triggered in the lung, its involvement is present in the majority of patients.


#

Clinical Features

There are similarities in the presentation of ASyS: including some combination of fever, myositis, ILD, inflammatory polyarthritis, Raynaud's phenomenon (RP) or mechanic's hand (MH). The heterogeneity in manifestations is thought to be largely driven by the different ASAs.[49] The classic triad of ILD, myositis, and arthritis has only been seen in 15% of patients included in a large international cohort,[22] and a similar 14% in a smaller ILD-specific cohort from China.[50] The proportion of patients with the classic triad rose to 44% during follow-up,[22] highlighting the evolving nature of ASyS.

Muscle Disease

Muscular involvement in ASyS can occur in the form of myalgia and/or weakness, with a subset of patients also suffering from dysphagia.[19] [51] The prevalence of muscle weakness and myalgia varies significantly across different cohorts of ASyS, ranging from 41 to 100% for muscle weakness and 30.4 to 88.9% for myalgia,[14] [15] [17] [19] [22] [52] [53] [54] [55] while primarily involving the proximal muscles.[19] The presence of anti-OJ appears to confer more severe muscle involvement, while anti-PL-12 has milder forms of muscle disease.[56] Despite this, anti-Jo-1 positivity remains the highest predictor for muscle involvement.[22] Respiratory muscle involvement has also been documented to occur in approximately 8%.[19]


#

Joint Disease

Arthritis and arthralgia are common in patients with ASyS. The prevalence may vary depending on the studied cohort and the underlying ASA profile. In smaller studies joint involvement ranged between 12[18] and 88%.[57] However, when examining larger patient samples, the frequency is closer to 65%.[22] [53] [54] [55] Individuals with anti-Jo-1 antibodies are more likely to have joint involvement compared with others.[22] Joint disease is often symmetrical, polyarticular, and predominantly affects the hands and wrists. While predominantly nonerosive in nature, some patients may develop erosive arthritis with concomitant seropositivity for anti-rheumatoid factor (anti-RF) antibody or anti-citrullinated protein (anti-CCP) antibody, potentially causing misclassification as rheumatoid arthritis.[53] [58] [59] [60] Erosive arthritis has also been documented in up to 35%, even in the absence of anti- RF antibody or anti-CCP antibody.[61] A substantial number of patients also exhibit oligoarthritis or asymmetrical arthritis, with more than 70% of anti-OJ patients presenting with such features.[22]

Cutaneous

RP is a significant manifestation of ASyS and has been reported to occur in approximately 39% of patients in large international registries.[14] [22] RP has also been shown to be the initial symptom in patients with anti-Jo-1 profiles and may predispose to an arthritis phenotype.[53] [54] Another hallmark sign of ASyS is MH, a nonpruritic hyperkeratotic, fissured, scaly, eruptions mainly affect the palms and radial aspects of the hands and index finger. These occur in nearly 40% of ASyS although reported more variably in smaller sized studies. More importantly, MH often improves with treatment and can serve as a sign of uncontrolled disease or a flare that may include the lungs.[62] Additionally, while MH is an important feature of ASyS, it is not pathognomonic and can be present in DM.[14] Conversely, cutaneous manifestations of DM including Gottron's sign, heliotrope rash, shawl sign, periungual erythema, digit ulceration, calcinosis, and the holster sign can be observed in ASyS in varying frequencies, reaching up to 44%.[14] [17] [51] Another cutaneous finding that resembles MH is hiker's feet ([Fig. 1]) with similar features involving the toes and feet and often occurring in conjunction with MH.[63]

Zoom Image
Fig. 1 Extrapulmonary manifestations of ASyS. (A) V-neck rash. (B) Gottron's papules. (C) Gottron's papules. (D) Mechanic's hand. (E) Mechanic's feet. (F) Calcinosis. (G) Ragged cuticles and periungual erythema. ASyS, antisynthetase syndrome.

#

Other Features

Fever affects over one-third of patients throughout their clinical course.[19] [22] [51] [53] There is a suggestion that patients positive for anti-EJ or anti-PL-12 are more likely to suffer from fever, but these findings have not been replicated elsewhere.[15] [22] Individuals may additionally suffer from sicca symptoms, reported to affect up to 33%[25] and possibly associate with anti-KS antibody positivity.[64] Cardiac involvement has also been detected in patients with ASyS. In the most recent published cohort, this accounted for 7% of a 649 patient registry.[14] Cardiovascular manifestations include myocarditis, arrhythmias, elevated cardiac enzymes in the absence of a coronary insult, pericarditis, and pericardial effusion.[49]


#
#
#

Pulmonary Manifestations

Interstitial Lung Disease

ILD stands out as one of the most important and characteristic feature of ASyS. Findings from large patient cohorts and registries indicate that ASyS-associated ILD (ASyS-ILD) occurs in up to 68 to 94%,[14] [17] [22] [53] [55] affirming the high prevalence observed in smaller studies.[25] [65] [66] In patients with anti-Jo-1 antibodies, the most common form of ASyS, 29 to 84% have been noted to have lung disease across a wide range of studies.[49] However, ILD rates appear to be higher in anti-EJ positive patients noted as 90 to 100%.[17] Importantly, despite anti-EJ positive patients appearing to have the highest prevalence of ILD, previous reports suggested that anti-PL-7- and PL-12-positive patients more commonly have ILD.[18] [67] This assertion has been somewhat challenged by data from the international study of 828 ASyS patients conducted by the American and European Network of Antisynthetase Syndrome (AENEAS). The study demonstrated that at disease onset, 69% of anti-PL-12 and 73.7% of anti-EJ positive patients had ILD, and both percentages were significantly higher than in patients with anti-Jo-1 antibodies (p = 0.001 and p = 0.005, respectively). This suggests that anti-EJ-positive patients may have a more prominent association with ILD. Nevertheless, in a group of 233 patients with anti-Jo-1, anti-PL7, and anti-PL12 antibodies, hierarchical cluster analysis revealed that patients positive for anti-PL7 and anti-PL12 exhibited a more lung-restricted phenotype correlating with the higher occurrence of lung involvement in these two groups.[55] Together, these data support the notion that patients with anti-PL7, PL-12, and EJ antibodies present a phenotype characterized by high pulmonary involvement. Moreover, ethnicity may play a role in the development of lung disease in ASyS. Black individuals are more likely to have non-Jo 1 serology[18] [68] and when compared with white patients, had worse ILD with an 18% lower forced vital capacity (FVC; p < 0.001) and 12% lower diffusing capacity of the lungs for carbon monoxide (DLCO; p < 0.01), independent of the antibody status.[18]

While some may have subclinical ILD, patients often present with cough or dyspnea on exertion.[53] Although many individuals with ASyS present with a gradual onset of symptoms, a substantial number manifest with acute-onset ILD across all antibody subtypes. Close to half the patients from a 32-patient study in France[69] required supplementary oxygen less than 9 weeks from symptom onset. In total, 21% of patients from another small study who were seropositive for anti-Jo-1 were additionally classified to have acute onset of ILD,[52] while a similar percentage was seen in a 234 patient Chinese cohort with a more expansive ASA profile.[15] Patients from the large AENEAS study showed that 20 out of 38 (74.1%) anti-EJ patients had acute onset of ILD defined as onset and worsening of symptoms within a 4- to 6-week time frame. Strikingly, the rate was 37.5 to 48.3% in the other antibody groups (anti-Jo-1, anti-PL7, andti-PL12, and anti-OJ were available).[22] Yet, there are contrasting data showing a much lower 9.6% rate of acute-onset ILD in a Spanish cohort of 148 anti-Jo-1-positive patients.[53] Similarly, a Chinese study demonstrated a comparable low rate of 8.9% of 124 patients with a diverse antibody profile that developed acute ILD despite their more liberal definition (worsening dyspnea and hypoxemia within 3 months of symptom onset).[17] Certainly, while the data may be conflicting, there is substantial evidence supporting the occurrence of acute ILD. Analysis of bronchoalveolar lavage fluid in patients with acute ILD demonstrated a neutrophil-predominant cell differential, significantly higher than their nonacute counterparts (66.9% vs. 29.1% respectively, p = 0.004).[69] This may suggest an element acute lung injury (ALI).

Co-existence of anti-Ro-52 antibodies has been shown by several investigators to associate with worse lung disease in ASyS.[17] [23] [70] [71] [72] [73] [74] However, this has not been universally observed especially in larger more diverse populations.[18] [53] [55] [75] Based on these discrepant reports, it is unclear to what degree anti-Ro-52 contributes to worse pulmonary manifestations in ASyS.

A restrictive physiology is typically seen in ASyS. In the assessment of FVC in 96 subjects with anti-Jo-1-positive antibodies, a mild restrictive physiology was observed with a mean of 65.7% with a corresponding mean total lung capacity (TLC) of 67.2% predicted.[76] Additionally, DLCO was moderately reduced, with a mean of 56.3% predicted. In another study with multiple ASA-positive patients, the baseline FVC and DLCO across the different ASA groups was mild to moderately reduced at 63.3 to 71.2% predicted and 57.6 to 68% predicted respectively with no significant differences.[77] In cases where there is a more significant reduction in FVC, TLC, or vital capacity than would be expected based on radiological disease involvement, respiratory muscle weakness should be considered.[9] Measurement of the maximum inspiratory pressure, the maximal sniff nasal inspiratory pressure, and the maximum expiratory pressure can help obtain more insight into the strength of different respiratory muscle groups.[78] [79]


#

Imaging

High-resolution computed tomography (HRCT) imaging of individuals with ASyS is essential for characterization and assessment of disease burden. Changes observed in ASyS-ILD include ground glass opacities (GGOs), air space opacities, traction bronchiectasis, reticulation, interlobular septal thickening, and volume loss indicative of ILD ([Fig. 2]). These changes are often seen in the lower lobes or sometimes diffusely, with peripheral or peribronchovascular predominance. The imaging findings correspond to a particular radiographic pattern in most cases. In an analysis of 33 patients with anti-Jo-1, anti-PL7, and anti-PL12 antibodies, all demonstrated GGOs at diagnosis with almost half of the patients having concomitant consolidative changes.[80] A nonspecific interstitial pneumonia (NSIP) pattern was most common (45%), while organizing pneumonia (OP) or a mixed NSIP-OP CT pattern occurred in 21 and 24%, respectively. Waseda et al reviewed retrospectively 64 patients with ASyS-ILD in patients with anti-EJ, anti-KS, and anti-OJ.[81] GGOs dominated HRCT changes in tandem with reticulations and interlobular septal thickening. Distribution of changes were almost exclusively peripheral and lower zone predominant. NSIP and OP were the principal radiographic patterns occurring in 55.6 and 41.2% of patients, respectively. In another review focusing on 51 anti-EJ-positive patients with ILD, GGOs were again universally observed.[82] Changes consistent with NSIP or OP were again primarily observed suggesting no clear difference in presentation of ASyS-ILD across the different antibody profiles. Whereas radiographically, NSIP is most frequently observed in ASyS, Zhan et al reported on 108 patients where OP appeared more commonly than NSIP representing 49.1%.[77] Liu et al[82] also found that in patients with anti-EJ-positive antibodies (n = 23, 78.3%) had imaging consistent with OP.

Zoom Image
Fig. 2 Different CT patterns in patients with ASyS. (A) Ground glass opacities, reticular thickening and subpleural sparing consistent with a NSIP in a patient with anti-PL12 serology. (B) Ground glass opacities, reticular thickening, subpleural sparing, and traction bronchiectasis consistent with a fibrotic NSIP in a patient with anti-PL7 serology. (C) Interstitial infiltrates with ground glass opacities, alveolar nodules, and subpleural bands consistent with an OP in a patient with anti-Jo1 serology. (D) Diffuse subpleural reticulation, fibrosis, traction bronchiectasis, and honeycombing consistent with an UIP pattern in a patient with anti-Jo1 serology. ASyS, antisynthetase syndrome; CT, computed tomography; OP, organizing pneumonia; NSIP, nonspecific interstitial pneumonia; UIP, usual interstitial pneumonia.

While NSIP, OP, or an NSIP/OP overlap patterns are most frequently observed, other changes have also been described. Usual interstitial pneumonia (UIP) has been observed in a minority of patients with ASyS. Zamora et al found that 13% of their anti-Jo-1 patients demonstrated a possible UIP pattern on imaging,[76] but these changes have been seen at rates of 3.7% or less in other retrospective analysis.[77] [81] [82] Notably, a proportion of patients with UIP may demonstrate anterior upper lobe, straight edge, or/and an exuberant honeycombing sign.[83] Other investigators have reported on imaging consistent with acute interstitial pneumonia (AIP), but this represents a very small percentage of patients. In a multicenter retrospective analysis from the United Kingdom, only 1 out of 76 patients with ASyS had imaging consistent with diffuse alveolar damage (DAD), corresponding to AIP.[84] However, a smaller study noted a higher percentage of patients with AIP (18%).[85] Other reports have been equally variable with AIP pattern in 0 to 6% of their cohorts.[86] [87] [88] [89] [90] More conclusively, in a systematic review and meta-analysis involving 306 patients with available radiographic information, NSIP was most common (67%), followed by OP (24%) and UIP (9%), while AIP represented 1% and 3% remained unclassifiable.[91]


#

Histopathology

Radiographically, NSIP and OP patterns dominate, though histological changes do not completely correspond. It is important to stress, however, that histological evaluation of patients with ASyS is limited due to factors such as the underlying severity of illness at baseline, concerns about disease flare with biopsy, and perceived lack of benefit or change in management obtained from a biopsy. In a group of 41 patients with ASyS for whom 18 surgical lung biopsy results were available, there was an equal distribution of OP, NSIP, and UIP histological findings.[68] The mean time interval between ILD diagnosis and undergoing a biopsy was 15 months, a factor that may affect tissue evolutionary changes. In a case series of 39 subjects who were anti-Jo-1 positive with biopsy data, OP and NSIP (26 and 23% of cases, respectively) were the most common findings with UIP and DAD representing close to 15% of the cases each.[76] In an autopsy-focused histological assessment of 22 patients with anti-Jo-1 serology, there was a predominance of DAD (55%) and UIP (45%) changes with a minority having a NSIP subtype (14%) (some had multiple histological patterns).[66] These findings should be interpreted with caution given the postmortem nature of the study.

Looking at ASA groups separately, in 12 patients with anti-PL12 serology for which lung histology was available, the majority showed a UIP pattern (8/12, 66.7%) with an equal remaining number for NSIP and OP (2 each, 16.7% each).[92] In other reports that included smaller numbers of PL-12-positive patients, UIP was seen in 60%, OP and NSIP in 20% each (5 total patients),[68] while NSIP was seen in all the patients included in another study (2 total patients).[93] For PL-7 positive patients, one study showed equal distribution of NSIP, OP, and UIP (3 total patients).[68] Another demonstrated UIP in 50%, OP in 25%, and NSIP in 12.5%[94]; while a total of 5 biopsies available in one report showed UIP in 60%, and both OP and NSIP in 20% each.[95] In a group of 12 patients with anti-EJ, biopsy results that were obtained in a median interval of 28 days from the date of the HRCT, 75% had NSIP while the remainder were unclassifiable.[96] A smaller cohort of four patient positive for anti-EJ also echoed these findings with 75% exhibiting NSIP while the remainder had UIP.[97] Among four patients with anti-EJ, half of them had DAD while the remainder had UIP.[98] In the largest cohort of anti-KS positive patients, 14 surgical lung biopsy results were available; 85.7% had NSIP, whereas the remaining 14.3% were unclassifiable.[99] Interestingly, the authors categorized any signs of ALI on biopsy as unclassifiable, arguing for the possibility of higher rates of DAD. In contrast, surgical lung biopsies from five patients exhibited predominantly UIP changes (80%) with the final patient showing an OP pattern.[100] Limited reports are available for histopathological findings in anti-OJ-positive patients. However, in a study from Japan, surgical lung biopsies were available in 3, showing an equal distribution of NSIP, UIP, and COP.[101] Finally, no biopsy results have been reported for other rare ASA profiles including anti-Zo and anti-YRS.

Flashner et al collated all the reported histopathological reports of individuals with ASA-associated ILD allowing for a better overview of pathological patterns in ASyS.[102] NSIP remained the most prominent manifestation for patients with anti-Jo-1, anti-KS, and anti-EJ. Anti-PL12 patients had significantly more UIP pattern than other antibody profiles, and both anti-KS and anti-EJ patients were statistically least likely to have changes consistent with OP. Anti-EJ and anti-Jo-1 patients had the largest proportion of histological ALI changes. Overall, NSIP remained the most common pathological finding accounting for 41%, UIP for 24%, OP for 19%, and ALI representing 13.5%. This contrasts with the radiographic findings where NSIP was more frequently seen (67%), OP (24%) was more common than UIP (9%), and AIP was only described in 1%. Part of this discrepancy may be explained by the inclusion of multiple pathology patterns on each biopsy report, and the more granular detail that arises from microscopic assessment of tissue biopsies. The biopsy discrepancy from imaging findings may additionally be driven by the mode of biopsy (surgical vs. transbronchial), the interval between the initial HRCT, and/or the motivation for biopsy acquisition (e.g., worsening clinical picture) that may predispose to more ALI pathologic changes.


#
#

Pulmonary Vascular Disease

Pulmonary vascular involvement in the form of pulmonary hypertension (PH) may be difficult to categorize due to the recent change in the diagnostic parameter that lowered the threshold of PH diagnosis.[103] Regardless, PH in ASyS is often attributed to group 3 disease owing to the extensive lung involvement in many patients and limited reports of group 1 (connective tissue disease [CTD]-related) disease exists.[104] [105] [106] [107] [108] In reports where pulmonary arterial hypertension (PAH) is diagnosed, this is often due to either limited parenchymal disease and/or hypoxemia, or when the PH is out of proportion to the degree of ILD. The prevalence of PH in ASyS can be overestimated when patients are solely screened with transthoracic echocardiography (TTE), as demonstrated by Abel et al.[104] In their cohort, there was TTE evidence of PH in 23%. Of their cohort, 16 of 21 were confirmed on right heart catheterization (RHC). Overall, most large cohorts that reported PH based on RHC estimated variable prevalence of 7.9 to 20%.[54] [104] [109] [110] Reports of successful management of PH-associated ASyS have been inconsistent with several case reports documenting improvement in symptoms and hemodynamics upon treatment with PAH-specific therapies.[104] [111] However, in the largest collection of patients with RHC diagnosed precapillary PH, where 19 patients were started on PAH-specific therapy, improvement was not seen.[111]

Many patients with a diagnosis of PH have additional serology for anti-Ro-52. Whether this is an associated marker for PH development or related to worse pulmonary disease that is reflected in a higher rate of group 3 PH is difficult to discern from the available literature. Overall, while there is clear evidence for precapillary PH in patients with ASyS, it is unclear which individuals may suffer from PAH (group 1 disease). Therefore, better characterization and patient selection may be necessary to establish benefits from PAH-specific therapy.


#

Pleural Disease

Pleural involvement is rare in ASyS and should prompt investigation to rule other causes. Nevertheless, there have been a handful of reports describing primarily pleural effusions developing in patients with ASS. A 69-year-old female who presented with fever and left-sided eosinophilic pleural effusion that was unresponsive to antibiotics[112] later developed other signs of ASyS and was positive for PL-7 antibody. Her pleural biopsy showed evidence of intercostal muscle myositis and pleuritis requiring immunosuppression to control her disease. Another young female with anti-Jo-1-positive serology presented with OP but quickly progressed without treatment to arthralgia, profound weakness, MH, and bilateral pleural effusion.[113] The pleural fluid was exudative and lymphocytic but sterile. Her effusions resolved with immunosuppression. Other case reports show a similar sterile exudative pleural effusion that resolves when the disease activity is controlled, suggesting an element of ASyS-mediated pleuritis.[114] [115] [116] [117] Interestingly, in an extensive retrospective analysis for evidence of serositis in ASyS, 93 patients were isolated.[118] Pleural effusions were present in 44% of patients, 75% of which were bilateral. Patients with PL-12 antibodies were statistically more likely to develop pleural effusions (p = 0.003). This study demonstrated a higher rate of pleural effusions in ASyS than reported elsewhere, and may be limited by its retrospective nature, ability to attribute the pleural effusions to ASyS, and the lack of exclusion of other etiologies for pleural effusions. In conclusion, pleural involvement in ASS remains limited in large registry databases but can occur in select patients.

Mode of Presentation

The initial association of anti-Jo-1 antibody with myositis, ILD and MH has led to the description of a triad presentation in ASyS. However, the presentation of ASyS can be widely variable. In those who lack the triad, either isolated organ involvement or a combination of other symptoms may constitute their initial presentation. In a meta-analysis from 2014, individuals with ASA often presented with arthralgia (62%) or ILD (72%).[119] It is unsurprising that the pleiotropic presentation of ASyS may be at least partially driven by the specific ASAs. In a large American and European cohort of patients,[22] the mode of presentation was slightly different according to the antibody profile. For individuals with anti-Jo-1, isolated arthralgia was the main presentation in 22%, whereas ILD predominated the initial presentation in those with anti-PL-7 (36%), anti- PL-12 (43%), and anti-EJ (39%). This emphasizes the necessity for pulmonologists to maintain a high index of suspicion when evaluating patients with ILD. Interestingly, presentations with isolated myositis (10–17%), arthritis (11–23%), as well as a combination of arthritis + myositis (3–13%), or arthritis + ILD (6–24%) were not statistically different across the remainder of antibody profiles in the same cohort. Overall, isolated organ involvement at presentation occurred in 64% of the study participants. Antibody profiling additionally predicted the likelihood of progression to include lung disease in the same study.[22] This was predominantly observed in patients with anti-Jo-1, where 63% developed new-onset ILD during follow-up, and a comparable 60% increase was noted in patients with anti-EJ. New ILD was seen in approximately 50% for both anti-PL-17 and anti-PL12, but only in 30% of anti-OJ. Therefore, while ILD remains as a major component of ASyS, it can occur as a primary or subsequent presentation.


#

Antisynthetase Antibodies

tRNA synthetases are a family of enzymes found in either the cytosol or mitochondria and are involved in cellular protein synthesis.[120] In total, there are 20 tRNA synthetases, with autoantibodies discovered against many of them. However, only 10 (originally 8 with recently discovered anti-Ly[121] and anti-VRS[122]) have been directly linked with ASyS. Other ASA such as anti-SC and anti-WARS have been linked to other autoimmune diseases.[123] [124] [125] Anti-Jo-1 was first discovered in 1983,[126] followed quickly by others and later linking them to autoimmunity and myositis. While most tRNA synthetases can function independently, eight of them require binding together in a complex known as the multi-tRNA synthetase complex.[51] Anti-OJ antibodies primarily target the isoleucyl-tRNA synthetase component of this complex.[51]

Anti-Jo-1 antibodies are the most common ASA amongst patients with ASyS although its prevalence varies according to the cohort. Given that anti-Jo-1 is more readily available for testing compared with other ASAs, this may lead to disparate detection rates compared with other ASA. ASyS may behave differently according to the specific ASA; clinical distinctions amongst various ASAs are outlined in [Table 1]. Moreover, antibody levels can fluctuate through the disease course depending on disease activity and in response to treatment.[127] [128] [129]

Table 1

Antisynthetase antibodies and prevalence

Antisynthetase antibody

tRNA synthetase auto-antigen

Prevalence (%)[14] [15] [16] [17] [18] [19] [22] [23] [24] [25] [26] [27] [50] [77] [88] [89] [250] [256] [257] [259] [260] [263] [264] [265] [266] [267] [268] [269]

Features

Anti-Jo1

Histidyl

60–65

Often have classic triad

Anti-PL7

Theronyl

15–20

More severe ILD, higher rates of esophageal involvement

Anti-PL12

Alanyl

10–15

More severe ILD, higher rates of esophageal involvement, lower myositis rates, worse survival

Anti-OJ

Components of MSC/isoleucyl

1–5

Possibly affects older patients, lower rates of arthritis, mainly asymmetric arthritis.[22] May be associated with malignancy.[270]

Anti-EJ

Glycyl

5–10

More acute onset ILD, low rates of arthritis at presentation, worse survival

Anti-KS

Asparaginyl

<1

Mainly ILD, low rates of arthritis, myositis rare

Anti-Zo

Phenylalanyl

<1

Often have classic triad

Anti-YRS/Ha

Tyrosyl

<1

Rare, no distinct features

Anti-Ly

Cysteinyl

<1

Rare, no distinct features

Anti-VRS

Valyl

<1

Rare, no distinct features

Abbreviations: ILD, interstitial lung disease; MSC, multienzyme synthetase complex.



#

Diagnosis

Diagnosis of ASyS is made using one of the proposed criteria outlined in [Table 2]. Given the heterogenous presentation of ASyS, its evolving nature, and overlapping symptoms with other disease, the performance of specific criteria will differ. There is a lack of evidence in the literature of how these differing criteria perform. All the criteria currently available include the presence of ASA as a prerequisite for the diagnosis but differ in the number and type of additional symptoms required. Currently, there are no existing data-driven or validated classification criteria for ASyS. A recent systematic review revealed numerous variabilities in defining ASyS across the literature, coupled with a lack of sensitivity and specificity of its clinical variables including muscle biopsy findings.[130]

Table 2

Diagnostic criteria for antisynthetase syndrome

Connors[4]

Solomon[5]

Lega (revised Connors)[6]

Anti-aminoacyl-tRNA synthetase autoantibody positivity plus one among:

• Myositis (Bohan and Peter's criteria)

• ILD by ATS criteria

• Arthritis (clinic, X-rays, self-report)

• Unexplained fever

• Raynaud's phenomenon

• Mechanic's hands

Anti-aminoacyl-tRNA synthetase autoantibody positivity

plus (2 major criteria or 1 major plus 2 minor criteria)

Major criteria:

• Myositis (Bohan and Peter's criteria)

• ILD by ATS criteria

Minor criteria:

• Arthritis

• Raynaud's phenomenon

• Mechanic's hands

Anti-aminoacyl-tRNA synthetase autoantibody positivity plus one among:

• Myositis (overt or hypomyopathic)

• ILD by ATS criteria

• Arthritis or arthralgia

Or two among:

• Unexplained fever

• Raynaud's phenomenon

• Mechanic's hands

Abbreviations: ATS, American Thoracic Society; ILD, interstitial lung disease.


In our practice, we have a high index of suspicion for ASyS even in the absence of extrapulmonary symptoms, especially in patients with an NSIP, OP, or NSIP-OP overlap pattern of ILD; and routinely screen for ASA. Despite the above-mentioned limitations, we rely on Connor's criteria because of our long-standing experience with it and its relative simplicity. We therefore seldom send patients for lung biopsies. We prefer a comprehensive myositis panel that utilizes multiple different assays for antibody detection including immunoprecipitation (considered the gold standard).[131] We also screen for other organ involvement when indicated (e.g., magnetic resonance imaging for myositis[132] [133]).


#
#

Management of Pulmonary Manifestations

Much of the management of ASyS-ILD is extrapolated from care established in IIM or other CTD-associated ILD (CTD-ILD). Therefore, the mainstay of treatment is immunosuppression. The use of immunosuppressive therapies has shown to reduce the pulmonary disease burden reflected by functional improvements in IIM-associated ILD (IIM-ILD).[134]

Treatment for ASyS-ILD can be challenging with presentations varying from asymptomatic, to chronically progressive, or rapidly progressive ILD (RP-ILD). In patients with significantly clinical ILD, disease severity can range from mild to life-threatening. Additionally, patients with stable lung disease can have episodes of acute exacerbations that can be unpredictable necessitating alteration in their therapy. Differences in medication pharmacokinetics are also important to keep in mind. Glucocorticoids have a pivotal role in initial therapy, but use of steroid-sparing agents is necessary to mitigate steroid side effects. mycophenolate mofetil (MMF) and azathioprine (AZA) take longer time to reach their maximum effect and therefore appropriate for mild–moderate ASyS-ILD, whereas tacro and intravenous immunoglobulin (IVIG) have a more rapid onset making them more appealing for severe disease and acute exacerbations. Some patients also exhibit resistant disease despite therapy with corticosteroids and two immunosuppressive agents forcing the trial of other therapies that include rituximab (RTX) and IVIG. An algorithm for management is presented in [Fig. 3].

Zoom Image
Fig. 3 Algorithm suggesting management for antisynthetase associated ILD (ASyS-ILD) according to severity of disease. Decision for the use of chronic immunosuppression and agent of choice is governed by several variables including disease severity, response to initial therapy, familiarity of physician with medication, co-morbidities, and disease course. AZA, azathioprine; ECMO, extracorporeal membrane oxygenation; IVIG, IV immunoglobulin; MMF, mycophenolate; PFT, pulmonary function test; PLEX, plasma exchange; RTX, rituximab; TAC, tacrolimus. Adapted from Hallowell & Danoff.[145]

Nonpharmacological treatment also plays a crucial role in the care of patients with ASyS-ILD. Supportive measures include supplemental oxygen therapy as well as adequate prevention of infections. With advances in medical practices, patients with acute exacerbations may benefit from a tailored approach when hospitalized in the intensive care unit (ICU). Finally, lung transplantation may be an option for some patients with ASyS-ILD unresponsive to therapy and should be considered on a case-by-case basis.

Immunosuppression

Corticosteroids

Corticosteroids are the backbone of initial therapy in ASyS-ILD. Much of the data for their use stem from observational studies in patients with IIM that have shown either improvement or stability of disease with their use.[135] [136] [137] Specifically in ASyS-ILD, 37 patients classified as dermatomyositis (DM)/polymyositis (PM) with concurrent presence of ASA had improvement in the pulmonary function tests (PFTs) and/or HRCT findings with corticosteroids being part of their management.[138] In other case series, 28 (42%) of 66 patients[65] and 6 (43%) of 14 patients with anti-PL-7 positivity[95] showed favorable response with corticosteroid therapy alone. There are currently no available indicators that can predict steroid responsiveness in ASyS-ILD. The literature suggests that patients with a more prominent inflammatory component in NSIP or OP pattern may exhibit more favorable responses to corticosteroids.[138] [139] [140] [141]

Consistent with expert opinion, we recommend the initiation of prednisone at 0.5 to 1 mg/kg/day depending on disease severity with monitoring for improvement of symptoms.[142] [143] [144] [145] For patients with severe disease, RP-ILD, acute exacerbations, or impending respiratory failure, we administer high-dose intravenous methylprednisolone 1 g daily for 3 days. In these patients we also simultaneously initiate a second immunosuppressive agent with a rapid onset, such as IVIG before transitioning to oral prednisone 1 mg/kg/day. Lastly, we include a steroid sparing agent.

We favor maintaining the steroid therapy for 1 month and if improvements are observed in symptoms, PFTs, and imaging, prednisone is then tapered down to 40 mg/day. Subsequently, we decrease prednisone by 25% every 2 to 4 weeks until reaching a dose of 20 mg/day, followed by a slower taper of 2.5 mg/day every 2 weeks. Based on our experience, we have noted severe or refractory ILD flares during tapering of glucocorticoids. Therefore, we maintain the prednisone dose at approximately 5 to 10 mg/day while continuing the steroid sparing agent until improvement or stabilization of symptoms, lung function tests, and CT abnormalities have plateaued. Once stabilized on a steroid sparing agent, prednisone can be weaned off with care to identify any evidence of symptomatic worsening or flare and a low threshold to escalate the steroid dose in the setting. Prophylaxis for Pneumocystis jirovecii is strongly recommended in ASyS-ILD patients.


#
#

Steroid Sparing Agents

Purine Synthesis Inhibitors—Azathioprine and Mycophenolate Mofetil

AZA, a prodrug, functions primarily via its conversion into 6-mercaptopurine to interfere with DNA and RNA synthesis in leukocytes thereby limiting their proliferation and effector function.[146] The metabolism of 6-mercaptopurine depends on thiopurine methyltransferase (TPMT). Patients can undergo screening for TPMT deficiency, as it may result in increased AZA toxicity. MMF, also a prodrug, inhibits the formation of de novo guanine nucleotides, necessary for the generation of DNA; this leads to the inhibition of lymphocyte proliferation, leukocyte recruitment, and cytokine production.[147] [148] No direct comparison of these two drugs exists, but their efficacy in controlling disease and improving lung function has largely been established from observational studies. In an analysis of 66 patients with ASyS-ILD, 20 subjects dependent on AZA or MMF for ILD improvement.[65] In a larger retrospective review of 110 patients with myositis-related ILD, over 60% had ASA, significant improvements in PFTs, and steroid lowering effect was achieved with the use of either MMF or AZA.[149]

We start AZA at a dose of 50 mg/day before the dose is increased by 50 mg every 1 to 2 weeks with ongoing monitoring for any blood dyscrasia or liver toxicity until reaching a dose of 2 to 2.5 mg/kg day (typically not exceeding 200 mg/day). For those with intermediate TMPT levels, lower doses may be necessary, and we avoid AZA in individuals with low TMPT levels. We prefer AZA for patients considering conception. We typically initiate MMF at 500 mg twice daily, aiming for a target dose of 2 to 3 g/day. In some patients, MMF may not be well-tolerated due to gastrointestinal side effects and can be substituted with its active metabolite mycophenolic acid.


#

Calcineurin Inhibitors

Cyclosporin A (CsA), a cyclic peptide, and tacrolimus (TAC), a macrolide antibiotic bind to a family of cytoplasmic proteins that inhibit calcineurin suppressing cell mediated and humoral immune responses.[150] [151] Both CsA and TAC have been shown to be effective steroid sparing agents in the treatment of ASyS-ILD.[69] [152] [153] [154] In a cohort of 17 anti-Jo-1 positive patients with ILD, CsA was effective at significantly improving FVC and DLCO, reducing prednisone to a median of 2.5 mg/day, and adequate control of disease as evident by ILD flares in 75% of patients who had interruptions in their CsA therapy.[153] TAC is often preferred over CsA due to it being more potent and its perceived greater efficacy primarily in myositis-associated ILD.[155] [156] In 13 patients with ASyS-ILD, TAC was successful at significantly improving both FVC and DLCO values, control of myositis, and significant reductions in corticosteroid dosing.[157] In a larger retrospective review of 57 ASyS-ILD patients, CsA (n = 31) and TAC (n = 26) were used in all patients and were associated with significant improvement in pulmonary functions with near normalization of FVC and DLCO.[158] Extrapolating from the myositis literature, early use of calcineurin inhibitors in the clinical course of ILD has been linked with better survival, encouraging its earlier incorporation in the treatment plan.[159]

CsA is dosed 2 to 5 mg/kg twice daily while monitoring and maintaining a blood trough level of 100 to 150 ng/mL. TAC is started at 0.075 mg/kg twice daily with similar monitoring for blood trough levels. Many studies using TAC for IIM targeted a trough of 5 to 10 ng/mL.[155] [157] [158] [160] [161] However, in our practice, we find maintaining a more conservative trough of 3 to 6 ng/mL achieves excellent disease control with a more favorable side-effect profile.


#

Cyclophosphamide

Cyclophosphamide (CYC) is an alkylating agent that primarily works by cross-linking DNA strands leading to reduced replication function in lymphocytes. It has been mainly employed in patients with severe ILD or RP-ILD. In a retrospective analysis of 62 patients with ASyS-ILD, where CYC and RTX were used in 34 and 28 cases respectively, pulmonary progression free survival (85 and 92%, respectively) and steroid dosing at 6 months were similar.[162] The authors did note that survival was higher at 2 years in the RTX group (24% CYC vs. 54% RTX). It should be noted that there was some selection bias in this study, as patients in the CYC group had significantly lower lung function tests, while those started on RTX had more refractory disease at baseline. In a meta-analysis of myositis-associated ILD therapies that included seven studies that used CYC (n = 71 cases),[134] CYC use was least associated with improvement in patients with chronic ILD. Moreover, in a recently published randomized controlled trial comparing the use of CYC with RTX in patients with connective tissue-related ILD, there was no differences in FVC improvement at follow-up although RTX showed a better side-effect profile.[163]

Because of the associated toxicity[164] [165] [166] [167] and the availability of other options with at least comparable efficacy, we do not routinely treat our patients with CYC. When used for RP-ILD or severe exacerbations, we favor the intravenous route initially prescribed at 500 to 750 mg/m2 of body surface area (BSA) repeated every 2 to 4 weeks with 20 to 25% dose (suggested maximum dose of 1 g/m2 BSA) adjustments based on the white blood cell count response to the initial dose.


#

Rituximab

RTX is a monoclonal antibody that targets anti-CD20+ B cells leading to their depletion and subsequent immunosuppression. It is often used in combination with other immunosuppressant agents for refractory disease or during acute exacerbations.[52] [88] In a cohort of 112 patients with ASyS-ILD, 34 of whom who received RTX for a median duration of 52 months showed improvements in FVC, DLCO, and imaging by 24, 17, and 34%, respectively.[127] In an open-label phase II trial of RTX in refractory ASyS, 9 out of the 10 treated patients showed improvement or stabilization of their ILD.[168] Another retrospective study of 25 ASyS-ILD patients showed that stability or improvement can be achieved with the use of RTX up to 3 years with repeat dosing of RTX,[88] but noted a decline in DLCO in patients who received a one-time dose of RTX. A meta-analysis RTX treatment demonstrated a 76.6% (95% confidence interval [CI]: 50.4–96.0; 3 studies, n = 20) improvement rate in IIM-ILD.[134] In a larger meta-analysis with subgroup analysis for ASyS-ILD, RTX was associated with an improvement rate of 48.1% (95% CI: 0.373–0.620, 5 studies, n = 68), and a stability rate of 43.4% (95% CI: 0.310–0.607, 4 studies, n = 53).[169] Lastly, as mentioned previously, when compared with CYC, RTX performed better in pulmonary progression-free survival at 2 years.[162]

We typically prescribe RTX at 1 g on days 0 and 14 with another dose at 6 months for severe ASyS-ILD or in those with an exacerbation. Subsequent repeat dosing every 4 to 6 months is considered on a case-by-case basis. Prior to therapy we advocate for the assessment of CD19/CD20 B cell counts and serum immunoglobulin levels to help with possible complications (impaired B cell reconstitution and hypogammaglobulinemia).[170] Similarly, hepatitis status and tuberculosis exposure should be ascertained prior to treatment and treatment initiated as needed. When possible, we also ensure adequate vaccinations before starting therapy.


#

Intravenous Immunoglobulins

IVIG is derived from the pooled immunoglobulins of thousands of healthy donors. IVIG is used at higher doses (1–2 g/kg divided over 2–5 days) for autoimmune conditions and is believed to result in immunomodulation through several pathways. These include blockade of cell surface receptors, neutralization of proinflammatory cytokines, abating complement and autoantibody-directed cellular damage, and both saturation of the Fc receptors and modulation of its expression on immune cells.[171] Much of the literature supports the use of IVIG in IIM for extrapulmonary disease.[172] [173] [174] [175] Observational reports also exist, documenting the benefit of IVIG in IIM-ILD.[176] [177] [178] [179] Specifically for ASyS-ILD, a retrospective study of 17 patients treated with IVIG showed significant improvements in FVC, DLCO, and a reduction in the mean prednisone dose.[180]

We use IVIG at a dose of 2 g/kg divided over 5 days and repeated monthly. Typically, we continue IVIG monthly with another immunosuppressant agent for a duration of 6 months but can be extended further in some patients. Due to its perceived rapid onset of action, we also employ it in RP-ILD and acute exacerbations. Notable side effects of IVIG can include aseptic meningitis, hemolytic anemia, and thromboembolic disease. Vigilance is warranted to identify side effects.


#

Other Therapies

Tofacitinib, a Janus kinase inhibitor, has been beneficial in treating cutaneous manifestations in refractory DM.[181] Explorations for its use in anti-MDA-5-related ILD[182] [183] and ASyS-ILD[184] [185] have been limited and more research is needed. Abatacept, an inhibitor of anti-CD80/CD-86 which prevents T cell activation, was evaluated in a retrospective report of eight patients with ASyS-ILD. It was able to alleviate symptoms and significantly improve DLCO.[186] A recent phase 2 trial demonstrated the safety of abatacept, paving the way for a phase 3 exploration.[187]

Plasmapheresis has been explored as an appealing option for those with critical illness to allow for immunosuppression and recovery. The overwhelming majority of the data are for its use in clinically amyopathic DM and specifically anti-MDA5 disease.[188] [189] However, there are case reports[190] [191] [192] and one case series[193] showing favorable outcomes in those with ASyS-ILD. Other therapies that have been trialed for IIM-ILD include antitumor necrosis factor-α,[194] [195] tocilizumab,[196] [197] [198] and anakinra.[199] More research is required to better evaluate these options for ASyS-ILD.


#

Antifibrotics

Despite immunosuppression being the primary mode of therapy for ASS-ILD, UIP changes have been shown to be less responsive to therapy[65] and can represent up to one-quarter of patients with ASyS-ILD[102]; some of whom can develop a progressive fibrotic phenotype. Nintedanib has been approved for the use in non-IPF (idiopathic pulmonary fibrosis) patients with progressive fibrosis based on its ability to slow the rate of FVC decline in the INBUILD trial,[200] of which 25% had autoimmune ILD. In IIM-ILD the use of pirfenidone or nintedanib has been limited with mixed results.[201] [202] More research is warranted in this area.


#

PH-Specific Therapies

As mentioned previously, there are several reports linking ASyS with Group 1 PH, although a substantial proportion of patients develop Group 3 disease. In individuals that develop Group 3 PH (based on RHC), inhaled treprostinil can be used based on the INCREASE trial where it was administered to 163 individuals with ILD (25% with CTD-ILD) and demonstrated improvements in both 6-minute walk distance (6MWD) and brain natriuretic peptide levels.[203] Patients with PH and minimal ILD may be classified as Group 1 disease after ruling other possible etiologies and treated accordingly.[204] Given the complexity and challenges with managing such patients, we routinely refer them to PH specialists. Our threshold for referral is a right ventricular systolic pressure >30 mm Hg, or any signs of right ventricular dysfunction on echocardiography.


#
#
#

Our Approach to Therapy

The timing of therapy is often dictated by the disease severity, patient's age, comorbidities, and associated ASyS manifestations. In cases of mild disease, where symptoms are minimal, and there is no PFT derangement or significant pulmonary parenchymal involvement (<10%) on CT scans, it may be sufficient to observe patients with close monitoring. However, in instances where there is a more substantial symptom burden, significant radiographic involvement or progression, or a decline in PFTs (5% or 10% for FVC and DLCO, respectively), we have a low threshold for initiating a prednisone taper with a concomitant steroid-sparing agent ([Fig. 3]).

No clear evidence is available for the superiority of one agent over another. Therefore, factors such as the prescribing provider's familiarity with different agents, the patient's underlying comorbidities, drug availability, and the associated side effects affect the eventual choice. This can be highlighted in our practice of favoring RTX in patients with concurrent inflammatory arthritis, choosing an alternative to TAC in patients with renal impairment, or the judicious use of IVIG in patients at risk of thromboembolism or volume overload. Management of severe and acute exacerbation of ASyS-ILD is often challenging, requires hospitalization, and may necessitate ICU level of care. These issues are discussed separately.

While there is no clear evidence for the optimal duration of immunosuppressive therapy, we aim to limit long-term steroid exposure. We achieve this by overlapping a steroid-sparing agent with prednisone and ideally decreasing it within 3 months, to a prednisone equivalent of ≤10 mg/d. We subsequently continue the target dose steroid-sparing agent(s) until stability is achieved in symptoms, PFTs, and radiographs. We often maintain therapy for at least 18 to 24 months before consideration of a slow taper of the steroid-sparing agent accompanied with close monitoring.

We typically use PFTs to monitor disease progression and response to treatment. A comprehensive PFT (spirometry, lung volumes, and DLCO) is conducted at our initial encounter with patients and before starting therapy with repeat spirometry and DLCO assessments every 3 to 4 months, and later every 6 to 12 months once stable. Monitoring patient-centered outcomes, such as symptoms and quality of life, is crucial for tracking disease progression and therapy. Additionally, we vigilantly assess therapy-related adverse events as outlined in [Table 3]. Importantly, we do not routinely rely on imaging to monitor or assess response to treatment; instead, we employ it when clinically indicated. We incorporate guideline-directed prophylaxis for osteoporosis and pneumocystis jirovecii pneumonia (PJP), gastroesophageal reflux (GERD), and aspiration.

Table 3

Drug therapies used in the treatment of antisynthetase-associated ILD

Drug

Dose

Monitoring

Adverse effects

Comments

First line

Corticosteroids

0.5–1 mg/kg/d prednisone; methylprednisolone 500–1,000 mg/d for 3 d

Annual bone density scan, glucose monitoring

Osteoporosis, glaucoma, hyperglycemia, insomnia, weight gain, bruising, mood changes, myopathy, risk for PJP

Disease severity dictates dosing. Taper slowly when steroid-sparing agent has reached therapeutic effect.

Second line

Azathioprine

2–3 mg/kg/d (generally not higher than 200 mg/d)

TPMT level before initiation; CBC and CMP every 2 wk during dose titration and 4–8 wk thereafter; yearly skin exam

Transaminitis, leukopenia, GI intolerance, increased risk of malignancy

Start at 50 mg/d and titrate by 50 mg/wk if tolerating. May need dose adjustment based on TPMT level. Controls myositis effectively.

Mycophenolate mofetil

2,000–3,000 mg/d divided in two doses

CBC count and CMP every 2 wk during dose titration and every 4–8 wk thereafter; yearly skin examination

Transaminitis, leukopenia, GI intolerance, increased risk of malignancy, difficulty concentrating

Start at 500 mg twice a day; titrate slowly over a few weeks according to tolerability.

Controls skin disease effectively

Third line

Cyclosporin A

2–5 mg/kg/d divided in two doses

CBC, CMP, CsA trough levels every wk for first month and every 4 wk thereafter; yearly skin exam and lipid panel

Renal toxicity, hypertension, hyperlipidemia, tremors, hyperglycemia, increased risk of malignancy

Start at 2 mg/kg and titrate dose by 0.5 mg/kg Maintain serum trough level 100–200 ng/mL.

Controls myositis and joint disease effectively

Tacrolimus

Typical starting dose is 0.5–1.0 mg or 0.075 mg/kg twice a day

CBC count, CMP, TAC trough level every week for the first month and every 4 wk thereafter; yearly skin exam and lipid panel

Renal toxicity, hypertension, hyperlipidemia, tremors, hyperglycemia, increased risk of malignancy

Titrate to a trough of 3–6 ng/mL

Controls myositis and joint disease effectively

Cyclophosphamide

2 mg/kg po daily; 500–750 mg/m2 IV monthly

CBC count and CMP every 2 wk initially, UA monthly, lifelong urine cytologic analysis annually

Myelosuppression, malignancy, hemorrhagic cystitis, infertility

Maintain WBC >3,500/mm3.

Dose adjust by 25% according to WBC count of initial IV dose; not to exceed 1,000 mg/m2

Often reserved for acute exacerbation

Adjunct therapy

Rituximab

1,000 mg day 0 and day 14; repeat about every 6 months

Hepatitis and latent TB screening; immunoglobulin, CBC prior to infusions; CD19/CD20 levels before initiation and sometimes during therapy

Hepatitis reactivation, increased risk of severe SARS-CoV-2 infection, infusion reactions, progressive multifocal leukoencephalopathy

Reserved for severe or resistant disease as an adjunct therapy.

Controls myositis and joint disease effectively

Human intravenous immunoglobulin

2 g/kg/mo divided over 3–5 d

Screen for IgA deficiency before initiation

VTE, volume overload, headaches (aseptic meningitis), antibody-mediated cytopenias, anaphylaxis, infusion reactions

Initially prescribed for 6 months before slowly tapering by spacing out therapy every 5–8 wk before withdrawal.

Not considered to be immune-suppressive. Reserved for severe and resistant disease as an adjunct therapy. Can be used in acute exacerbations.

Abbreviations: CBC, complete blood count; CMP, comprehensive metabolic panel; CsA, ciclosporin A; ILD, interstitial lung disease; IgA, immunoglobulin A; IVIG, IV immunoglobulin; mo, month(s); PJP, Pneumocystis jirovecii pneumonia; TAC, tacrolimus; TB, tuberculosis; TPMT, thiopurine S-methyltransferase; UA, urinalysis; VTE, venous thromboembolism; WBC, white blood count; wk, week(s).


Severe Disease and Critically Ill Patients

Although the data for IIM-ILD (and specifically ASyS-ILD) developing respiratory failure are limited, it confers a grim prognosis, with mortality estimates from acute exacerbations upwards of 40%.[205] [206] [207] The acute respiratory failure that develops in these patients resembles acute respiratory distress syndrome (ARDS) with histological correlates of DAD.[208] [209] Despite the literature outlining a poor prognosis for patients with CTD-ILD and acute respiratory failure, we believe that ASyS-ILD presents a unique subset of patients that may have better outcomes[210] especially if invasive mechanical ventilation support is optimized.[211] This stems from our observation that many of the patients are younger, have otherwise less co-morbidities, are primarily with single organ (lung) diseases (making them candidates for other advanced interventions, e.g., lung transplantation), and the development of acute respiratory failure is likely reversible.

To this end, we actively search for an infectious etiology. We find that deep endotracheal aspirate or, in patients who can tolerate it, bronchoalveolar lavage has a better yield to pick up infectious agents in individuals on background immunosuppression.[212] [213] [214] Nevertheless, since infection has been documented as a common etiology in acute respiratory worsening,[215] we often empirically treat patients with broad-spectrum antibiotics that include azithromycin for its immunomodulating effects in ILD exacerbations[216] and ALI.[217] We do not incorporate procalcitonin levels in our antimicrobial coverage algorithm.

As acute respiratory failure may be the first manifestation of ASyS-ILD, we have a high index of suspicion in patients with new-onset ILD. In the absence of an active infection, or other causes of dyspnea, we depend on thorough history and physical examination. We focus on hand, neck, and back examinations that are often overlooked in critically ill patients for dermatological or rheumatic features of ASyS that can provide further insight into the patients' underlying disease activity. This is vital for patients presenting for the first time with ASyS-ILD in expediting their diagnosis. We also search for other markers, including creatinine kinase levels, aldolase levels, and repeat of their antibody serology that may increase above baseline during flares.

We pulse such patients with 1 g of intravenous methylprednisolone for 3 days followed by prednisone 1 mg/kg/d (max 60 mg). Although CYC has traditionally been used in severe disease and acute exacerbations for remission, we prefer using IVIG due to its lower associated toxicity. A steroid-sparing agent (AZA, MMF, or TAC) is also routinely started based on other ASyS organ involvement. We prefer using TAC based on our anecdotal experience of better efficacy and faster onset action and data for its use in refractory disease. In patients who fail to respond within the first 5 to 7 days or present with fulminant respiratory failure, we typically add a second steroid-sparing agent (CYC, RTX, and TAC, if not used previously). We rarely use plasmapheresis except in select cases. Early referral for extracorporeal membrane oxygenation (ECMO) and consideration of lung transplant evaluation where available are also critical.

Respiratory support is often needed in these patients. We ensure that the patient and their family are aware of the reserved prognosis of invasive mechanical ventilation use during respiratory failure[218] and explore their goals of care; we also engage the palliative care team early in the patient's disease course. We incorporate noninvasive mechanical ventilation use due to its potential to decrease intubation rates, extrapolated from cohorts of respiratory failure without ILD.[219] We prefer high-flow nasal cannula due to less concern with risk of large tidal volume ventilation.[220] [221] When intubated, patients are maintained with lung-protective ventilation in line with the ARDS literature.[222] Moreover, patients are kept net-neutral or net-negative fluid balance.[211] [223] Eligibility or pre-existing enrollment for lung transplantation is explored and may help in choosing the appropriate care pathways.

ECMO has become an attractive temporizing therapy. Several cases have demonstrated its successful use in ASyS-ILD.[224] [225] [226] [227] [228] [229] [230] [231] We consider this modality in patients that fulfil our center's selection criteria, especially for those awaiting lung transplantation. Additionally, we believe that the lung healing benefits of ECMO[232] [233] complement immunosuppression therapy and provides sufficient time for it to take effect.


#
#

Nonpharmacological Treatment

Supplemental Oxygen

Supplemental oxygen therapy can help patients with ASyS-ILD in symptomatic relief, improved exercise tolerance, and theoretical risk reduction of developing PH. Hypoxemia in addition to disruption of the pulmonary vascular beds by fibrosis contributes to the development PH in patients with ILD.[234] The physiological hypoxic pulmonary vasoconstriction can be somewhat mitigated by supplemental oxygen.[235] [236] Studies on supplemental oxygen effect on dyspnea are conflicting. A small retrospective study in ILD (not limited to CTD-ILD) found that dyspnea improved with supplemental oxygen,[237] while a larger retrospective review of 3,126 patients demonstrated worsened dyspnea on exertion.[238] In a systematic review of 1,509 ILD patients, no improvement in dyspnea was observed although exercise capacity increased with oxygen.[239] A more robust prospective, single-blinded, randomized study found that dyspnea improves with supplemental oxygen at baseline but not during peak exercise, despite a significant improvement in exercise endurance.[240] We routinely screen all our ASyS-ILD patients who complain of significant dyspnea, reduced exercise tolerance, or those with DLCO is below 50% predicted. We prescribe oxygen if their saturation falls below 88%, either at rest or on exertion.[241] [242] Nocturnal oxygen is encouraged for all patients with resting hypoxemia.


#

Pulmonary Rehabilitation and Exercise

Poor exercise capacity is linked with worse survival in patients with ILD.[243] In a Cochrane review of pulmonary rehabilitation,[244] improvements in dyspnea and 6MWD were observed and maintained at 6 to 12 months. Muscle strength also improves after 6 months of pulmonary rehabilitation with improved exercise capacity and quality of life. We therefore offer pulmonary rehabilitation to all our ASyS-ILD patients[245] and emphasize regular exercise.


#

Immunization and Prophylaxis

We routinely offer patients the pneumococcal, COVID-19, influenza, and respiratory syncytial virus (in those who qualify) vaccines irrespective of their immunosuppression regimen. Alongside vitamin D supplementation, we prescribe oral bisphosphonates for those we anticipate requiring continued prednisone of ≥2.5 mg/day for >3 months if they have a moderate risk of fracture.[246] PJP occurs more frequently in IIM-ILD patients,[247] [248] and is associated with a high mortality rate.[249] We typically prescribe prophylaxis in patients receiving prednisone at a dose of ≥15 mg/d for ≥4 weeks, RTX, or any combination of two or more steroid-sparing agents. CD4 levels <200/mm3 may also aid in the decision of initiating prophylaxis.

GERD is common in ASyS-ILD especially when esophageal dysmotility is present.[250] We emphasize lifestyle and dietary measures, weight loss, elevation of the head of bed, and avoiding dietary triggers[251] to limit reflux or micro aspirations. We offer proton pump inhibitors therapy when indicated.


#

Lung Transplantation

In patients with end-stage ASyS-ILD, lung transplantation may be their only viable option. In retrospective studies, the survival of transplanted patients with IIM appeared to be slightly worse in some,[252] while similar in others[253] [254] when compared with CTD-ILD. In line with international recommendations,[255] we refer our patients for lung transplant evaluation if they are on supplemental oxygen, DLCO <40%, progression of disease despite treatment, rapid decline in PFTs, or any evidence of PH.


#

ILD Prognosis

Patients with ASyS-ILD have variable disease outcome. Generally, most patients respond to therapy with either improvement or stabilization in their lung functions. A subset may continue to deteriorate, are refractory to therapy, or recur. In a cohort of 51 patients with anti-EJ antibody-positive ILD,[82] of whom 44 were followed to assess response to immunosuppression, 34 (77%) improved, and 10 (23%) stabilized. Disease recurrence was seen in 25%. In another cohort of 87 ASA-ILD with positive anti-Ro-52,[256] 56 (64%), 10 (11.5%), and 21 (24.5%) patients improved, stabilized, and worsened, respectively. The higher rate of nonresponsive disease in this group may be due to patient selection that was initially based on positive anti-Ro-52 antibodies prior to testing for ASA; possibly causing preselection of patients with a worse phenotype in addition to the lack of anti-Jo-1 positive patients. In 202 patients with ASyS,[257] patients with non-Jo-1 serology had a worse 5- and 10-year cumulative survival, with the main cause of death in all the population being pulmonary fibrosis and PH.

ASyS-ILD typically shows response to therapy in the first 6 to 12 months,[65] [80] [258] [259] unless refractory. Factors that have been demonstrated to show worse outcomes in patients with ASyS-ILD include the presence of a UIP pattern on HRCT,[50] [65] [89] [95] [260] non-Jo-1 serology,[50] [55] [77] [257] [260] [261] male gender,[76] and older age at diagnosis.[17] [53] Mortality in patients with ASyS is considered higher than the average[17] [53] [55] often due to pulmonary complications.[261] [262]


#
#

Conclusion

ASyS has a high pulmonary disease burden, with ILD being its most prominent and common manifestation. Although various classification criteria exist, a high index of suspicion is crucial when assessing patients with ILD. Different ASAs are associated with distinct clinical features, often presenting with radiographic disease consistent with NSIP or OP. In most cases, lung biopsy is unnecessary. Various therapeutic options are available demonstrating relative success in disease control. However, a subset of patients experiencing clinical deterioration, unresponsiveness, or becoming critically ill emphasizes the need for future prospective studies focused specifically on ASyS-ILD management.


#
#

Conflict of Interest

None declared.

  • References

  • 1 Lundberg IE, Tjärnlund A, Bottai M. et al. 2017 European League Against Rheumatism/American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups. Ann Rheum Dis 2017; 76 (12) 1955-1964
  • 2 Hochberg MC, Feldman D, Stevens MB, Arnett FC, Reichlin M. Antibody to Jo-1 in polymyositis/dermatomyositis: association with interstitial pulmonary disease. J Rheumatol 1984; 11 (05) 663-665
  • 3 Marguerie C, Bunn CC, Beynon HLC. et al. Polymyositis, pulmonary fibrosis and autoantibodies to aminoacyl-tRNA synthetase enzymes. Q J Med 1990; 77 (282) 1019-1038
  • 4 Connors GR, Christopher-Stine L, Oddis CV, Danoff SK. Interstitial lung disease associated with the idiopathic inflammatory myopathies: what progress has been made in the past 35 years?. Chest 2010; 138 (06) 1464-1474
  • 5 Solomon J, Swigris JJ, Brown KK. Myositis-related interstitial lung disease and antisynthetase syndrome. J Bras Pneumol 2011; 37 (01) 100-109
  • 6 Lega JC, Reynaud Q, Belot A, Fabien N, Durieu I, Cottin V. Idiopathic inflammatory myopathies and the lung. Eur Respir Rev 2015; 24 (136) 216-238
  • 7 Khoo T, Lilleker JB, Thong BYH, Leclair V, Lamb JA, Chinoy H. Epidemiology of the idiopathic inflammatory myopathies. Nat Rev Rheumatol 2023; 19 (11) 695-712
  • 8 Galindo-Feria AS, Notarnicola A, Lundberg IE, Horuluoglu B. Aminoacyl-tRNA synthetases: on anti-synthetase syndrome and beyond. Front Immunol 2022; 13: 866087
  • 9 Wells M, Alawi S, Thin KYM. et al. A multidisciplinary approach to the diagnosis of antisynthetase syndrome. Front Med (Lausanne) 2022; 9: 959653
  • 10 Tan JA, Roberts-Thomson PJ, Blumbergs P, Hakendorf P, Cox SR, Limaye V. Incidence and prevalence of idiopathic inflammatory myopathies in South Australia: a 30-year epidemiologic study of histology-proven cases. Int J Rheum Dis 2013; 16 (03) 331-338
  • 11 Limaye V, Hakendorf P, Woodman RJ, Blumbergs P, Roberts-Thomson P. Mortality and its predominant causes in a large cohort of patients with biopsy-determined inflammatory myositis. Intern Med J 2012; 42 (02) 191-198
  • 12 Limaye V, Luke C, Tucker G. et al. The incidence and associations of malignancy in a large cohort of patients with biopsy-determined idiopathic inflammatory myositis. Rheumatol Int 2013; 33 (04) 965-971
  • 13 Barsotti S, Lundberg IE. Myositis an evolving spectrum of disease. Immunol Med 2018; 41 (02) 46-54
  • 14 Hum RM, Lilleker JB, Lamb JA. et al. MYONET registry. Comparison of clinical features between patients with anti-synthetase syndrome and dermatomyositis: results from the MYONET registry. Rheumatology (Oxford) 2023; 00: 1-8
  • 15 Zhang Y, Ge Y, Yang H. et al. Clinical features and outcomes of the patients with anti-glycyl tRNA synthetase syndrome. Clin Rheumatol 2020; 39 (08) 2417-2424
  • 16 Koenig M, Fritzler MJ, Targoff IN, Troyanov Y, Senécal JL. Heterogeneity of autoantibodies in 100 patients with autoimmune myositis: insights into clinical features and outcomes. Arthritis Res Ther 2007; 9 (04) R78
  • 17 Shi J, Li S, Yang H. et al. Clinical profiles and prognosis of patients with distinct antisynthetase autoantibodies. J Rheumatol 2017; 44 (07) 1051-1057
  • 18 Pinal-Fernandez I, Casal-Dominguez M, Huapaya JA. et al. A longitudinal cohort study of the anti-synthetase syndrome: increased severity of interstitial lung disease in black patients and patients with anti-PL7 and anti-PL12 autoantibodies. Rheumatology (Oxford) 2017; 56 (06) 999-1007
  • 19 Noguchi E, Uruha A, Suzuki S. et al. Skeletal muscle involvement in antisynthetase syndrome. JAMA Neurol 2017; 74 (08) 992-999
  • 20 Orphanet: Antisynthetase syndrome. Accessed December 17, 2023 at: https://www.orpha.net/consor/cgi-bin/OC_Exp.php?Lng=GB&Expert=81
  • 21 Lilleker JB, Vencovsky J, Wang G. et al. all EuroMyositis contributors. The EuroMyositis registry: an international collaborative tool to facilitate myositis research. Ann Rheum Dis 2018; 77 (01) 30-39
  • 22 Cavagna L, Trallero-Araguás E, Meloni F. et al. Influence of Antisynthetase antibodies specificities on antisynthetase syndrome clinical spectrum time course. J Clin Med 2019; 8 (11) 2013
  • 23 Wang R, Zhao Y, Qi F. et al. Analysis of the clinical features of antisynthetase syndrome: a retrospective cohort study in China. Clin Rheumatol 2023; 42 (03) 703-709
  • 24 Deligny C, Dueymes M, Arfi S. et al. Epidemiology and characteristics of antisynthetase syndrome in the African descent population of martinique. Arthritis Rheumatol 2014; 66: S556
  • 25 Hamaguchi Y, Fujimoto M, Matsushita T. et al. Common and distinct clinical features in adult patients with anti-aminoacyl-tRNA synthetase antibodies: heterogeneity within the syndrome. PLoS One 2013; 8 (04) e60442
  • 26 Li W, Li J, Xie WM, Ren YH, Dai HP. Clinical characteristics of patients with antisynthetase syndrome and interstitial pulmonary disease [in Chinese]. Zhonghua Yi Xue Za Zhi 2020; 100 (24) 1861-1865
  • 27 Dugar M, Cox S, Limaye V, Blumbergs P, Roberts-Thomson PJ. Clinical heterogeneity and prognostic features of South Australian patients with anti-synthetase autoantibodies. Intern Med J 2011; 41 (09) 674-679
  • 28 Mescam-Mancini L, Allenbach Y, Hervier B. et al. Anti-Jo-1 antibody-positive patients show a characteristic necrotizing perifascicular myositis. Brain 2015; 138 (Pt 9): 2485-2492
  • 29 Uruha A, Nishikawa A, Tsuburaya RS. et al. Sarcoplasmic MxA expression: a valuable marker of dermatomyositis. Neurology 2017; 88 (05) 493-500
  • 30 Selva-O'Callaghan A, Pinal-Fernandez I, Trallero-Araguás E, Milisenda JC, Grau-Junyent JM, Mammen AL. Classification and management of adult inflammatory myopathies. Lancet Neurol 2018; 17 (09) 816-828
  • 31 Gutierrez-Arcelus M, Rich SS, Raychaudhuri S. Autoimmune diseases - connecting risk alleles with molecular traits of the immune system. Nat Rev Genet 2016; 17 (03) 160-174
  • 32 Rubtsova K, Marrack P, Rubtsov AV. Sexual dimorphism in autoimmunity. J Clin Invest 2015; 125 (06) 2187-2193
  • 33 Haghikia A, Jörg S, Duscha A. et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity 2015; 43 (04) 817-829
  • 34 Gallay L, Gayed C, Hervier B. Antisynthetase syndrome pathogenesis: knowledge and uncertainties. Curr Opin Rheumatol 2018; 30 (06) 664-673
  • 35 Svensson J, Holmqvist M, Lundberg IE, Arkema EV. Infections and respiratory tract disease as risk factors for idiopathic inflammatory myopathies: a population-based case-control study. Ann Rheum Dis 2017; 76 (11) 1803-1808
  • 36 Webber MP, Moir W, Zeig-Owens R. et al. Nested case-control study of selected systemic autoimmune diseases in World Trade Center rescue/recovery workers. Arthritis Rheumatol 2015; 67 (05) 1369-1376
  • 37 Katsumata Y, Ridgway WM, Oriss T. et al. Species-specific immune responses generated by histidyl-tRNA synthetase immunization are associated with muscle and lung inflammation. J Autoimmun 2007; 29 (2-3): 174-186
  • 38 Albrecht I, Eddie J, Fathi M. et al. Characterization of histidyl-tRNA synthetase specific Th cell response in blood and broncheoalveolar lavage (BAL) of myositis patients. ACR Meeting Abstracts. Published online 2013. Accessed November 18, 2023 at: https://acrabstracts.org/abstract/characterization-of-histidyl-trna-synthetase-specific-th-cell-response-in-blood-and-broncheoalveolar-lavage-bal-of-myositis-patients/
  • 39 Englund P, Wahlström J, Fathi M. et al. Restricted T cell receptor BV gene usage in the lungs and muscles of patients with idiopathic inflammatory myopathies. Arthritis Rheum 2007; 56 (01) 372-383
  • 40 Galindo-Feria AS, Albrecht I, Notarnicola A. et al. 02.09 Identification of a novel pro-inflammatory T cell epitope from his-trna-synthetase associated with interstitial lung disease in anti-jo-1 positive patients. Ann Rheum Dis 2017; 76: A11.2-A12
  • 41 Barbasso Helmers S, Englund P, Engström M. et al. Sera from anti-Jo-1-positive patients with polymyositis and interstitial lung disease induce expression of intercellular adhesion molecule 1 in human lung endothelial cells. Arthritis Rheum 2009; 60 (08) 2524-2530
  • 42 Guo M, Schimmel P. Essential nontranslational functions of tRNA synthetases. Nat Chem Biol 2013; 9 (03) 145-153
  • 43 Lo WS, Gardiner E, Xu Z. et al. Human tRNA synthetase catalytic nulls with diverse functions. Science 2014; 345 (6194) 328-332
  • 44 Adams RA, Fernandes-Cerqueira C, Notarnicola A. et al. Serum-circulating His-tRNA synthetase inhibits organ-targeted immune responses. Cell Mol Immunol 2021; 18 (06) 1463-1475
  • 45 Miller FW, Chen W, O'Hanlon TP. et al. Myositis Genetics Consortium. Genome-wide association study identifies HLA 8.1 ancestral haplotype alleles as major genetic risk factors for myositis phenotypes. Genes Immun 2015; 16 (07) 470-480
  • 46 Rothwell S, Lamb JA, Chinoy H. New developments in genetics of myositis. Curr Opin Rheumatol 2016; 28 (06) 651-656
  • 47 Rothwell S, Chinoy H, Lamb JA. et al. Myositis Genetics Consortium (MYOGEN). Focused HLA analysis in Caucasians with myositis identifies significant associations with autoantibody subgroups. Ann Rheum Dis 2019; 78 (07) 996-1002
  • 48 Remuzgo-Martínez S, Atienza-Mateo B, Ocejo-Vinyals JG. et al. HLA association with the susceptibility to anti-synthetase syndrome. Joint Bone Spine 2021; 88 (03) 105115
  • 49 Opinc AH, Makowska JS. Antisynthetase syndrome - much more than just a myopathy. Semin Arthritis Rheum 2021; 51 (01) 72-83
  • 50 Jiang M, Dong X, Zheng Y. Clinical characteristics of interstitial lung diseases positive to different anti-synthetase antibodies. Medicine (Baltimore) 2021; 100 (19) e25816
  • 51 Vulsteke JB, Satoh M, Malyavantham K, Bossuyt X, De Langhe E, Mahler M. Anti-OJ autoantibodies: rare or underdetected?. Autoimmun Rev 2019; 18 (07) 658-664
  • 52 Marie I, Hatron PY, Dominique S. et al. Short-term and long-term outcome of anti-Jo1-positive patients with anti-Ro52 antibody. Semin Arthritis Rheum 2012; 41 (06) 890-899
  • 53 Trallero-Araguás E, Grau-Junyent JM, Labirua-Iturburu A. et al. IIM Study Group and Autoimmune Diseases Study Group (GEAS) of the Spanish Society of Internal Medicine (SEMI). Clinical manifestations and long-term outcome of anti-Jo1 antisynthetase patients in a large cohort of Spanish patients from the GEAS-IIM group. Semin Arthritis Rheum 2016; 46 (02) 225-231
  • 54 Cavagna L, Nuño L, Scirè CA. et al. AENEAS (American, European NEtwork of Antisynthetase Syndrome) collaborative group.. Clinical spectrum time course in anti jo-1 positive antisynthetase syndrome: results from an international retrospective multicenter study. Medicine (Baltimore) 2015; 94 (32) e1144
  • 55 Hervier B, Devilliers H, Stanciu R. et al. Hierarchical cluster and survival analyses of antisynthetase syndrome: phenotype and outcome are correlated with anti-tRNA synthetase antibody specificity. Autoimmun Rev 2012; 12 (02) 210-217
  • 56 Tanboon J, Inoue M, Hirakawa S. et al. Muscle pathology of antisynthetase syndrome according to antibody subtypes. Brain Pathol 2023; 33 (04) e13155
  • 57 Szabó K, Bodoki L, Nagy-Vincze M. et al. Effect of genetic and laboratory findings on clinical course of antisynthetase syndrome in a Hungarian cohort. BioMed Res Int 2018; 2018: 6416378
  • 58 Kaneko Y, Hanaoka H, Hirakata M, Takeuchi T, Kuwana M. Distinct arthropathies of the hands in patients with anti-aminoacyl tRNA synthetase antibodies: usefulness of autoantibody profiles in classifying patients. Rheumatology (Oxford) 2014; 53 (06) 1120-1124
  • 59 Meyer A, Lefevre G, Bierry G. et al. Club Rhumatismes et Inflammation. In antisynthetase syndrome, ACPA are associated with severe and erosive arthritis: an overlapping rheumatoid arthritis and antisynthetase syndrome. Medicine (Baltimore) 2015; 94 (20) e523
  • 60 Kumar RR, Jha S, Dhooria A. et al. Anti-Jo-1 syndrome often misdiagnosed as rheumatoid arthritis (for many years): a single-center experience. J Clin Rheumatol 2021; 27 (04) 150-155
  • 61 Miller JB, Danoff SK, Bingham III CO. et al. Sonographic findings from inflammatory arthritis due to antisynthetase syndrome. Clin Rheumatol 2019; 38 (05) 1477-1483
  • 62 Gusdorf L, Morruzzi C, Goetz J, Lipsker D, Sibilia J, Cribier B. Mechanics hands in patients with antisynthetase syndrome: 25 cases. Ann Dermatol Venereol 2019; 146 (01) 19-25
  • 63 Cox JT, Gullotti DM, Mecoli CA. et al. “Hiker's feet”: a novel cutaneous finding in the inflammatory myopathies. Clin Rheumatol 2017; 36 (07) 1683-1686
  • 64 Hosono Y, Ishii A, Izumi Y. et al. New aspects of clinical and immunological characteristics in patients with anti-asparaginyl tRNA synthetase (anti-KS) autoantibody. Mod Rheumatol 2023; 34 (01) 122-128
  • 65 Marie I, Josse S, Hatron PY. et al. Interstitial lung disease in anti-Jo-1 patients with antisynthetase syndrome. Arthritis Care Res (Hoboken) 2013; 65 (05) 800-808
  • 66 Richards TJ, Eggebeen A, Gibson K. et al. Characterization and peripheral blood biomarker assessment of anti-Jo-1 antibody-positive interstitial lung disease. Arthritis Rheum 2009; 60 (07) 2183-2192
  • 67 Marie I, Josse S, Decaux O. et al. Comparison of long-term outcome between anti-Jo1- and anti-PL7/PL12 positive patients with antisynthetase syndrome. Autoimmun Rev 2012; 11 (10) 739-745
  • 68 Johnson C, Connors GR, Oaks J. et al. Clinical and pathologic differences in interstitial lung disease based on antisynthetase antibody type. Respir Med 2014; 108 (10) 1542-1548
  • 69 Tillie-Leblond I, Wislez M, Valeyre D. et al. Interstitial lung disease and anti-Jo-1 antibodies: difference between acute and gradual onset. Thorax 2008; 63 (01) 53-59
  • 70 Cruellas MG, Viana VdosS, Levy-Neto M, Souza FH, Shinjo SK. Myositis-specific and myositis-associated autoantibody profiles and their clinical associations in a large series of patients with polymyositis and dermatomyositis. Clinics (São Paulo) 2013; 68 (07) 909-914
  • 71 Bauhammer J, Blank N, Max R. et al. Rituximab in the treatment of Jo1 antibody-associated antisynthetase syndrome: anti-Ro52 positivity as a marker for severity and treatment response. J Rheumatol 2016; 43 (08) 1566-1574
  • 72 Zhao Y, Zhang C, Su H. et al. Predictive factors for progressive fibrosing interstitial lung disease in anti-synthetase syndrome. Int J Rheum Dis 2023; 26 (05) 885-894
  • 73 Váncsa A, Csípo I, Németh J, Dévényi K, Gergely L, Dankó K. Characteristics of interstitial lung disease in SS-A positive/Jo-1 positive inflammatory myopathy patients. Rheumatol Int 2009; 29 (09) 989-994
  • 74 La Corte R, Lo Mo Naco A, Locaputo A, Dolzani F, Trotta F. In patients with antisynthetase syndrome the occurrence of anti-Ro/SSA antibodies causes a more severe interstitial lung disease. Autoimmunity 2006; 39 (03) 249-253
  • 75 Pepper E, Vilar L, Ward IM. Clinical characteristics and prognostic value of Ro52/SSA antibodies in idiopathic inflammatory myopathies. J Clin Rheumatol 2023; 29 (07) 347-353
  • 76 Zamora AC, Hoskote SS, Abascal-Bolado B. et al. Clinical features and outcomes of interstitial lung disease in anti-Jo-1 positive antisynthetase syndrome. Respir Med 2016; 118: 39-45
  • 77 Zhan X, Yan W, Wang Y. et al. Clinical features of anti-synthetase syndrome associated interstitial lung disease: a retrospective cohort in China. BMC Pulm Med 2021; 21 (01) 57
  • 78 American Thoracic Society/European Respiratory Society. ATS/ERS Statement on respiratory muscle testing. Am J Respir Crit Care Med 2002; 166 (04) 518-624
  • 79 Laveneziana P, Albuquerque A, Aliverti A. et al. ERS statement on respiratory muscle testing at rest and during exercise. Eur Respir J 2019; 53 (06) 1801214
  • 80 Debray MP, Borie R, Revel MP. et al. Interstitial lung disease in anti-synthetase syndrome: initial and follow-up CT findings. Eur J Radiol 2015; 84 (03) 516-523
  • 81 Waseda Y, Johkoh T, Egashira R. et al. Antisynthetase syndrome: pulmonary computed tomography findings of adult patients with antibodies to aminoacyl-tRNA synthetases. Eur J Radiol 2016; 85 (08) 1421-1426
  • 82 Liu Y, Liu X, Xie M. et al. Clinical characteristics of patients with anti-EJ antisynthetase syndrome associated interstitial lung disease and literature review. Respir Med 2020; 165: 105920
  • 83 Chung JH, Cox CW, Montner SM. et al. CT features of the usual interstitial pneumonia pattern: differentiating connective tissue disease-associated interstitial lung disease from idiopathic pulmonary fibrosis. AJR Am J Roentgenol 2018; 210 (02) 307-313
  • 84 Barratt SL, Adamali HH, Cotton C. et al. Clinicoserological features of antisynthetase syndrome (ASyS)-associated interstitial lung disease presenting to respiratory services: comparison with idiopathic pulmonary fibrosis and ASyS diagnosed in rheumatology services. BMJ Open Respir Res 2021; 8 (01) e000829
  • 85 Baratella E, Marrocchio C, Cifaldi R. et al. Interstitial lung disease in patients with antisynthetase syndrome: a retrospective case series study. Jpn J Radiol 2021; 39 (01) 40-46
  • 86 Chartrand S, Swigris JJ, Peykova L, Chung J, Fischer A. A multidisciplinary evaluation helps identify the antisynthetase syndrome in patients presenting as idiopathic interstitial pneumonia. J Rheumatol 2016; 43 (05) 887-892
  • 87 Yura H, Sakamoto N, Satoh M. et al. Clinical characteristics of patients with anti-aminoacyl-tRNA synthetase antibody positive idiopathic interstitial pneumonia. Respir Med 2017; 132: 189-194
  • 88 Doyle TJ, Dhillon N, Madan R. et al. Rituximab in the treatment of interstitial lung disease associated with antisynthetase syndrome: a multi-center retrospective case review. J Rheumatol 2018; 45 (06) 841-850
  • 89 Cobo-Ibáñez T, López-Longo FJ, Joven B. et al. Long-term pulmonary outcomes and mortality in idiopathic inflammatory myopathies associated with interstitial lung disease. Clin Rheumatol 2019; 38 (03) 803-815
  • 90 Fukamatsu H, Hirai Y, Miyake T. et al. Clinical manifestations of skin, lung and muscle diseases in dermatomyositis positive for anti-aminoacyl tRNA synthetase antibodies. J Dermatol 2019; 46 (10) 886-897
  • 91 Teel A, Lu J, Park J, Singh N, Basharat P. The role of myositis-specific autoantibodies and the management of interstitial lung disease in idiopathic inflammatory myopathies: a systematic review. Semin Arthritis Rheum 2022; 57: 152088
  • 92 Schneider F, Yousem SA, Oddis CV, Aggarwal R. Pulmonary pathologic manifestations of anti-alanyl-tRNA synthetase (anti-PL-12)-related inflammatory myopathy. Arch Pathol Lab Med 2018; 142 (02) 191-197
  • 93 Yamasaki Y, Yamada H, Yamasaki M. et al. Intravenous cyclophosphamide therapy for progressive interstitial pneumonia in patients with polymyositis/dermatomyositis. Rheumatology (Oxford) 2007; 46 (01) 124-130
  • 94 Yousem SA, Schneider F, Bi D, Oddis CV, Gibson K, Aggarwal R. The pulmonary histopathologic manifestations of the anti-PL7/antithreonyl transfer RNA synthetase syndrome. Hum Pathol 2014; 45 (06) 1199-1204
  • 95 Marie I, Josse S, Decaux O. et al. Clinical manifestations and outcome of anti-PL7 positive patients with antisynthetase syndrome. Eur J Intern Med 2013; 24 (05) 474-479
  • 96 Sasano H, Hagiwara E, Kitamura H. et al. Long-term clinical course of anti-glycyl tRNA synthetase (anti-EJ) antibody-related interstitial lung disease pathologically proven by surgical lung biopsy. BMC Pulm Med 2016; 16 (01) 168
  • 97 Watanabe K, Handa T, Tanizawa K. et al. Detection of antisynthetase syndrome in patients with idiopathic interstitial pneumonias. Respir Med 2011; 105 (08) 1238-1247
  • 98 Schneider F, Yousem SA, Bi D, Gibson KF, Oddis CV, Aggarwal R. Pulmonary pathologic manifestations of anti-glycyl-tRNA synthetase (anti-EJ)-related inflammatory myopathy. J Clin Pathol 2014; 67 (08) 678-683
  • 99 Aiko N, Yamakawa H, Iwasawa T. et al. Clinical, radiological, and pathological features of anti-asparaginyl tRNA synthetase antibody-related interstitial lung disease. Respir Investig 2020; 58 (03) 196-203
  • 100 Schneider F, Aggarwal R, Bi D, Gibson K, Oddis C, Yousem SA. The pulmonary histopathology of anti-KS transfer RNA synthetase syndrome. Arch Pathol Lab Med 2015; 139 (01) 122-125
  • 101 Sato S, Kuwana M, Hirakata M. Clinical characteristics of Japanese patients with anti-OJ (anti-isoleucyl-tRNA synthetase) autoantibodies. Rheumatology (Oxford) 2007; 46 (05) 842-845
  • 102 Flashner BM, VanderLaan PA, Nurhussien L, Rice MB, Hallowell RW. Pulmonary histopathology of interstitial lung disease associated with antisynthetase antibodies. Respir Med 2022; 191: 106697
  • 103 Condon DF, Nickel NP, Anderson R, Mirza S, de Jesus Perez VA. The 6th World Symposium on Pulmonary Hypertension: what's old is new. F1000Res 2019; 8: F1000 Faculty Rev-888
  • 104 Hervier B, Meyer A, Dieval C. et al. Pulmonary hypertension in antisynthetase syndrome: prevalence, aetiology and survival. Eur Respir J 2013; 42 (05) 1271-1282
  • 105 Lecomte R, Perrin F, Journeau L. et al. Syndrome des antisynthétases compliqué d'une hypertension pulmonaire : 4 observations originales. Rev Med Interne 2015; 36 (12) 794-799
  • 106 Hervier B, Wallaert B, Hachulla E. et al. Clinical manifestations of anti-synthetase syndrome positive for anti-alanyl-tRNA synthetase (anti-PL12) antibodies: a retrospective study of 17 cases. Rheumatology (Oxford) 2010; 49 (05) 972-976
  • 107 Kalluri M, Sahn SA, Oddis CV. et al. Clinical profile of anti-PL-12 autoantibody. Cohort study and review of the literature. Chest 2009; 135 (06) 1550-1556
  • 108 Bryan JL, Matar R, Raviprasad A. et al. Echocardiographic characteristics of patients with antisynthetase syndrome. Pulm Circ 2022; 12 (02) e12084
  • 109 Syed N, Yadav R, O'Rourke C, Chatterjee S. Pulmonary hypertension in anti-JO1 syndrome. Arthritis Rheum 2013; 65: S886
  • 110 Reddy R, Bryan JL, Raviprasad A. et al. Pulmonary hypertension in anti-synthetase syndrome. Am J Respir Crit Care Med 2021; 203 (09) A3581
  • 111 Mason T, Samaranayake C, Mccabe C. et al. Use of pulmonary vasodilator therapy in anti-synthetase syndrome-associated pulmonary hypertension. Eur Respir J 2023; 62 (Suppl. 67) PA3495
  • 112 Saito G, Kono M, Tsutsumi A. et al. Anti-PL-7 antisynthetase syndrome with eosinophilic pleural effusion. Intern Med 2018; 57 (15) 2227-2232
  • 113 Mogulkoc N, Kabasakal Y, Ekren PK, Bishop PW. An unusual presentation of anti-Jo-1 syndrome, mimicking lung metastases, with massive pleural and pericardial effusions. J Clin Rheumatol 2006; 12 (02) 90-92
  • 114 Wu Y, Chhaya S, Hurowitz B, Ardiles T, Carlson R. Clinically amyopathic dermatomyositis complicated by pleural effusion case report, literature review, and proposed mechanism. Bull Hosp Jt Dis 2015; 73 (03) 217-220
  • 115 Sugie K, Tonomura Y, Ueno S. Characterization of dermatomyositis with coexistence of anti-Jo-1 and anti-SRP antibodies. Intern Med 2012; 51 (07) 799-802
  • 116 Chen L, Somers A, Liu J. Antisynthetase syndrome and pleural effusion: a case report. Chest 2020; 158 (04) A1991-A1992
  • 117 Bhandari BS, Yeramareddy S, Akella S, Rawal H, Martin KB. An unusual case of anti-synthetase syndrome with pleurisy and pleural effusions. Am J Respir Crit Care Med 2019; 199 (09) A3116
  • 118 Katz A, Bena J, Chatterjee S. Antisynthetase syndrome: prevalence of serositis in autoantibody subsets. Arthritis Rheumatol 2018; 70: 397-399
  • 119 Lega JC, Fabien N, Reynaud Q. et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun Rev 2014; 13 (09) 883-891
  • 120 Jiang L, Jones J, Yang XL. Human diseases linked to cytoplasmic aminoacyl-tRNA synthetases. Enzymes 2020; 48: 277-319
  • 121 Vulsteke JB, Derua R, Dubucquoi S. et al. Mass spectrometry-based identification of new anti-Ly and known antisynthetase autoantibodies. Ann Rheum Dis 2023; 82 (04) 546-555
  • 122 Sasai T, Nakashima R, Shirakashi M. et al. A new autoantibody to valyl transfer RNA synthetase associated with anti-synthetase syndrome. Rheumatology (Oxford) 2023; 62 (05) e155-e157
  • 123 Sidorik LL. Seryl-tRNA synthetase &ndash; a novel autoantigen during systemic autoimmune diseases. Biopolymers Cell 1994; 10 (3–4): 68-74
  • 124 Vartanyan OA. Detection of antibodies against tryptophanyl-, tyrosyl- and phenylalanyl-tRNA synthetase and antiidiotypic antibodies aganist them in sera of patients bearing autoimmune diseases. Molekulyarnaya Biologiya 1991; 25 (04) 1033-1039
  • 125 Paley EL, Alexandrova N, Smelansky L. Tryptophanyl-tRNA synthetase as a human autoantigen. Immunol Lett 1995; 48 (03) 201-207
  • 126 Mathews MB, Bernstein RM. Myositis autoantibody inhibits histidyl-tRNA synthetase: a model for autoimmunity. Nature 1983; 304 (5922) 177-179
  • 127 Andersson H, Sem M, Lund MB. et al. Long-term experience with rituximab in anti-synthetase syndrome-related interstitial lung disease. Rheumatology (Oxford) 2015; 54 (08) 1420-1428
  • 128 Bernstein RM, Morgan SH, Chapman J. et al. Anti-Jo-1 antibody: a marker for myositis with interstitial lung disease. Br Med J (Clin Res Ed) 1984; 289 (6438) 151-152
  • 129 Stone KB, Oddis CV, Fertig N. et al. Anti-Jo-1 antibody levels correlate with disease activity in idiopathic inflammatory myopathy. Arthritis Rheum 2007; 56 (09) 3125-3131
  • 130 Zanframundo G, Faghihi-Kashani S, Scirè CA. et al. Defining anti-synthetase syndrome: a systematic literature review. Clin Exp Rheumatol 2022; 40 (02) 309-319
  • 131 Mecoli CA, Albayda J, Tiniakou E. et al. Myositis Autoantibodies: a comparison of results from the Oklahoma Medical Research Foundation Myositis panel to the euroimmun research line blot. Arthritis Rheumatol 2020; 72 (01) 192-194
  • 132 Zhang W, Zheng Y, Wang Y. et al. Thigh MRI in antisynthetase syndrome, and comparisons with dermatomyositis and immune-mediated necrotizing myopathy. Rheumatology (Oxford) 2022; 62 (01) 310-320
  • 133 Andersson H, Kirkhus E, Garen T, Walle-Hansen R, Merckoll E, Molberg Ø. Comparative analyses of muscle MRI and muscular function in anti-synthetase syndrome patients and matched controls: a cross-sectional study. Arthritis Res Ther 2017; 19 (01) 17
  • 134 Barba T, Fort R, Cottin V. et al. Treatment of idiopathic inflammatory myositis associated interstitial lung disease: a systematic review and meta-analysis. Autoimmun Rev 2019; 18 (02) 113-122
  • 135 Schwarz MI, Matthay RA, Sahn SA, Stanford RE, Marmorstein BL, Scheinhorn DJ. Interstitial lung disease in polymyositis and dermatomyositis: analysis of six cases and review of the literature. Medicine (Baltimore) 1976; 55 (01) 89-104
  • 136 Marie I, Hachulla E, Chérin P. et al. Interstitial lung disease in polymyositis and dermatomyositis. Arthritis Rheum 2002; 47 (06) 614-622
  • 137 Schnabel A, Reuter M, Biederer J, Richter C, Gross WL. Interstitial lung disease in polymyositis and dermatomyositis: clinical course and response to treatment. Semin Arthritis Rheum 2003; 32 (05) 273-284
  • 138 Marie I, Hatron PY, Dominique S, Cherin P, Mouthon L, Menard JF. Short-term and long-term outcomes of interstitial lung disease in polymyositis and dermatomyositis: a series of 107 patients. Arthritis Rheum 2011; 63 (11) 3439-3447
  • 139 Takizawa H, Shiga J, Moroi Y, Miyachi S, Nishiwaki M, Miyamoto T. Interstitial lung disease in dermatomyositis: clinicopathological study. J Rheumatol 1987; 14 (01) 102-107
  • 140 Marie I, Hatron PY, Hachulla E, Wallaert B, Michon-Pasturel U, Devulder B. Pulmonary involvement in polymyositis and in dermatomyositis. J Rheumatol 1998; 25 (07) 1336-1343
  • 141 Tazelaar HD, Viggiano RW, Pickersgill J, Colby TV. Interstitial lung disease in polymyositis and dermatomyositis. Clinical features and prognosis as correlated with histologic findings. Am Rev Respir Dis 1990; 141 (03) 727-733
  • 142 Oddis CV, Aggarwal R. Treatment in myositis. Nat Rev Rheumatol 2018; 14 (05) 279-289
  • 143 Sawal N, Mukhopadhyay S, Rayancha S. et al. A narrative review of interstitial lung disease in anti-synthetase syndrome: a clinical approach. J Thorac Dis 2021; 13 (09) 5556-5571
  • 144 Huapaya J, Wiley K, Danoff S. Treatment in antisynthetase syndrome-associated interstitial lung disease. Curr Treatm Opt Rheumatol 2021; 7 (03) 243-257
  • 145 Hallowell RW, Danoff SK. Diagnosis and management of myositis-associated lung disease. Chest 2023; 163 (06) 1476-1491
  • 146 Van Os EC, Zins BJ, Sandborn WJ. et al. Azathioprine pharmacokinetics after intravenous, oral, delayed release oral and rectal foam administration. Gut 1996; 39 (01) 63-68
  • 147 Allison AC. Mechanisms of action of mycophenolate mofetil. Lupus 2005; 14 (3, Suppl 1): s2-s8
  • 148 Ransom JT. Mechanism of action of mycophenolate mofetil. Ther Drug Monit 1995; 17 (06) 681-684
  • 149 Huapaya JA, Silhan L, Pinal-Fernandez I. et al. Long-term treatment with azathioprine and mycophenolate mofetil for myositis-related interstitial lung disease. Chest 2019; 156 (05) 896-906
  • 150 Broen JCA, van Laar JM. Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology. Nat Rev Rheumatol 2020; 16 (03) 167-178
  • 151 Schreiber SL, Crabtree GR. The mechanism of action of cyclosporin A and FK506. Immunol Today 1992; 13 (04) 136-142
  • 152 Labirua-Iturburu A, Selva-O'Callaghan A, Martínez-Gómez X, Trallero-Araguás E, Labrador-Horrillo M, Vilardell-Tarrés M. Calcineurin inhibitors in a cohort of patients with antisynthetase-associated interstitial lung disease. Clin Exp Rheumatol 2013; 31 (03) 436-439
  • 153 Cavagna L, Caporali R, Abdì-Alì L, Dore R, Meloni F, Montecucco C. Cyclosporine in anti-Jo1-positive patients with corticosteroid-refractory interstitial lung disease. J Rheumatol 2013; 40 (04) 484-492
  • 154 Koreeda Y, Higashimoto I, Yamamoto M. et al. Clinical and pathological findings of interstitial lung disease patients with anti-aminoacyl-tRNA synthetase autoantibodies. Intern Med 2010; 49 (05) 361-369
  • 155 Sharma N, Putman MS, Vij R, Strek ME, Dua A. Myositis-associated interstitial lung disease: predictors of failure of conventional treatment and response to tacrolimus in a US cohort. J Rheumatol 2017; 44 (11) 1612-1618
  • 156 Fujisawa T. Management of myositis-associated interstitial lung disease. Medicina (Kaunas) 2021; 57 (04) 347
  • 157 Wilkes MR, Sereika SM, Fertig N, Lucas MR, Oddis CV. Treatment of antisynthetase-associated interstitial lung disease with tacrolimus. Arthritis Rheum 2005; 52 (08) 2439-2446
  • 158 Takei R, Yamano Y, Kataoka K. et al. Predictive factors for the recurrence of anti-aminoacyl-tRNA synthetase antibody-associated interstitial lung disease. Respir Investig 2020; 58 (02) 83-90
  • 159 Go DJ, Park JK, Kang EH. et al. Survival benefit associated with early cyclosporine treatment for dermatomyositis-associated interstitial lung disease. Rheumatol Int 2016; 36 (01) 125-131
  • 160 Witt LJ, Demchuk C, Curran JJ, Strek ME. Benefit of adjunctive tacrolimus in connective tissue disease-interstitial lung disease. Pulm Pharmacol Ther 2016; 36: 46-52
  • 161 Chen Y, Bai Z, Zhang Z, Hu Q, Zhong J, Dong L. The efficacy and safety of tacrolimus on top of glucocorticoids in the management of IIM-ILD: a retrospective and prospective study. Front Immunol 2022; 13: 978429
  • 162 Langlois V, Gillibert A, Uzunhan Y. et al. Rituximab and cyclophosphamide in antisynthetase syndrome-related interstitial lung disease: an observational retrospective study. J Rheumatol 2020; 47 (11) 1678-1686
  • 163 Maher TM, Tudor VA, Saunders P. et al. RECITAL Investigators. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir Med 2023; 11 (01) 45-54
  • 164 Baughman RP, Meyer KC, Nathanson I. et al. Monitoring of nonsteroidal immunosuppressive drugs in patients with lung disease and lung transplant recipients: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012; 142 (05) e1S-e111S
  • 165 Knight A, Askling J, Granath F, Sparen P, Ekbom A. Urinary bladder cancer in Wegener's granulomatosis: risks and relation to cyclophosphamide. Ann Rheum Dis 2004; 63 (10) 1307-1311
  • 166 Tashkin DP, Elashoff R, Clements PJ. et al. Scleroderma Lung Study Research Group. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med 2006; 354 (25) 2655-2666
  • 167 Barnes H, Holland AE, Westall GP, Goh NSL, Glaspole IN. Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst Rev 2018; 1 (01) CD010908
  • 168 Allenbach Y, Guiguet M, Rigolet A. et al. Efficacy of rituximab in refractory inflammatory myopathies associated with anti- synthetase auto-antibodies: an open-label, phase II trial. PLoS One 2015; 10 (11) e0133702
  • 169 Xu L, Wang F, Luo F. Rituximab for the treatment of connective tissue disease-associated interstitial lung disease: a systematic review and meta-analysis. Front Pharmacol 2022; 13: 1019915
  • 170 Sacco KA, Abraham RS. Consequences of B-cell-depleting therapy: hypogammaglobulinemia and impaired B-cell reconstitution. Immunotherapy 2018; 10 (08) 713-728
  • 171 Lünemann JD, Nimmerjahn F, Dalakas MC. Intravenous immunoglobulin in neurology–mode of action and clinical efficacy. Nat Rev Neurol 2015; 11 (02) 80-89
  • 172 Kampylafka EI, Kosmidis ML, Panagiotakos DB, Dalakas M, Moutsopoulos HM, Tzioufas AG. The effect of intravenous immunoglobulin (IVIG) treatment on patients with dermatomyositis: a 4-year follow-up study. Clin Exp Rheumatol 2012; 30 (03) 397-401
  • 173 Cherin P, Pelletier S, Teixeira A. et al. Results and long-term followup of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients. Arthritis Rheum 2002; 46 (02) 467-474
  • 174 Marie I, Menard JF, Hatron PY. et al. Intravenous immunoglobulins for steroid-refractory esophageal involvement related to polymyositis and dermatomyositis: a series of 73 patients. Arthritis Care Res (Hoboken) 2010; 62 (12) 1748-1755
  • 175 Goswami RP, Haldar SN, Chatterjee M. et al. Efficacy and safety of intravenous and subcutaneous immunoglobulin therapy in idiopathic inflammatory myopathy: a systematic review and meta-analysis. Autoimmun Rev 2022; 21 (02) 102997
  • 176 Bakewell CJ, Raghu G. Polymyositis associated with severe interstitial lung disease: remission after three doses of IV immunoglobulin. Chest 2011; 139 (02) 441-443
  • 177 Suzuki Y, Hayakawa H, Miwa S. et al. Intravenous immunoglobulin therapy for refractory interstitial lung disease associated with polymyositis/dermatomyositis. Lung 2009; 187 (03) 201-206
  • 178 Diot E, Carmier D, Marquette D, Marchand-Adam S, Diot P, Lesire V. IV immunoglobulin might be considered as a first-line treatment of severe interstitial lung disease associated with polymyositis. Chest 2011; 140 (02) 562-563
  • 179 Hallowell RW, Amariei D, Danoff SK. Intravenous immunoglobulin as potential adjunct therapy for interstitial lung disease. Ann Am Thorac Soc 2016; 13 (10) 1682-1688
  • 180 Huapaya JA, Hallowell R, Silhan L. et al. Long-term treatment with human immunoglobulin for antisynthetase syndrome-associated interstitial lung disease. Respir Med 2019; 154: 6-11
  • 181 Paik JJ, Casciola-Rosen L, Shin JY. et al. Study of tofacitinib in refractory dermatomyositis: an open-label pilot study of ten patients. Arthritis Rheumatol 2021; 73 (05) 858-865
  • 182 Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis-associated interstitial lung disease. N Engl J Med 2019; 381 (03) 291-293
  • 183 Kurasawa K, Arai S, Namiki Y. et al. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford) 2018; 57 (12) 2114-2119
  • 184 Pineton de Chambrun M, Hervier B, Chauveau S, Tandjaoui-Lambiotte Y, Combes A, Uzunhan Y. Tofacitinib in antisynthetase syndrome-related rapidly progressive interstitial lung disease. Rheumatology (Oxford) 2020; 59 (12) e142-e143
  • 185 Sugino K, Ono H, Saito M, Ando M, Tsuboi E. Successful baricitinib treatment of refractory anti-synthetase syndrome associated with interstitial lung disease. Respirol Case Rep 2023; 11 (04) e01129
  • 186 Xia N, Hong SM, Zhang X. et al. Efficacy and safety of abatacept for interstitial lung disease associated with antisynthetase syndrome: a case series. Clin Exp Rheumatol 2024; 42 (02) 377-385
  • 187 Aggarwal R, Moghadam-Kia S, Koontz D. et al. POS1240 EFFICACY AND SAFETY OF ABATACEPT IN MYOSITIS ASSOCIATED INTERSTITIAL LUNG DISEASE. Ann Rheum Dis 2023; 82 (Suppl. 01) 959
  • 188 Ning Y, Yang G, Sun Y, Chen S, Liu Y, Shi G. Efficiency of therapeutic plasma-exchange in acute interstitial lung disease, associated with polymyositis/dermatomyositis resistant to glucocorticoids and immunosuppressive drugs: a retrospective study. Front Med (Lausanne) 2019; 6: 239
  • 189 Komai T, Iwasaki Y, Tsuchida Y. et al. Efficacy and safety of plasma exchange in interstitial lung diseases with anti-melanoma differentiation-associated 5 gene antibody positive clinically amyopathic dermatomyositis. Scand J Rheumatol 2023; 52 (01) 77-83
  • 190 Bozkirli DEE, Kozanoglu I, Bozkirli E, Yucel E. Antisynthetase syndrome with refractory lung involvement and myositis successfully treated with double filtration plasmapheresis. J Clin Apher 2013; 28 (06) 422-425
  • 191 Omotoso BA, Ogden MI, Balogun RA. Therapeutic plasma exchange in antisynthetase syndrome with severe interstitial lung disease. J Clin Apher 2015; 30 (06) 375-379
  • 192 Werst D, Scarpato B, Callahan SJ, Scholand MBA. A 64-year-old man with multifocal infiltrates. Chest 2021; 159 (03) e151-e154
  • 193 Thompson TZ, Bobr A, Juskewitch JE, Winters JL. Therapeutic plasma exchange for steroid refractory idiopathic inflammatory myopathies with interstitial lung disease. J Clin Apher 2023; 38 (04) 481-490
  • 194 Park JK, Yoo HG, Ahn DS, Jeon HS, Yoo WH. Successful treatment for conventional treatment-resistant dermatomyositis-associated interstitial lung disease with adalimumab. Rheumatol Int 2012; 32 (11) 3587-3590
  • 195 da Silva TCP, Zon Pretti F, Shinjo SK. Adalimumab in anti-synthetase syndrome. Joint Bone Spine 2013; 80 (04) 432
  • 196 Teng F, Peng JM, Wang Q, Tian XL, Huo Z, Weng L. Successful treatment with tocilizumab in a patient with rapidly progressive interstitial lung disease with positive anti-melanoma differentiation-associated gene-5 antibody. Chin Med J (Engl) 2020; 134 (08) 999-1000
  • 197 Zhang X, Zhou S, Wu C. et al. Tocilizumab for refractory rapidly progressive interstitial lung disease related to anti-MDA5-positive dermatomyositis. Rheumatology (Oxford) 2021; 60 (07) e227-e228
  • 198 Su CF, Liao HT, Tsai CY. Tocilizumab and rituximab for anti-MDA-5 positive amyopathic dermatomyositis complicated with macrophage activation syndrome and progressive fibrosing interstitial lung disease. Scand J Rheumatol 2022; 51 (02) 166-168
  • 199 Furlan A, Botsios C, Ruffatti A, Todesco S, Punzi L. Antisynthetase syndrome with refractory polyarthritis and fever successfully treated with the IL-1 receptor antagonist, anakinra: a case report. Joint Bone Spine 2008; 75 (03) 366-367
  • 200 Flaherty KR, Wells AU, Cottin V. et al. INBUILD Trial Investigators. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med 2019; 381 (18) 1718-1727
  • 201 Li T, Guo L, Chen Z. et al. Pirfenidone in patients with rapidly progressive interstitial lung disease associated with clinically amyopathic dermatomyositis. Sci Rep 2016; 6 (01) 33226
  • 202 Liang J, Cao H, Yang Y. et al. Efficacy and tolerability of nintedanib in idiopathic-inflammatory-myopathy-related interstitial lung disease: a pilot study. Front Med (Lausanne) 2021; 8: 626953
  • 203 Waxman A, Restrepo-Jaramillo R, Thenappan T. et al. Inhaled treprostinil in pulmonary hypertension due to interstitial lung disease. N Engl J Med 2021; 384 (04) 325-334
  • 204 Khangoora V, Bernstein EJ, King CS, Shlobin OA. Connective tissue disease-associated pulmonary hypertension: a comprehensive review. Pulm Circ 2023; 13 (04) e12276
  • 205 Tachikawa R, Tomii K, Ueda H. et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia: collagen vascular diseases-related versus idiopathic. Respiration 2012; 83 (01) 20-27
  • 206 Toyoda Y, Hanibuchi M, Kishi J. et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia associated with connective tissue disease. J Med Invest 2016; 63 (3–4): 294-299
  • 207 Cao H, Huan C, Wang Q, Xu G, Lin J, Zhou J. Predicting survival across acute exacerbation of interstitial lung disease in patients with idiopathic inflammatory myositis: the GAP-ILD model. Rheumatol Ther 2020; 7 (04) 967-978
  • 208 Rice AJ, Wells AU, Bouros D. et al. Terminal diffuse alveolar damage in relation to interstitial pneumonias. An autopsy study. Am J Clin Pathol 2003; 119 (05) 709-714
  • 209 Churg A, Müller NL, Silva CIS, Wright JL. Acute exacerbation (acute lung injury of unknown cause) in UIP and other forms of fibrotic interstitial pneumonias. Am J Surg Pathol 2007; 31 (02) 277-284
  • 210 Vuillard C, Pineton de Chambrun M, de Prost N. et al. Clinical features and outcome of patients with acute respiratory failure revealing anti-synthetase or anti-MDA-5 dermato-pulmonary syndrome: a French multicenter retrospective study. Ann Intensive Care 2018; 8 (01) 87
  • 211 Martin MJ, Moua T. Mechanical ventilation and predictors of in-hospital mortality in fibrotic interstitial lung disease with acute respiratory failure: a cohort analysis through the paradigm of acute respiratory distress syndrome. Crit Care Med 2020; 48 (07) 993-1000
  • 212 Canet E, Osman D, Lambert J. et al. Acute respiratory failure in kidney transplant recipients: a multicenter study. Crit Care 2011; 15 (02) R91
  • 213 Choo R, Naser NSH, Nadkarni NV, Anantham D. Utility of bronchoalveolar lavage in the management of immunocompromised patients presenting with lung infiltrates. BMC Pulm Med 2019; 19 (01) 51
  • 214 Tomassetti S, Colby TV, Wells AU, Poletti V, Costabel U, Matucci-Cerinic M. Bronchoalveolar lavage and lung biopsy in connective tissue diseases, to do or not to do?. Ther Adv Musculoskelet Dis 2021; 13: X211059605
  • 215 Moua T, Westerly BD, Dulohery MM, Daniels CE, Ryu JH, Lim KG. Patients with fibrotic interstitial lung disease hospitalized for acute respiratory worsening: a large cohort analysis. Chest 2016; 149 (05) 1205-1214
  • 216 Kawamura K, Ichikado K, Suga M, Yoshioka M. Efficacy of azithromycin for treatment of acute exacerbation of chronic fibrosing interstitial pneumonia: a prospective, open-label study with historical controls. Respiration 2014; 87 (06) 478-484
  • 217 Walkey AJ, Wiener RS. Macrolide antibiotics and survival in patients with acute lung injury. Chest 2012; 141 (05) 1153-1159
  • 218 Gaudry S, Vincent F, Rabbat A. et al. Invasive mechanical ventilation in patients with fibrosing interstitial pneumonia. J Thorac Cardiovasc Surg 2014; 147 (01) 47-53
  • 219 Yasuda H, Okano H, Mayumi T, Nakane M, Shime N. Association of noninvasive respiratory support with mortality and intubation rates in acute respiratory failure: a systematic review and network meta-analysis. J Intensive Care 2021; 9 (01) 32
  • 220 Slutsky AS, Ranieri VM. Ventilator-induced lung injury. N Engl J Med 2013; 369 (22) 2126-2136
  • 221 MacIntyre NR. Physiologic effects of noninvasive ventilation. Respir Care 2019; 64 (06) 617-628
  • 222 Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med 2017; 377 (06) 562-572
  • 223 Mizuno Y, Iwata H, Shirahashi K. et al. The importance of intraoperative fluid balance for the prevention of postoperative acute exacerbation of idiopathic pulmonary fibrosis after pulmonary resection for primary lung cancer. Eur J Cardiothorac Surg 2012; 41 (06) e161-e165
  • 224 Shah A, Patel SR. Acute onset anti-synthetase syndrome with pericardial effusion and non-specific interstitial pneumonia. J Clin Med Res 2016; 8 (09) 683-687
  • 225 Todd EM, Biswas Roy S, Hashimi AS. et al. Extracorporeal membrane oxygenation as a bridge to lung transplantation: a single-center experience in the present era. J Thorac Cardiovasc Surg 2017; 154 (05) 1798-1809
  • 226 Vijayasingam A, Alcalde I, Shah S. et al. AIP, Jo-1 and ECMO. Thorax 2018; 73 (12) 1190-1192
  • 227 Maloir Q, Ghysen K, von Frenckell C, Louis R, Guiot J. Acute respiratory distress revealing antisynthetase syndrome [in French]. Rev Med Liege 2018; 73 (7–8): 370-375
  • 228 Ralki M, De Langhe E, Wilmer A. et al. EJ-antisynthetase syndrome presenting as severe acute respiratory distress syndrome. Int J Tuberc Lung Dis 2021; 25 (08) 671-674
  • 229 Bay P, Lebreton G, Mathian A. et al. Outcomes of severe systemic rheumatic disease patients requiring extracorporeal membrane oxygenation. Ann Intensive Care 2021; 11 (01) 29
  • 230 Sampson C, Taylor J, Dyson L, Hassanein M. Life-threatening respiratory failure requiring extra-corporeal membrane oxygenation secondary to the anti-synthetase syndrome. J Intensive Care Soc 2021; 22 (01) 83-87
  • 231 Oda N, Taniguchi A, Aokage T. et al. Fulminant respiratory failure caused by anti-asparaginyl tRNA synthetase (Anti-KS) antibody syndrome-related interstitial lung disease. Intern Med 2022; 61 (22) 3409-3414
  • 232 Brodie D, Slutsky AS, Combes A. Extracorporeal life support for adults with respiratory failure and related indications: a review. JAMA 2019; 322 (06) 557-568
  • 233 Combes A, Schmidt M, Hodgson CL. et al. Extracorporeal life support for adults with acute respiratory distress syndrome. Intensive Care Med 2020; 46 (12) 2464-2476
  • 234 Kolb TM, Hassoun PM. Right ventricular dysfunction in chronic lung disease. Cardiol Clin 2012; 30 (02) 243-256
  • 235 Leggett RJ, Cooke NJ, Clancy L, Leitch AG, Kirby BJ, Flenley DC. Long-term domiciliary oxygen therapy in cor pulmonale complicating chronic bronchitis and emphysema. Thorax 1976; 31 (04) 414-418
  • 236 Zieliński J, Tobiasz M, Hawryłkiewicz I, Sliwiński P, Pałasiewicz G. Effects of long-term oxygen therapy on pulmonary hemodynamics in COPD patients: a 6-year prospective study. Chest 1998; 113 (01) 65-70
  • 237 Visca D, Montgomery A, de Lauretis A. et al. Ambulatory oxygen in interstitial lung disease. Eur Respir J 2011; 38 (04) 987-990
  • 238 Cao M, Wamboldt FS, Brown KK. et al. Supplemental oxygen users with pulmonary fibrosis perceive greater dyspnea than oxygen non-users. Multidiscip Respir Med 2015; 10 (01) 37
  • 239 Bell EC, Cox NS, Goh N. et al. Oxygen therapy for interstitial lung disease: a systematic review. Eur Respir Rev 2017; 26 (143) 160080
  • 240 Schaeffer MR, Ryerson CJ, Ramsook AH. et al. Effects of hyperoxia on dyspnoea and exercise endurance in fibrotic interstitial lung disease. Eur Respir J 2017; 49 (05) 1602494
  • 241 Johannson KA, Pendharkar SR, Mathison K. et al. Supplemental oxygen in interstitial lung disease: an art in need of science. Ann Am Thorac Soc 2017; 14 (09) 1373-1377
  • 242 Jacobs SS, Krishnan JA, Lederer DJ. et al. Home oxygen therapy for adults with chronic lung disease. an official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2020; 202 (10) e121-e141
  • 243 Lederer DJ, Arcasoy SM, Wilt JS, D'Ovidio F, Sonett JR, Kawut SM. Six-minute-walk distance predicts waiting list survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006; 174 (06) 659-664
  • 244 Dowman L, Hill CJ, May A, Holland AE. Pulmonary rehabilitation for interstitial lung disease. Cochrane Database Syst Rev 2021; 2 (02) CD006322
  • 245 Rochester CL, Alison JA, Carlin B. et al. Pulmonary rehabilitation for adults with chronic respiratory disease: an official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2023; 208 (04) e7-e26
  • 246 Humphrey MB, Russell L, Danila MI. et al. 2022 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Rheumatol 2023; 75 (12) 2088-2102
  • 247 Mecoli CA, Danoff SK. Pneumocystis jirovecii pneumonia and other infections in idiopathic inflammatory myositis. Curr Rheumatol Rep 2020; 22 (02) 7
  • 248 Chan SCW, Chung HY, Lau CS, Li PH. Epidemiology, mortality and effectiveness of prophylaxis for Pneumocystis jiroveci pneumonia among rheumatic patients: a territory-wide study. Ann Clin Microbiol Antimicrob 2021; 20 (01) 78
  • 249 Lee H, Chung SJ, Kim SH. et al. Treatment outcomes of infectious and non-infectious acute exacerbation of myositis-related interstitial lung disease. Front Med (Lausanne) 2022; 8: 801206
  • 250 Wilfong EM, Young-Glazer JJ, Sohn BK. et al. Anti-tRNA synthetase syndrome interstitial lung disease: a single center experience. Respir Med 2022; 191: 106432
  • 251 Ness-Jensen E, Hveem K, El-Serag H, Lagergren J. Lifestyle intervention in gastroesophageal reflux disease. Clin Gastroenterol Hepatol 2016; 14 (02) 175-82.e1 , 3
  • 252 Yang X, Wei D, Liu M. et al. Survival and outcomes after lung transplantation for connective tissue disease-associated interstitial lung diseases. Clin Rheumatol 2021; 40 (09) 3789-3795
  • 253 Courtwright AM, El-Chemaly S, Dellaripa PF, Goldberg HJ. Survival and outcomes after lung transplantation for non-scleroderma connective tissue-related interstitial lung disease. J Heart Lung Transplant 2017; 36 (07) 763-769
  • 254 Takagishi T, Ostrowski R, Alex C, Rychlik K, Pelletiere K, Tehrani R. Survival and extrapulmonary course of connective tissue disease after lung transplantation. J Clin Rheumatol 2012; 18 (06) 283-289
  • 255 Leard LE, Holm AM, Valapour M. et al. Consensus document for the selection of lung transplant candidates: an update from the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2021; 40 (11) 1349-1379
  • 256 Shao C, Sun Y, Huang H. et al. Myositis specific antibodies are associated with isolated anti-Ro-52 associated interstitial lung disease. Rheumatology (Oxford) 2022; 61 (03) 1083-1091
  • 257 Aggarwal R, Cassidy E, Fertig N. et al. Patients with non-Jo-1 anti-tRNA-synthetase autoantibodies have worse survival than Jo-1 positive patients. Ann Rheum Dis 2014; 73 (01) 227-232
  • 258 Yamakawa H, Hagiwara E, Kitamura H. et al. Predictive factors for the long-term deterioration of pulmonary function in interstitial lung disease associated with anti-aminoacyl-tRNA synthetase antibodies. Respiration 2018; 96 (03) 210-221
  • 259 González-Pérez MI, Mejía-Hurtado JG, Pérez-Román DI. et al. Evolution of pulmonary function in a cohort of patients with interstitial lung disease and positive for antisynthetase antibodies. J Rheumatol 2020; 47 (03) 415-423
  • 260 Liu H, Xie S, Liang T. et al. Prognostic factors of interstitial lung disease progression at sequential HRCT in anti-synthetase syndrome. Eur Radiol 2019; 29 (10) 5349-5357
  • 261 Rojas-Serrano J, Herrera-Bringas D, Mejía M, Rivero H, Mateos-Toledo H, Figueroa JE. Prognostic factors in a cohort of antisynthetase syndrome (ASS): serologic profile is associated with mortality in patients with interstitial lung disease (ILD). Clin Rheumatol 2015; 34 (09) 1563-1569
  • 262 Marie I, Hatron PY, Cherin P. et al. Functional outcome and prognostic factors in anti-Jo1 patients with antisynthetase syndrome. Arthritis Res Ther 2013; 15 (05) R149
  • 263 Mecoli CA, Igusa T, Chen M. et al. Subsets of idiopathic inflammatory myositis enriched for contemporaneous cancer relative to the general population. Arthritis Rheumatol 2023; 75 (04) 620-629
  • 264 Da Silva LMB, Rathore U, Agarwal V, Gupta L, Shinjo SK. Demographic, clinical, laboratory data, prognostic, and treatment features of patients with antisynthetase syndrome: an international, two-center cohort study. Arch Rheumatol 2022; 37 (03) 424-434
  • 265 Martins P, Dourado E, Melo AT. et al. Clinical characterisation of a multicentre nationwide cohort of patients with antisynthetase syndrome. ARP Rheumatol 2022; 1 (ARP Rheumatology, n°3 2022): 190-196
  • 266 Tang M, Wang Q, Wei W. et al. A multi-center clinical cohort study of Chinese anti-synthetase syndrome patients. Arthritis Rheumatol 2022; 74: 3671-3672
  • 267 Tang HS, Tang IYK, Ho RTC. et al. Clinical heterogeneity and prognostic factors of anti-synthetase syndrome: a multi-centered retrospective cohort study. Rheumatology (Oxford) 2023; (e-pub ahead of print). DOI: 10.1093/RHEUMATOLOGY/KEAD671.
  • 268 Sodsri T, Petnak T, Ngamjanyaporn P. Clinical characteristics of anti-synthetase syndrome and variables associated with interstitial lung disease and mortality: a retrospective cohort study. J Clin Med 2023; 12 (21) 6849
  • 269 Guimarães F, Yildirim R, Isenberg DA. Long-term survival of patients with idiopathic inflammatory myopathies: anatomy of a single-centre cohort. Clin Exp Rheumatol 2023; 41 (02) 322-329
  • 270 Muro Y, Nishida K, Yamashita Y. et al. Comment on: favourable complete remission of anti-OJ antibody-positive myositis after lung cancer resection. Rheumatology (Oxford) 2022; 61 (08) e232-e234

Address for correspondence

Sonye K. Danoff, MD, PhD
Division of Pulmonary and Critical Care Medicine
1830 E. Monument Street, 5th Floor, Baltimore, MD 21205

Publication History

Article published online:
06 May 2024

© 2024. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

  • References

  • 1 Lundberg IE, Tjärnlund A, Bottai M. et al. 2017 European League Against Rheumatism/American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups. Ann Rheum Dis 2017; 76 (12) 1955-1964
  • 2 Hochberg MC, Feldman D, Stevens MB, Arnett FC, Reichlin M. Antibody to Jo-1 in polymyositis/dermatomyositis: association with interstitial pulmonary disease. J Rheumatol 1984; 11 (05) 663-665
  • 3 Marguerie C, Bunn CC, Beynon HLC. et al. Polymyositis, pulmonary fibrosis and autoantibodies to aminoacyl-tRNA synthetase enzymes. Q J Med 1990; 77 (282) 1019-1038
  • 4 Connors GR, Christopher-Stine L, Oddis CV, Danoff SK. Interstitial lung disease associated with the idiopathic inflammatory myopathies: what progress has been made in the past 35 years?. Chest 2010; 138 (06) 1464-1474
  • 5 Solomon J, Swigris JJ, Brown KK. Myositis-related interstitial lung disease and antisynthetase syndrome. J Bras Pneumol 2011; 37 (01) 100-109
  • 6 Lega JC, Reynaud Q, Belot A, Fabien N, Durieu I, Cottin V. Idiopathic inflammatory myopathies and the lung. Eur Respir Rev 2015; 24 (136) 216-238
  • 7 Khoo T, Lilleker JB, Thong BYH, Leclair V, Lamb JA, Chinoy H. Epidemiology of the idiopathic inflammatory myopathies. Nat Rev Rheumatol 2023; 19 (11) 695-712
  • 8 Galindo-Feria AS, Notarnicola A, Lundberg IE, Horuluoglu B. Aminoacyl-tRNA synthetases: on anti-synthetase syndrome and beyond. Front Immunol 2022; 13: 866087
  • 9 Wells M, Alawi S, Thin KYM. et al. A multidisciplinary approach to the diagnosis of antisynthetase syndrome. Front Med (Lausanne) 2022; 9: 959653
  • 10 Tan JA, Roberts-Thomson PJ, Blumbergs P, Hakendorf P, Cox SR, Limaye V. Incidence and prevalence of idiopathic inflammatory myopathies in South Australia: a 30-year epidemiologic study of histology-proven cases. Int J Rheum Dis 2013; 16 (03) 331-338
  • 11 Limaye V, Hakendorf P, Woodman RJ, Blumbergs P, Roberts-Thomson P. Mortality and its predominant causes in a large cohort of patients with biopsy-determined inflammatory myositis. Intern Med J 2012; 42 (02) 191-198
  • 12 Limaye V, Luke C, Tucker G. et al. The incidence and associations of malignancy in a large cohort of patients with biopsy-determined idiopathic inflammatory myositis. Rheumatol Int 2013; 33 (04) 965-971
  • 13 Barsotti S, Lundberg IE. Myositis an evolving spectrum of disease. Immunol Med 2018; 41 (02) 46-54
  • 14 Hum RM, Lilleker JB, Lamb JA. et al. MYONET registry. Comparison of clinical features between patients with anti-synthetase syndrome and dermatomyositis: results from the MYONET registry. Rheumatology (Oxford) 2023; 00: 1-8
  • 15 Zhang Y, Ge Y, Yang H. et al. Clinical features and outcomes of the patients with anti-glycyl tRNA synthetase syndrome. Clin Rheumatol 2020; 39 (08) 2417-2424
  • 16 Koenig M, Fritzler MJ, Targoff IN, Troyanov Y, Senécal JL. Heterogeneity of autoantibodies in 100 patients with autoimmune myositis: insights into clinical features and outcomes. Arthritis Res Ther 2007; 9 (04) R78
  • 17 Shi J, Li S, Yang H. et al. Clinical profiles and prognosis of patients with distinct antisynthetase autoantibodies. J Rheumatol 2017; 44 (07) 1051-1057
  • 18 Pinal-Fernandez I, Casal-Dominguez M, Huapaya JA. et al. A longitudinal cohort study of the anti-synthetase syndrome: increased severity of interstitial lung disease in black patients and patients with anti-PL7 and anti-PL12 autoantibodies. Rheumatology (Oxford) 2017; 56 (06) 999-1007
  • 19 Noguchi E, Uruha A, Suzuki S. et al. Skeletal muscle involvement in antisynthetase syndrome. JAMA Neurol 2017; 74 (08) 992-999
  • 20 Orphanet: Antisynthetase syndrome. Accessed December 17, 2023 at: https://www.orpha.net/consor/cgi-bin/OC_Exp.php?Lng=GB&Expert=81
  • 21 Lilleker JB, Vencovsky J, Wang G. et al. all EuroMyositis contributors. The EuroMyositis registry: an international collaborative tool to facilitate myositis research. Ann Rheum Dis 2018; 77 (01) 30-39
  • 22 Cavagna L, Trallero-Araguás E, Meloni F. et al. Influence of Antisynthetase antibodies specificities on antisynthetase syndrome clinical spectrum time course. J Clin Med 2019; 8 (11) 2013
  • 23 Wang R, Zhao Y, Qi F. et al. Analysis of the clinical features of antisynthetase syndrome: a retrospective cohort study in China. Clin Rheumatol 2023; 42 (03) 703-709
  • 24 Deligny C, Dueymes M, Arfi S. et al. Epidemiology and characteristics of antisynthetase syndrome in the African descent population of martinique. Arthritis Rheumatol 2014; 66: S556
  • 25 Hamaguchi Y, Fujimoto M, Matsushita T. et al. Common and distinct clinical features in adult patients with anti-aminoacyl-tRNA synthetase antibodies: heterogeneity within the syndrome. PLoS One 2013; 8 (04) e60442
  • 26 Li W, Li J, Xie WM, Ren YH, Dai HP. Clinical characteristics of patients with antisynthetase syndrome and interstitial pulmonary disease [in Chinese]. Zhonghua Yi Xue Za Zhi 2020; 100 (24) 1861-1865
  • 27 Dugar M, Cox S, Limaye V, Blumbergs P, Roberts-Thomson PJ. Clinical heterogeneity and prognostic features of South Australian patients with anti-synthetase autoantibodies. Intern Med J 2011; 41 (09) 674-679
  • 28 Mescam-Mancini L, Allenbach Y, Hervier B. et al. Anti-Jo-1 antibody-positive patients show a characteristic necrotizing perifascicular myositis. Brain 2015; 138 (Pt 9): 2485-2492
  • 29 Uruha A, Nishikawa A, Tsuburaya RS. et al. Sarcoplasmic MxA expression: a valuable marker of dermatomyositis. Neurology 2017; 88 (05) 493-500
  • 30 Selva-O'Callaghan A, Pinal-Fernandez I, Trallero-Araguás E, Milisenda JC, Grau-Junyent JM, Mammen AL. Classification and management of adult inflammatory myopathies. Lancet Neurol 2018; 17 (09) 816-828
  • 31 Gutierrez-Arcelus M, Rich SS, Raychaudhuri S. Autoimmune diseases - connecting risk alleles with molecular traits of the immune system. Nat Rev Genet 2016; 17 (03) 160-174
  • 32 Rubtsova K, Marrack P, Rubtsov AV. Sexual dimorphism in autoimmunity. J Clin Invest 2015; 125 (06) 2187-2193
  • 33 Haghikia A, Jörg S, Duscha A. et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity 2015; 43 (04) 817-829
  • 34 Gallay L, Gayed C, Hervier B. Antisynthetase syndrome pathogenesis: knowledge and uncertainties. Curr Opin Rheumatol 2018; 30 (06) 664-673
  • 35 Svensson J, Holmqvist M, Lundberg IE, Arkema EV. Infections and respiratory tract disease as risk factors for idiopathic inflammatory myopathies: a population-based case-control study. Ann Rheum Dis 2017; 76 (11) 1803-1808
  • 36 Webber MP, Moir W, Zeig-Owens R. et al. Nested case-control study of selected systemic autoimmune diseases in World Trade Center rescue/recovery workers. Arthritis Rheumatol 2015; 67 (05) 1369-1376
  • 37 Katsumata Y, Ridgway WM, Oriss T. et al. Species-specific immune responses generated by histidyl-tRNA synthetase immunization are associated with muscle and lung inflammation. J Autoimmun 2007; 29 (2-3): 174-186
  • 38 Albrecht I, Eddie J, Fathi M. et al. Characterization of histidyl-tRNA synthetase specific Th cell response in blood and broncheoalveolar lavage (BAL) of myositis patients. ACR Meeting Abstracts. Published online 2013. Accessed November 18, 2023 at: https://acrabstracts.org/abstract/characterization-of-histidyl-trna-synthetase-specific-th-cell-response-in-blood-and-broncheoalveolar-lavage-bal-of-myositis-patients/
  • 39 Englund P, Wahlström J, Fathi M. et al. Restricted T cell receptor BV gene usage in the lungs and muscles of patients with idiopathic inflammatory myopathies. Arthritis Rheum 2007; 56 (01) 372-383
  • 40 Galindo-Feria AS, Albrecht I, Notarnicola A. et al. 02.09 Identification of a novel pro-inflammatory T cell epitope from his-trna-synthetase associated with interstitial lung disease in anti-jo-1 positive patients. Ann Rheum Dis 2017; 76: A11.2-A12
  • 41 Barbasso Helmers S, Englund P, Engström M. et al. Sera from anti-Jo-1-positive patients with polymyositis and interstitial lung disease induce expression of intercellular adhesion molecule 1 in human lung endothelial cells. Arthritis Rheum 2009; 60 (08) 2524-2530
  • 42 Guo M, Schimmel P. Essential nontranslational functions of tRNA synthetases. Nat Chem Biol 2013; 9 (03) 145-153
  • 43 Lo WS, Gardiner E, Xu Z. et al. Human tRNA synthetase catalytic nulls with diverse functions. Science 2014; 345 (6194) 328-332
  • 44 Adams RA, Fernandes-Cerqueira C, Notarnicola A. et al. Serum-circulating His-tRNA synthetase inhibits organ-targeted immune responses. Cell Mol Immunol 2021; 18 (06) 1463-1475
  • 45 Miller FW, Chen W, O'Hanlon TP. et al. Myositis Genetics Consortium. Genome-wide association study identifies HLA 8.1 ancestral haplotype alleles as major genetic risk factors for myositis phenotypes. Genes Immun 2015; 16 (07) 470-480
  • 46 Rothwell S, Lamb JA, Chinoy H. New developments in genetics of myositis. Curr Opin Rheumatol 2016; 28 (06) 651-656
  • 47 Rothwell S, Chinoy H, Lamb JA. et al. Myositis Genetics Consortium (MYOGEN). Focused HLA analysis in Caucasians with myositis identifies significant associations with autoantibody subgroups. Ann Rheum Dis 2019; 78 (07) 996-1002
  • 48 Remuzgo-Martínez S, Atienza-Mateo B, Ocejo-Vinyals JG. et al. HLA association with the susceptibility to anti-synthetase syndrome. Joint Bone Spine 2021; 88 (03) 105115
  • 49 Opinc AH, Makowska JS. Antisynthetase syndrome - much more than just a myopathy. Semin Arthritis Rheum 2021; 51 (01) 72-83
  • 50 Jiang M, Dong X, Zheng Y. Clinical characteristics of interstitial lung diseases positive to different anti-synthetase antibodies. Medicine (Baltimore) 2021; 100 (19) e25816
  • 51 Vulsteke JB, Satoh M, Malyavantham K, Bossuyt X, De Langhe E, Mahler M. Anti-OJ autoantibodies: rare or underdetected?. Autoimmun Rev 2019; 18 (07) 658-664
  • 52 Marie I, Hatron PY, Dominique S. et al. Short-term and long-term outcome of anti-Jo1-positive patients with anti-Ro52 antibody. Semin Arthritis Rheum 2012; 41 (06) 890-899
  • 53 Trallero-Araguás E, Grau-Junyent JM, Labirua-Iturburu A. et al. IIM Study Group and Autoimmune Diseases Study Group (GEAS) of the Spanish Society of Internal Medicine (SEMI). Clinical manifestations and long-term outcome of anti-Jo1 antisynthetase patients in a large cohort of Spanish patients from the GEAS-IIM group. Semin Arthritis Rheum 2016; 46 (02) 225-231
  • 54 Cavagna L, Nuño L, Scirè CA. et al. AENEAS (American, European NEtwork of Antisynthetase Syndrome) collaborative group.. Clinical spectrum time course in anti jo-1 positive antisynthetase syndrome: results from an international retrospective multicenter study. Medicine (Baltimore) 2015; 94 (32) e1144
  • 55 Hervier B, Devilliers H, Stanciu R. et al. Hierarchical cluster and survival analyses of antisynthetase syndrome: phenotype and outcome are correlated with anti-tRNA synthetase antibody specificity. Autoimmun Rev 2012; 12 (02) 210-217
  • 56 Tanboon J, Inoue M, Hirakawa S. et al. Muscle pathology of antisynthetase syndrome according to antibody subtypes. Brain Pathol 2023; 33 (04) e13155
  • 57 Szabó K, Bodoki L, Nagy-Vincze M. et al. Effect of genetic and laboratory findings on clinical course of antisynthetase syndrome in a Hungarian cohort. BioMed Res Int 2018; 2018: 6416378
  • 58 Kaneko Y, Hanaoka H, Hirakata M, Takeuchi T, Kuwana M. Distinct arthropathies of the hands in patients with anti-aminoacyl tRNA synthetase antibodies: usefulness of autoantibody profiles in classifying patients. Rheumatology (Oxford) 2014; 53 (06) 1120-1124
  • 59 Meyer A, Lefevre G, Bierry G. et al. Club Rhumatismes et Inflammation. In antisynthetase syndrome, ACPA are associated with severe and erosive arthritis: an overlapping rheumatoid arthritis and antisynthetase syndrome. Medicine (Baltimore) 2015; 94 (20) e523
  • 60 Kumar RR, Jha S, Dhooria A. et al. Anti-Jo-1 syndrome often misdiagnosed as rheumatoid arthritis (for many years): a single-center experience. J Clin Rheumatol 2021; 27 (04) 150-155
  • 61 Miller JB, Danoff SK, Bingham III CO. et al. Sonographic findings from inflammatory arthritis due to antisynthetase syndrome. Clin Rheumatol 2019; 38 (05) 1477-1483
  • 62 Gusdorf L, Morruzzi C, Goetz J, Lipsker D, Sibilia J, Cribier B. Mechanics hands in patients with antisynthetase syndrome: 25 cases. Ann Dermatol Venereol 2019; 146 (01) 19-25
  • 63 Cox JT, Gullotti DM, Mecoli CA. et al. “Hiker's feet”: a novel cutaneous finding in the inflammatory myopathies. Clin Rheumatol 2017; 36 (07) 1683-1686
  • 64 Hosono Y, Ishii A, Izumi Y. et al. New aspects of clinical and immunological characteristics in patients with anti-asparaginyl tRNA synthetase (anti-KS) autoantibody. Mod Rheumatol 2023; 34 (01) 122-128
  • 65 Marie I, Josse S, Hatron PY. et al. Interstitial lung disease in anti-Jo-1 patients with antisynthetase syndrome. Arthritis Care Res (Hoboken) 2013; 65 (05) 800-808
  • 66 Richards TJ, Eggebeen A, Gibson K. et al. Characterization and peripheral blood biomarker assessment of anti-Jo-1 antibody-positive interstitial lung disease. Arthritis Rheum 2009; 60 (07) 2183-2192
  • 67 Marie I, Josse S, Decaux O. et al. Comparison of long-term outcome between anti-Jo1- and anti-PL7/PL12 positive patients with antisynthetase syndrome. Autoimmun Rev 2012; 11 (10) 739-745
  • 68 Johnson C, Connors GR, Oaks J. et al. Clinical and pathologic differences in interstitial lung disease based on antisynthetase antibody type. Respir Med 2014; 108 (10) 1542-1548
  • 69 Tillie-Leblond I, Wislez M, Valeyre D. et al. Interstitial lung disease and anti-Jo-1 antibodies: difference between acute and gradual onset. Thorax 2008; 63 (01) 53-59
  • 70 Cruellas MG, Viana VdosS, Levy-Neto M, Souza FH, Shinjo SK. Myositis-specific and myositis-associated autoantibody profiles and their clinical associations in a large series of patients with polymyositis and dermatomyositis. Clinics (São Paulo) 2013; 68 (07) 909-914
  • 71 Bauhammer J, Blank N, Max R. et al. Rituximab in the treatment of Jo1 antibody-associated antisynthetase syndrome: anti-Ro52 positivity as a marker for severity and treatment response. J Rheumatol 2016; 43 (08) 1566-1574
  • 72 Zhao Y, Zhang C, Su H. et al. Predictive factors for progressive fibrosing interstitial lung disease in anti-synthetase syndrome. Int J Rheum Dis 2023; 26 (05) 885-894
  • 73 Váncsa A, Csípo I, Németh J, Dévényi K, Gergely L, Dankó K. Characteristics of interstitial lung disease in SS-A positive/Jo-1 positive inflammatory myopathy patients. Rheumatol Int 2009; 29 (09) 989-994
  • 74 La Corte R, Lo Mo Naco A, Locaputo A, Dolzani F, Trotta F. In patients with antisynthetase syndrome the occurrence of anti-Ro/SSA antibodies causes a more severe interstitial lung disease. Autoimmunity 2006; 39 (03) 249-253
  • 75 Pepper E, Vilar L, Ward IM. Clinical characteristics and prognostic value of Ro52/SSA antibodies in idiopathic inflammatory myopathies. J Clin Rheumatol 2023; 29 (07) 347-353
  • 76 Zamora AC, Hoskote SS, Abascal-Bolado B. et al. Clinical features and outcomes of interstitial lung disease in anti-Jo-1 positive antisynthetase syndrome. Respir Med 2016; 118: 39-45
  • 77 Zhan X, Yan W, Wang Y. et al. Clinical features of anti-synthetase syndrome associated interstitial lung disease: a retrospective cohort in China. BMC Pulm Med 2021; 21 (01) 57
  • 78 American Thoracic Society/European Respiratory Society. ATS/ERS Statement on respiratory muscle testing. Am J Respir Crit Care Med 2002; 166 (04) 518-624
  • 79 Laveneziana P, Albuquerque A, Aliverti A. et al. ERS statement on respiratory muscle testing at rest and during exercise. Eur Respir J 2019; 53 (06) 1801214
  • 80 Debray MP, Borie R, Revel MP. et al. Interstitial lung disease in anti-synthetase syndrome: initial and follow-up CT findings. Eur J Radiol 2015; 84 (03) 516-523
  • 81 Waseda Y, Johkoh T, Egashira R. et al. Antisynthetase syndrome: pulmonary computed tomography findings of adult patients with antibodies to aminoacyl-tRNA synthetases. Eur J Radiol 2016; 85 (08) 1421-1426
  • 82 Liu Y, Liu X, Xie M. et al. Clinical characteristics of patients with anti-EJ antisynthetase syndrome associated interstitial lung disease and literature review. Respir Med 2020; 165: 105920
  • 83 Chung JH, Cox CW, Montner SM. et al. CT features of the usual interstitial pneumonia pattern: differentiating connective tissue disease-associated interstitial lung disease from idiopathic pulmonary fibrosis. AJR Am J Roentgenol 2018; 210 (02) 307-313
  • 84 Barratt SL, Adamali HH, Cotton C. et al. Clinicoserological features of antisynthetase syndrome (ASyS)-associated interstitial lung disease presenting to respiratory services: comparison with idiopathic pulmonary fibrosis and ASyS diagnosed in rheumatology services. BMJ Open Respir Res 2021; 8 (01) e000829
  • 85 Baratella E, Marrocchio C, Cifaldi R. et al. Interstitial lung disease in patients with antisynthetase syndrome: a retrospective case series study. Jpn J Radiol 2021; 39 (01) 40-46
  • 86 Chartrand S, Swigris JJ, Peykova L, Chung J, Fischer A. A multidisciplinary evaluation helps identify the antisynthetase syndrome in patients presenting as idiopathic interstitial pneumonia. J Rheumatol 2016; 43 (05) 887-892
  • 87 Yura H, Sakamoto N, Satoh M. et al. Clinical characteristics of patients with anti-aminoacyl-tRNA synthetase antibody positive idiopathic interstitial pneumonia. Respir Med 2017; 132: 189-194
  • 88 Doyle TJ, Dhillon N, Madan R. et al. Rituximab in the treatment of interstitial lung disease associated with antisynthetase syndrome: a multi-center retrospective case review. J Rheumatol 2018; 45 (06) 841-850
  • 89 Cobo-Ibáñez T, López-Longo FJ, Joven B. et al. Long-term pulmonary outcomes and mortality in idiopathic inflammatory myopathies associated with interstitial lung disease. Clin Rheumatol 2019; 38 (03) 803-815
  • 90 Fukamatsu H, Hirai Y, Miyake T. et al. Clinical manifestations of skin, lung and muscle diseases in dermatomyositis positive for anti-aminoacyl tRNA synthetase antibodies. J Dermatol 2019; 46 (10) 886-897
  • 91 Teel A, Lu J, Park J, Singh N, Basharat P. The role of myositis-specific autoantibodies and the management of interstitial lung disease in idiopathic inflammatory myopathies: a systematic review. Semin Arthritis Rheum 2022; 57: 152088
  • 92 Schneider F, Yousem SA, Oddis CV, Aggarwal R. Pulmonary pathologic manifestations of anti-alanyl-tRNA synthetase (anti-PL-12)-related inflammatory myopathy. Arch Pathol Lab Med 2018; 142 (02) 191-197
  • 93 Yamasaki Y, Yamada H, Yamasaki M. et al. Intravenous cyclophosphamide therapy for progressive interstitial pneumonia in patients with polymyositis/dermatomyositis. Rheumatology (Oxford) 2007; 46 (01) 124-130
  • 94 Yousem SA, Schneider F, Bi D, Oddis CV, Gibson K, Aggarwal R. The pulmonary histopathologic manifestations of the anti-PL7/antithreonyl transfer RNA synthetase syndrome. Hum Pathol 2014; 45 (06) 1199-1204
  • 95 Marie I, Josse S, Decaux O. et al. Clinical manifestations and outcome of anti-PL7 positive patients with antisynthetase syndrome. Eur J Intern Med 2013; 24 (05) 474-479
  • 96 Sasano H, Hagiwara E, Kitamura H. et al. Long-term clinical course of anti-glycyl tRNA synthetase (anti-EJ) antibody-related interstitial lung disease pathologically proven by surgical lung biopsy. BMC Pulm Med 2016; 16 (01) 168
  • 97 Watanabe K, Handa T, Tanizawa K. et al. Detection of antisynthetase syndrome in patients with idiopathic interstitial pneumonias. Respir Med 2011; 105 (08) 1238-1247
  • 98 Schneider F, Yousem SA, Bi D, Gibson KF, Oddis CV, Aggarwal R. Pulmonary pathologic manifestations of anti-glycyl-tRNA synthetase (anti-EJ)-related inflammatory myopathy. J Clin Pathol 2014; 67 (08) 678-683
  • 99 Aiko N, Yamakawa H, Iwasawa T. et al. Clinical, radiological, and pathological features of anti-asparaginyl tRNA synthetase antibody-related interstitial lung disease. Respir Investig 2020; 58 (03) 196-203
  • 100 Schneider F, Aggarwal R, Bi D, Gibson K, Oddis C, Yousem SA. The pulmonary histopathology of anti-KS transfer RNA synthetase syndrome. Arch Pathol Lab Med 2015; 139 (01) 122-125
  • 101 Sato S, Kuwana M, Hirakata M. Clinical characteristics of Japanese patients with anti-OJ (anti-isoleucyl-tRNA synthetase) autoantibodies. Rheumatology (Oxford) 2007; 46 (05) 842-845
  • 102 Flashner BM, VanderLaan PA, Nurhussien L, Rice MB, Hallowell RW. Pulmonary histopathology of interstitial lung disease associated with antisynthetase antibodies. Respir Med 2022; 191: 106697
  • 103 Condon DF, Nickel NP, Anderson R, Mirza S, de Jesus Perez VA. The 6th World Symposium on Pulmonary Hypertension: what's old is new. F1000Res 2019; 8: F1000 Faculty Rev-888
  • 104 Hervier B, Meyer A, Dieval C. et al. Pulmonary hypertension in antisynthetase syndrome: prevalence, aetiology and survival. Eur Respir J 2013; 42 (05) 1271-1282
  • 105 Lecomte R, Perrin F, Journeau L. et al. Syndrome des antisynthétases compliqué d'une hypertension pulmonaire : 4 observations originales. Rev Med Interne 2015; 36 (12) 794-799
  • 106 Hervier B, Wallaert B, Hachulla E. et al. Clinical manifestations of anti-synthetase syndrome positive for anti-alanyl-tRNA synthetase (anti-PL12) antibodies: a retrospective study of 17 cases. Rheumatology (Oxford) 2010; 49 (05) 972-976
  • 107 Kalluri M, Sahn SA, Oddis CV. et al. Clinical profile of anti-PL-12 autoantibody. Cohort study and review of the literature. Chest 2009; 135 (06) 1550-1556
  • 108 Bryan JL, Matar R, Raviprasad A. et al. Echocardiographic characteristics of patients with antisynthetase syndrome. Pulm Circ 2022; 12 (02) e12084
  • 109 Syed N, Yadav R, O'Rourke C, Chatterjee S. Pulmonary hypertension in anti-JO1 syndrome. Arthritis Rheum 2013; 65: S886
  • 110 Reddy R, Bryan JL, Raviprasad A. et al. Pulmonary hypertension in anti-synthetase syndrome. Am J Respir Crit Care Med 2021; 203 (09) A3581
  • 111 Mason T, Samaranayake C, Mccabe C. et al. Use of pulmonary vasodilator therapy in anti-synthetase syndrome-associated pulmonary hypertension. Eur Respir J 2023; 62 (Suppl. 67) PA3495
  • 112 Saito G, Kono M, Tsutsumi A. et al. Anti-PL-7 antisynthetase syndrome with eosinophilic pleural effusion. Intern Med 2018; 57 (15) 2227-2232
  • 113 Mogulkoc N, Kabasakal Y, Ekren PK, Bishop PW. An unusual presentation of anti-Jo-1 syndrome, mimicking lung metastases, with massive pleural and pericardial effusions. J Clin Rheumatol 2006; 12 (02) 90-92
  • 114 Wu Y, Chhaya S, Hurowitz B, Ardiles T, Carlson R. Clinically amyopathic dermatomyositis complicated by pleural effusion case report, literature review, and proposed mechanism. Bull Hosp Jt Dis 2015; 73 (03) 217-220
  • 115 Sugie K, Tonomura Y, Ueno S. Characterization of dermatomyositis with coexistence of anti-Jo-1 and anti-SRP antibodies. Intern Med 2012; 51 (07) 799-802
  • 116 Chen L, Somers A, Liu J. Antisynthetase syndrome and pleural effusion: a case report. Chest 2020; 158 (04) A1991-A1992
  • 117 Bhandari BS, Yeramareddy S, Akella S, Rawal H, Martin KB. An unusual case of anti-synthetase syndrome with pleurisy and pleural effusions. Am J Respir Crit Care Med 2019; 199 (09) A3116
  • 118 Katz A, Bena J, Chatterjee S. Antisynthetase syndrome: prevalence of serositis in autoantibody subsets. Arthritis Rheumatol 2018; 70: 397-399
  • 119 Lega JC, Fabien N, Reynaud Q. et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun Rev 2014; 13 (09) 883-891
  • 120 Jiang L, Jones J, Yang XL. Human diseases linked to cytoplasmic aminoacyl-tRNA synthetases. Enzymes 2020; 48: 277-319
  • 121 Vulsteke JB, Derua R, Dubucquoi S. et al. Mass spectrometry-based identification of new anti-Ly and known antisynthetase autoantibodies. Ann Rheum Dis 2023; 82 (04) 546-555
  • 122 Sasai T, Nakashima R, Shirakashi M. et al. A new autoantibody to valyl transfer RNA synthetase associated with anti-synthetase syndrome. Rheumatology (Oxford) 2023; 62 (05) e155-e157
  • 123 Sidorik LL. Seryl-tRNA synthetase &ndash; a novel autoantigen during systemic autoimmune diseases. Biopolymers Cell 1994; 10 (3–4): 68-74
  • 124 Vartanyan OA. Detection of antibodies against tryptophanyl-, tyrosyl- and phenylalanyl-tRNA synthetase and antiidiotypic antibodies aganist them in sera of patients bearing autoimmune diseases. Molekulyarnaya Biologiya 1991; 25 (04) 1033-1039
  • 125 Paley EL, Alexandrova N, Smelansky L. Tryptophanyl-tRNA synthetase as a human autoantigen. Immunol Lett 1995; 48 (03) 201-207
  • 126 Mathews MB, Bernstein RM. Myositis autoantibody inhibits histidyl-tRNA synthetase: a model for autoimmunity. Nature 1983; 304 (5922) 177-179
  • 127 Andersson H, Sem M, Lund MB. et al. Long-term experience with rituximab in anti-synthetase syndrome-related interstitial lung disease. Rheumatology (Oxford) 2015; 54 (08) 1420-1428
  • 128 Bernstein RM, Morgan SH, Chapman J. et al. Anti-Jo-1 antibody: a marker for myositis with interstitial lung disease. Br Med J (Clin Res Ed) 1984; 289 (6438) 151-152
  • 129 Stone KB, Oddis CV, Fertig N. et al. Anti-Jo-1 antibody levels correlate with disease activity in idiopathic inflammatory myopathy. Arthritis Rheum 2007; 56 (09) 3125-3131
  • 130 Zanframundo G, Faghihi-Kashani S, Scirè CA. et al. Defining anti-synthetase syndrome: a systematic literature review. Clin Exp Rheumatol 2022; 40 (02) 309-319
  • 131 Mecoli CA, Albayda J, Tiniakou E. et al. Myositis Autoantibodies: a comparison of results from the Oklahoma Medical Research Foundation Myositis panel to the euroimmun research line blot. Arthritis Rheumatol 2020; 72 (01) 192-194
  • 132 Zhang W, Zheng Y, Wang Y. et al. Thigh MRI in antisynthetase syndrome, and comparisons with dermatomyositis and immune-mediated necrotizing myopathy. Rheumatology (Oxford) 2022; 62 (01) 310-320
  • 133 Andersson H, Kirkhus E, Garen T, Walle-Hansen R, Merckoll E, Molberg Ø. Comparative analyses of muscle MRI and muscular function in anti-synthetase syndrome patients and matched controls: a cross-sectional study. Arthritis Res Ther 2017; 19 (01) 17
  • 134 Barba T, Fort R, Cottin V. et al. Treatment of idiopathic inflammatory myositis associated interstitial lung disease: a systematic review and meta-analysis. Autoimmun Rev 2019; 18 (02) 113-122
  • 135 Schwarz MI, Matthay RA, Sahn SA, Stanford RE, Marmorstein BL, Scheinhorn DJ. Interstitial lung disease in polymyositis and dermatomyositis: analysis of six cases and review of the literature. Medicine (Baltimore) 1976; 55 (01) 89-104
  • 136 Marie I, Hachulla E, Chérin P. et al. Interstitial lung disease in polymyositis and dermatomyositis. Arthritis Rheum 2002; 47 (06) 614-622
  • 137 Schnabel A, Reuter M, Biederer J, Richter C, Gross WL. Interstitial lung disease in polymyositis and dermatomyositis: clinical course and response to treatment. Semin Arthritis Rheum 2003; 32 (05) 273-284
  • 138 Marie I, Hatron PY, Dominique S, Cherin P, Mouthon L, Menard JF. Short-term and long-term outcomes of interstitial lung disease in polymyositis and dermatomyositis: a series of 107 patients. Arthritis Rheum 2011; 63 (11) 3439-3447
  • 139 Takizawa H, Shiga J, Moroi Y, Miyachi S, Nishiwaki M, Miyamoto T. Interstitial lung disease in dermatomyositis: clinicopathological study. J Rheumatol 1987; 14 (01) 102-107
  • 140 Marie I, Hatron PY, Hachulla E, Wallaert B, Michon-Pasturel U, Devulder B. Pulmonary involvement in polymyositis and in dermatomyositis. J Rheumatol 1998; 25 (07) 1336-1343
  • 141 Tazelaar HD, Viggiano RW, Pickersgill J, Colby TV. Interstitial lung disease in polymyositis and dermatomyositis. Clinical features and prognosis as correlated with histologic findings. Am Rev Respir Dis 1990; 141 (03) 727-733
  • 142 Oddis CV, Aggarwal R. Treatment in myositis. Nat Rev Rheumatol 2018; 14 (05) 279-289
  • 143 Sawal N, Mukhopadhyay S, Rayancha S. et al. A narrative review of interstitial lung disease in anti-synthetase syndrome: a clinical approach. J Thorac Dis 2021; 13 (09) 5556-5571
  • 144 Huapaya J, Wiley K, Danoff S. Treatment in antisynthetase syndrome-associated interstitial lung disease. Curr Treatm Opt Rheumatol 2021; 7 (03) 243-257
  • 145 Hallowell RW, Danoff SK. Diagnosis and management of myositis-associated lung disease. Chest 2023; 163 (06) 1476-1491
  • 146 Van Os EC, Zins BJ, Sandborn WJ. et al. Azathioprine pharmacokinetics after intravenous, oral, delayed release oral and rectal foam administration. Gut 1996; 39 (01) 63-68
  • 147 Allison AC. Mechanisms of action of mycophenolate mofetil. Lupus 2005; 14 (3, Suppl 1): s2-s8
  • 148 Ransom JT. Mechanism of action of mycophenolate mofetil. Ther Drug Monit 1995; 17 (06) 681-684
  • 149 Huapaya JA, Silhan L, Pinal-Fernandez I. et al. Long-term treatment with azathioprine and mycophenolate mofetil for myositis-related interstitial lung disease. Chest 2019; 156 (05) 896-906
  • 150 Broen JCA, van Laar JM. Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology. Nat Rev Rheumatol 2020; 16 (03) 167-178
  • 151 Schreiber SL, Crabtree GR. The mechanism of action of cyclosporin A and FK506. Immunol Today 1992; 13 (04) 136-142
  • 152 Labirua-Iturburu A, Selva-O'Callaghan A, Martínez-Gómez X, Trallero-Araguás E, Labrador-Horrillo M, Vilardell-Tarrés M. Calcineurin inhibitors in a cohort of patients with antisynthetase-associated interstitial lung disease. Clin Exp Rheumatol 2013; 31 (03) 436-439
  • 153 Cavagna L, Caporali R, Abdì-Alì L, Dore R, Meloni F, Montecucco C. Cyclosporine in anti-Jo1-positive patients with corticosteroid-refractory interstitial lung disease. J Rheumatol 2013; 40 (04) 484-492
  • 154 Koreeda Y, Higashimoto I, Yamamoto M. et al. Clinical and pathological findings of interstitial lung disease patients with anti-aminoacyl-tRNA synthetase autoantibodies. Intern Med 2010; 49 (05) 361-369
  • 155 Sharma N, Putman MS, Vij R, Strek ME, Dua A. Myositis-associated interstitial lung disease: predictors of failure of conventional treatment and response to tacrolimus in a US cohort. J Rheumatol 2017; 44 (11) 1612-1618
  • 156 Fujisawa T. Management of myositis-associated interstitial lung disease. Medicina (Kaunas) 2021; 57 (04) 347
  • 157 Wilkes MR, Sereika SM, Fertig N, Lucas MR, Oddis CV. Treatment of antisynthetase-associated interstitial lung disease with tacrolimus. Arthritis Rheum 2005; 52 (08) 2439-2446
  • 158 Takei R, Yamano Y, Kataoka K. et al. Predictive factors for the recurrence of anti-aminoacyl-tRNA synthetase antibody-associated interstitial lung disease. Respir Investig 2020; 58 (02) 83-90
  • 159 Go DJ, Park JK, Kang EH. et al. Survival benefit associated with early cyclosporine treatment for dermatomyositis-associated interstitial lung disease. Rheumatol Int 2016; 36 (01) 125-131
  • 160 Witt LJ, Demchuk C, Curran JJ, Strek ME. Benefit of adjunctive tacrolimus in connective tissue disease-interstitial lung disease. Pulm Pharmacol Ther 2016; 36: 46-52
  • 161 Chen Y, Bai Z, Zhang Z, Hu Q, Zhong J, Dong L. The efficacy and safety of tacrolimus on top of glucocorticoids in the management of IIM-ILD: a retrospective and prospective study. Front Immunol 2022; 13: 978429
  • 162 Langlois V, Gillibert A, Uzunhan Y. et al. Rituximab and cyclophosphamide in antisynthetase syndrome-related interstitial lung disease: an observational retrospective study. J Rheumatol 2020; 47 (11) 1678-1686
  • 163 Maher TM, Tudor VA, Saunders P. et al. RECITAL Investigators. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir Med 2023; 11 (01) 45-54
  • 164 Baughman RP, Meyer KC, Nathanson I. et al. Monitoring of nonsteroidal immunosuppressive drugs in patients with lung disease and lung transplant recipients: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012; 142 (05) e1S-e111S
  • 165 Knight A, Askling J, Granath F, Sparen P, Ekbom A. Urinary bladder cancer in Wegener's granulomatosis: risks and relation to cyclophosphamide. Ann Rheum Dis 2004; 63 (10) 1307-1311
  • 166 Tashkin DP, Elashoff R, Clements PJ. et al. Scleroderma Lung Study Research Group. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med 2006; 354 (25) 2655-2666
  • 167 Barnes H, Holland AE, Westall GP, Goh NSL, Glaspole IN. Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst Rev 2018; 1 (01) CD010908
  • 168 Allenbach Y, Guiguet M, Rigolet A. et al. Efficacy of rituximab in refractory inflammatory myopathies associated with anti- synthetase auto-antibodies: an open-label, phase II trial. PLoS One 2015; 10 (11) e0133702
  • 169 Xu L, Wang F, Luo F. Rituximab for the treatment of connective tissue disease-associated interstitial lung disease: a systematic review and meta-analysis. Front Pharmacol 2022; 13: 1019915
  • 170 Sacco KA, Abraham RS. Consequences of B-cell-depleting therapy: hypogammaglobulinemia and impaired B-cell reconstitution. Immunotherapy 2018; 10 (08) 713-728
  • 171 Lünemann JD, Nimmerjahn F, Dalakas MC. Intravenous immunoglobulin in neurology–mode of action and clinical efficacy. Nat Rev Neurol 2015; 11 (02) 80-89
  • 172 Kampylafka EI, Kosmidis ML, Panagiotakos DB, Dalakas M, Moutsopoulos HM, Tzioufas AG. The effect of intravenous immunoglobulin (IVIG) treatment on patients with dermatomyositis: a 4-year follow-up study. Clin Exp Rheumatol 2012; 30 (03) 397-401
  • 173 Cherin P, Pelletier S, Teixeira A. et al. Results and long-term followup of intravenous immunoglobulin infusions in chronic, refractory polymyositis: an open study with thirty-five adult patients. Arthritis Rheum 2002; 46 (02) 467-474
  • 174 Marie I, Menard JF, Hatron PY. et al. Intravenous immunoglobulins for steroid-refractory esophageal involvement related to polymyositis and dermatomyositis: a series of 73 patients. Arthritis Care Res (Hoboken) 2010; 62 (12) 1748-1755
  • 175 Goswami RP, Haldar SN, Chatterjee M. et al. Efficacy and safety of intravenous and subcutaneous immunoglobulin therapy in idiopathic inflammatory myopathy: a systematic review and meta-analysis. Autoimmun Rev 2022; 21 (02) 102997
  • 176 Bakewell CJ, Raghu G. Polymyositis associated with severe interstitial lung disease: remission after three doses of IV immunoglobulin. Chest 2011; 139 (02) 441-443
  • 177 Suzuki Y, Hayakawa H, Miwa S. et al. Intravenous immunoglobulin therapy for refractory interstitial lung disease associated with polymyositis/dermatomyositis. Lung 2009; 187 (03) 201-206
  • 178 Diot E, Carmier D, Marquette D, Marchand-Adam S, Diot P, Lesire V. IV immunoglobulin might be considered as a first-line treatment of severe interstitial lung disease associated with polymyositis. Chest 2011; 140 (02) 562-563
  • 179 Hallowell RW, Amariei D, Danoff SK. Intravenous immunoglobulin as potential adjunct therapy for interstitial lung disease. Ann Am Thorac Soc 2016; 13 (10) 1682-1688
  • 180 Huapaya JA, Hallowell R, Silhan L. et al. Long-term treatment with human immunoglobulin for antisynthetase syndrome-associated interstitial lung disease. Respir Med 2019; 154: 6-11
  • 181 Paik JJ, Casciola-Rosen L, Shin JY. et al. Study of tofacitinib in refractory dermatomyositis: an open-label pilot study of ten patients. Arthritis Rheumatol 2021; 73 (05) 858-865
  • 182 Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis-associated interstitial lung disease. N Engl J Med 2019; 381 (03) 291-293
  • 183 Kurasawa K, Arai S, Namiki Y. et al. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford) 2018; 57 (12) 2114-2119
  • 184 Pineton de Chambrun M, Hervier B, Chauveau S, Tandjaoui-Lambiotte Y, Combes A, Uzunhan Y. Tofacitinib in antisynthetase syndrome-related rapidly progressive interstitial lung disease. Rheumatology (Oxford) 2020; 59 (12) e142-e143
  • 185 Sugino K, Ono H, Saito M, Ando M, Tsuboi E. Successful baricitinib treatment of refractory anti-synthetase syndrome associated with interstitial lung disease. Respirol Case Rep 2023; 11 (04) e01129
  • 186 Xia N, Hong SM, Zhang X. et al. Efficacy and safety of abatacept for interstitial lung disease associated with antisynthetase syndrome: a case series. Clin Exp Rheumatol 2024; 42 (02) 377-385
  • 187 Aggarwal R, Moghadam-Kia S, Koontz D. et al. POS1240 EFFICACY AND SAFETY OF ABATACEPT IN MYOSITIS ASSOCIATED INTERSTITIAL LUNG DISEASE. Ann Rheum Dis 2023; 82 (Suppl. 01) 959
  • 188 Ning Y, Yang G, Sun Y, Chen S, Liu Y, Shi G. Efficiency of therapeutic plasma-exchange in acute interstitial lung disease, associated with polymyositis/dermatomyositis resistant to glucocorticoids and immunosuppressive drugs: a retrospective study. Front Med (Lausanne) 2019; 6: 239
  • 189 Komai T, Iwasaki Y, Tsuchida Y. et al. Efficacy and safety of plasma exchange in interstitial lung diseases with anti-melanoma differentiation-associated 5 gene antibody positive clinically amyopathic dermatomyositis. Scand J Rheumatol 2023; 52 (01) 77-83
  • 190 Bozkirli DEE, Kozanoglu I, Bozkirli E, Yucel E. Antisynthetase syndrome with refractory lung involvement and myositis successfully treated with double filtration plasmapheresis. J Clin Apher 2013; 28 (06) 422-425
  • 191 Omotoso BA, Ogden MI, Balogun RA. Therapeutic plasma exchange in antisynthetase syndrome with severe interstitial lung disease. J Clin Apher 2015; 30 (06) 375-379
  • 192 Werst D, Scarpato B, Callahan SJ, Scholand MBA. A 64-year-old man with multifocal infiltrates. Chest 2021; 159 (03) e151-e154
  • 193 Thompson TZ, Bobr A, Juskewitch JE, Winters JL. Therapeutic plasma exchange for steroid refractory idiopathic inflammatory myopathies with interstitial lung disease. J Clin Apher 2023; 38 (04) 481-490
  • 194 Park JK, Yoo HG, Ahn DS, Jeon HS, Yoo WH. Successful treatment for conventional treatment-resistant dermatomyositis-associated interstitial lung disease with adalimumab. Rheumatol Int 2012; 32 (11) 3587-3590
  • 195 da Silva TCP, Zon Pretti F, Shinjo SK. Adalimumab in anti-synthetase syndrome. Joint Bone Spine 2013; 80 (04) 432
  • 196 Teng F, Peng JM, Wang Q, Tian XL, Huo Z, Weng L. Successful treatment with tocilizumab in a patient with rapidly progressive interstitial lung disease with positive anti-melanoma differentiation-associated gene-5 antibody. Chin Med J (Engl) 2020; 134 (08) 999-1000
  • 197 Zhang X, Zhou S, Wu C. et al. Tocilizumab for refractory rapidly progressive interstitial lung disease related to anti-MDA5-positive dermatomyositis. Rheumatology (Oxford) 2021; 60 (07) e227-e228
  • 198 Su CF, Liao HT, Tsai CY. Tocilizumab and rituximab for anti-MDA-5 positive amyopathic dermatomyositis complicated with macrophage activation syndrome and progressive fibrosing interstitial lung disease. Scand J Rheumatol 2022; 51 (02) 166-168
  • 199 Furlan A, Botsios C, Ruffatti A, Todesco S, Punzi L. Antisynthetase syndrome with refractory polyarthritis and fever successfully treated with the IL-1 receptor antagonist, anakinra: a case report. Joint Bone Spine 2008; 75 (03) 366-367
  • 200 Flaherty KR, Wells AU, Cottin V. et al. INBUILD Trial Investigators. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med 2019; 381 (18) 1718-1727
  • 201 Li T, Guo L, Chen Z. et al. Pirfenidone in patients with rapidly progressive interstitial lung disease associated with clinically amyopathic dermatomyositis. Sci Rep 2016; 6 (01) 33226
  • 202 Liang J, Cao H, Yang Y. et al. Efficacy and tolerability of nintedanib in idiopathic-inflammatory-myopathy-related interstitial lung disease: a pilot study. Front Med (Lausanne) 2021; 8: 626953
  • 203 Waxman A, Restrepo-Jaramillo R, Thenappan T. et al. Inhaled treprostinil in pulmonary hypertension due to interstitial lung disease. N Engl J Med 2021; 384 (04) 325-334
  • 204 Khangoora V, Bernstein EJ, King CS, Shlobin OA. Connective tissue disease-associated pulmonary hypertension: a comprehensive review. Pulm Circ 2023; 13 (04) e12276
  • 205 Tachikawa R, Tomii K, Ueda H. et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia: collagen vascular diseases-related versus idiopathic. Respiration 2012; 83 (01) 20-27
  • 206 Toyoda Y, Hanibuchi M, Kishi J. et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia associated with connective tissue disease. J Med Invest 2016; 63 (3–4): 294-299
  • 207 Cao H, Huan C, Wang Q, Xu G, Lin J, Zhou J. Predicting survival across acute exacerbation of interstitial lung disease in patients with idiopathic inflammatory myositis: the GAP-ILD model. Rheumatol Ther 2020; 7 (04) 967-978
  • 208 Rice AJ, Wells AU, Bouros D. et al. Terminal diffuse alveolar damage in relation to interstitial pneumonias. An autopsy study. Am J Clin Pathol 2003; 119 (05) 709-714
  • 209 Churg A, Müller NL, Silva CIS, Wright JL. Acute exacerbation (acute lung injury of unknown cause) in UIP and other forms of fibrotic interstitial pneumonias. Am J Surg Pathol 2007; 31 (02) 277-284
  • 210 Vuillard C, Pineton de Chambrun M, de Prost N. et al. Clinical features and outcome of patients with acute respiratory failure revealing anti-synthetase or anti-MDA-5 dermato-pulmonary syndrome: a French multicenter retrospective study. Ann Intensive Care 2018; 8 (01) 87
  • 211 Martin MJ, Moua T. Mechanical ventilation and predictors of in-hospital mortality in fibrotic interstitial lung disease with acute respiratory failure: a cohort analysis through the paradigm of acute respiratory distress syndrome. Crit Care Med 2020; 48 (07) 993-1000
  • 212 Canet E, Osman D, Lambert J. et al. Acute respiratory failure in kidney transplant recipients: a multicenter study. Crit Care 2011; 15 (02) R91
  • 213 Choo R, Naser NSH, Nadkarni NV, Anantham D. Utility of bronchoalveolar lavage in the management of immunocompromised patients presenting with lung infiltrates. BMC Pulm Med 2019; 19 (01) 51
  • 214 Tomassetti S, Colby TV, Wells AU, Poletti V, Costabel U, Matucci-Cerinic M. Bronchoalveolar lavage and lung biopsy in connective tissue diseases, to do or not to do?. Ther Adv Musculoskelet Dis 2021; 13: X211059605
  • 215 Moua T, Westerly BD, Dulohery MM, Daniels CE, Ryu JH, Lim KG. Patients with fibrotic interstitial lung disease hospitalized for acute respiratory worsening: a large cohort analysis. Chest 2016; 149 (05) 1205-1214
  • 216 Kawamura K, Ichikado K, Suga M, Yoshioka M. Efficacy of azithromycin for treatment of acute exacerbation of chronic fibrosing interstitial pneumonia: a prospective, open-label study with historical controls. Respiration 2014; 87 (06) 478-484
  • 217 Walkey AJ, Wiener RS. Macrolide antibiotics and survival in patients with acute lung injury. Chest 2012; 141 (05) 1153-1159
  • 218 Gaudry S, Vincent F, Rabbat A. et al. Invasive mechanical ventilation in patients with fibrosing interstitial pneumonia. J Thorac Cardiovasc Surg 2014; 147 (01) 47-53
  • 219 Yasuda H, Okano H, Mayumi T, Nakane M, Shime N. Association of noninvasive respiratory support with mortality and intubation rates in acute respiratory failure: a systematic review and network meta-analysis. J Intensive Care 2021; 9 (01) 32
  • 220 Slutsky AS, Ranieri VM. Ventilator-induced lung injury. N Engl J Med 2013; 369 (22) 2126-2136
  • 221 MacIntyre NR. Physiologic effects of noninvasive ventilation. Respir Care 2019; 64 (06) 617-628
  • 222 Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med 2017; 377 (06) 562-572
  • 223 Mizuno Y, Iwata H, Shirahashi K. et al. The importance of intraoperative fluid balance for the prevention of postoperative acute exacerbation of idiopathic pulmonary fibrosis after pulmonary resection for primary lung cancer. Eur J Cardiothorac Surg 2012; 41 (06) e161-e165
  • 224 Shah A, Patel SR. Acute onset anti-synthetase syndrome with pericardial effusion and non-specific interstitial pneumonia. J Clin Med Res 2016; 8 (09) 683-687
  • 225 Todd EM, Biswas Roy S, Hashimi AS. et al. Extracorporeal membrane oxygenation as a bridge to lung transplantation: a single-center experience in the present era. J Thorac Cardiovasc Surg 2017; 154 (05) 1798-1809
  • 226 Vijayasingam A, Alcalde I, Shah S. et al. AIP, Jo-1 and ECMO. Thorax 2018; 73 (12) 1190-1192
  • 227 Maloir Q, Ghysen K, von Frenckell C, Louis R, Guiot J. Acute respiratory distress revealing antisynthetase syndrome [in French]. Rev Med Liege 2018; 73 (7–8): 370-375
  • 228 Ralki M, De Langhe E, Wilmer A. et al. EJ-antisynthetase syndrome presenting as severe acute respiratory distress syndrome. Int J Tuberc Lung Dis 2021; 25 (08) 671-674
  • 229 Bay P, Lebreton G, Mathian A. et al. Outcomes of severe systemic rheumatic disease patients requiring extracorporeal membrane oxygenation. Ann Intensive Care 2021; 11 (01) 29
  • 230 Sampson C, Taylor J, Dyson L, Hassanein M. Life-threatening respiratory failure requiring extra-corporeal membrane oxygenation secondary to the anti-synthetase syndrome. J Intensive Care Soc 2021; 22 (01) 83-87
  • 231 Oda N, Taniguchi A, Aokage T. et al. Fulminant respiratory failure caused by anti-asparaginyl tRNA synthetase (Anti-KS) antibody syndrome-related interstitial lung disease. Intern Med 2022; 61 (22) 3409-3414
  • 232 Brodie D, Slutsky AS, Combes A. Extracorporeal life support for adults with respiratory failure and related indications: a review. JAMA 2019; 322 (06) 557-568
  • 233 Combes A, Schmidt M, Hodgson CL. et al. Extracorporeal life support for adults with acute respiratory distress syndrome. Intensive Care Med 2020; 46 (12) 2464-2476
  • 234 Kolb TM, Hassoun PM. Right ventricular dysfunction in chronic lung disease. Cardiol Clin 2012; 30 (02) 243-256
  • 235 Leggett RJ, Cooke NJ, Clancy L, Leitch AG, Kirby BJ, Flenley DC. Long-term domiciliary oxygen therapy in cor pulmonale complicating chronic bronchitis and emphysema. Thorax 1976; 31 (04) 414-418
  • 236 Zieliński J, Tobiasz M, Hawryłkiewicz I, Sliwiński P, Pałasiewicz G. Effects of long-term oxygen therapy on pulmonary hemodynamics in COPD patients: a 6-year prospective study. Chest 1998; 113 (01) 65-70
  • 237 Visca D, Montgomery A, de Lauretis A. et al. Ambulatory oxygen in interstitial lung disease. Eur Respir J 2011; 38 (04) 987-990
  • 238 Cao M, Wamboldt FS, Brown KK. et al. Supplemental oxygen users with pulmonary fibrosis perceive greater dyspnea than oxygen non-users. Multidiscip Respir Med 2015; 10 (01) 37
  • 239 Bell EC, Cox NS, Goh N. et al. Oxygen therapy for interstitial lung disease: a systematic review. Eur Respir Rev 2017; 26 (143) 160080
  • 240 Schaeffer MR, Ryerson CJ, Ramsook AH. et al. Effects of hyperoxia on dyspnoea and exercise endurance in fibrotic interstitial lung disease. Eur Respir J 2017; 49 (05) 1602494
  • 241 Johannson KA, Pendharkar SR, Mathison K. et al. Supplemental oxygen in interstitial lung disease: an art in need of science. Ann Am Thorac Soc 2017; 14 (09) 1373-1377
  • 242 Jacobs SS, Krishnan JA, Lederer DJ. et al. Home oxygen therapy for adults with chronic lung disease. an official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2020; 202 (10) e121-e141
  • 243 Lederer DJ, Arcasoy SM, Wilt JS, D'Ovidio F, Sonett JR, Kawut SM. Six-minute-walk distance predicts waiting list survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006; 174 (06) 659-664
  • 244 Dowman L, Hill CJ, May A, Holland AE. Pulmonary rehabilitation for interstitial lung disease. Cochrane Database Syst Rev 2021; 2 (02) CD006322
  • 245 Rochester CL, Alison JA, Carlin B. et al. Pulmonary rehabilitation for adults with chronic respiratory disease: an official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med 2023; 208 (04) e7-e26
  • 246 Humphrey MB, Russell L, Danila MI. et al. 2022 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Rheumatol 2023; 75 (12) 2088-2102
  • 247 Mecoli CA, Danoff SK. Pneumocystis jirovecii pneumonia and other infections in idiopathic inflammatory myositis. Curr Rheumatol Rep 2020; 22 (02) 7
  • 248 Chan SCW, Chung HY, Lau CS, Li PH. Epidemiology, mortality and effectiveness of prophylaxis for Pneumocystis jiroveci pneumonia among rheumatic patients: a territory-wide study. Ann Clin Microbiol Antimicrob 2021; 20 (01) 78
  • 249 Lee H, Chung SJ, Kim SH. et al. Treatment outcomes of infectious and non-infectious acute exacerbation of myositis-related interstitial lung disease. Front Med (Lausanne) 2022; 8: 801206
  • 250 Wilfong EM, Young-Glazer JJ, Sohn BK. et al. Anti-tRNA synthetase syndrome interstitial lung disease: a single center experience. Respir Med 2022; 191: 106432
  • 251 Ness-Jensen E, Hveem K, El-Serag H, Lagergren J. Lifestyle intervention in gastroesophageal reflux disease. Clin Gastroenterol Hepatol 2016; 14 (02) 175-82.e1 , 3
  • 252 Yang X, Wei D, Liu M. et al. Survival and outcomes after lung transplantation for connective tissue disease-associated interstitial lung diseases. Clin Rheumatol 2021; 40 (09) 3789-3795
  • 253 Courtwright AM, El-Chemaly S, Dellaripa PF, Goldberg HJ. Survival and outcomes after lung transplantation for non-scleroderma connective tissue-related interstitial lung disease. J Heart Lung Transplant 2017; 36 (07) 763-769
  • 254 Takagishi T, Ostrowski R, Alex C, Rychlik K, Pelletiere K, Tehrani R. Survival and extrapulmonary course of connective tissue disease after lung transplantation. J Clin Rheumatol 2012; 18 (06) 283-289
  • 255 Leard LE, Holm AM, Valapour M. et al. Consensus document for the selection of lung transplant candidates: an update from the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2021; 40 (11) 1349-1379
  • 256 Shao C, Sun Y, Huang H. et al. Myositis specific antibodies are associated with isolated anti-Ro-52 associated interstitial lung disease. Rheumatology (Oxford) 2022; 61 (03) 1083-1091
  • 257 Aggarwal R, Cassidy E, Fertig N. et al. Patients with non-Jo-1 anti-tRNA-synthetase autoantibodies have worse survival than Jo-1 positive patients. Ann Rheum Dis 2014; 73 (01) 227-232
  • 258 Yamakawa H, Hagiwara E, Kitamura H. et al. Predictive factors for the long-term deterioration of pulmonary function in interstitial lung disease associated with anti-aminoacyl-tRNA synthetase antibodies. Respiration 2018; 96 (03) 210-221
  • 259 González-Pérez MI, Mejía-Hurtado JG, Pérez-Román DI. et al. Evolution of pulmonary function in a cohort of patients with interstitial lung disease and positive for antisynthetase antibodies. J Rheumatol 2020; 47 (03) 415-423
  • 260 Liu H, Xie S, Liang T. et al. Prognostic factors of interstitial lung disease progression at sequential HRCT in anti-synthetase syndrome. Eur Radiol 2019; 29 (10) 5349-5357
  • 261 Rojas-Serrano J, Herrera-Bringas D, Mejía M, Rivero H, Mateos-Toledo H, Figueroa JE. Prognostic factors in a cohort of antisynthetase syndrome (ASS): serologic profile is associated with mortality in patients with interstitial lung disease (ILD). Clin Rheumatol 2015; 34 (09) 1563-1569
  • 262 Marie I, Hatron PY, Cherin P. et al. Functional outcome and prognostic factors in anti-Jo1 patients with antisynthetase syndrome. Arthritis Res Ther 2013; 15 (05) R149
  • 263 Mecoli CA, Igusa T, Chen M. et al. Subsets of idiopathic inflammatory myositis enriched for contemporaneous cancer relative to the general population. Arthritis Rheumatol 2023; 75 (04) 620-629
  • 264 Da Silva LMB, Rathore U, Agarwal V, Gupta L, Shinjo SK. Demographic, clinical, laboratory data, prognostic, and treatment features of patients with antisynthetase syndrome: an international, two-center cohort study. Arch Rheumatol 2022; 37 (03) 424-434
  • 265 Martins P, Dourado E, Melo AT. et al. Clinical characterisation of a multicentre nationwide cohort of patients with antisynthetase syndrome. ARP Rheumatol 2022; 1 (ARP Rheumatology, n°3 2022): 190-196
  • 266 Tang M, Wang Q, Wei W. et al. A multi-center clinical cohort study of Chinese anti-synthetase syndrome patients. Arthritis Rheumatol 2022; 74: 3671-3672
  • 267 Tang HS, Tang IYK, Ho RTC. et al. Clinical heterogeneity and prognostic factors of anti-synthetase syndrome: a multi-centered retrospective cohort study. Rheumatology (Oxford) 2023; (e-pub ahead of print). DOI: 10.1093/RHEUMATOLOGY/KEAD671.
  • 268 Sodsri T, Petnak T, Ngamjanyaporn P. Clinical characteristics of anti-synthetase syndrome and variables associated with interstitial lung disease and mortality: a retrospective cohort study. J Clin Med 2023; 12 (21) 6849
  • 269 Guimarães F, Yildirim R, Isenberg DA. Long-term survival of patients with idiopathic inflammatory myopathies: anatomy of a single-centre cohort. Clin Exp Rheumatol 2023; 41 (02) 322-329
  • 270 Muro Y, Nishida K, Yamashita Y. et al. Comment on: favourable complete remission of anti-OJ antibody-positive myositis after lung cancer resection. Rheumatology (Oxford) 2022; 61 (08) e232-e234

Zoom Image
Fig. 1 Extrapulmonary manifestations of ASyS. (A) V-neck rash. (B) Gottron's papules. (C) Gottron's papules. (D) Mechanic's hand. (E) Mechanic's feet. (F) Calcinosis. (G) Ragged cuticles and periungual erythema. ASyS, antisynthetase syndrome.
Zoom Image
Fig. 2 Different CT patterns in patients with ASyS. (A) Ground glass opacities, reticular thickening and subpleural sparing consistent with a NSIP in a patient with anti-PL12 serology. (B) Ground glass opacities, reticular thickening, subpleural sparing, and traction bronchiectasis consistent with a fibrotic NSIP in a patient with anti-PL7 serology. (C) Interstitial infiltrates with ground glass opacities, alveolar nodules, and subpleural bands consistent with an OP in a patient with anti-Jo1 serology. (D) Diffuse subpleural reticulation, fibrosis, traction bronchiectasis, and honeycombing consistent with an UIP pattern in a patient with anti-Jo1 serology. ASyS, antisynthetase syndrome; CT, computed tomography; OP, organizing pneumonia; NSIP, nonspecific interstitial pneumonia; UIP, usual interstitial pneumonia.
Zoom Image
Fig. 3 Algorithm suggesting management for antisynthetase associated ILD (ASyS-ILD) according to severity of disease. Decision for the use of chronic immunosuppression and agent of choice is governed by several variables including disease severity, response to initial therapy, familiarity of physician with medication, co-morbidities, and disease course. AZA, azathioprine; ECMO, extracorporeal membrane oxygenation; IVIG, IV immunoglobulin; MMF, mycophenolate; PFT, pulmonary function test; PLEX, plasma exchange; RTX, rituximab; TAC, tacrolimus. Adapted from Hallowell & Danoff.[145]