5,534
Views
2
CrossRef citations to date
0
Altmetric
Review

Management of progressive pulmonary fibrosis associated with connective tissue disease

, , , , , & show all
Pages 765-774 | Received 28 Apr 2022, Accepted 26 Jul 2022, Published online: 05 Aug 2022

ABSTRACT

Introduction

Fibrotic interstitial lung disease (ILD) is a frequent and severe complication of connective tissue disease (CTD).

Areas covered

In this narrative review, we update the most relevant differential characteristics of fibrotic ILD associated with CTD (CTD-ILD) and propose a diagnostic and therapeutic approach based on a review of the articles published between 2002 and 2022 through PubMed.

Expert opinion

The subset of ILD, mainly the radiological/histological pattern and the degree of fibrotic component, usually determines the prognosis and therapeutic strategy for these patients. Some patients with CTD-ILD can develop progressive pulmonary fibrosis (PPF) with severe deterioration of lung function, rapid progression to chronic respiratory failure, and high mortality. PPF has been described in many CTDs, mainly in systemic sclerosis and rheumatoid arthritis, and requires a multidisciplinary diagnostic and therapeutic approach to improve patient outcomes.

1. Introduction

Interstitial lung disease (ILD) has been described in almost all connective tissue diseases (CTDs) [Citation1–12], with higher prevalence in systemic sclerosis (SSc) [Citation2,Citation3], rheumatoid arthritis (RA) [Citation4,Citation5], and in some groups of inflammatory myositis (dermatomyositis, antisynthetase syndrome, and overlap syndromes) [Citation6–9].

The pathogenesis of ILD associated with CTD (CTD-ILD) is not completely elucidated, although the main hypothesis assumes that tissue fibrosis is preceded by an immune-mediate process [Citation1–6,Citation13–16]. In SSc, this process seems to be triggered by endothelial injury in the context of encompassing immune activity [Citation3,Citation14,Citation15]. In the last decades, some research studies have suggested different pathways to be implicated in the development of ILD in SSc and RA, including immune-mediated alveolar epithelial damage and endothelial disorders, and the subsequent and progressive abnormal extracellular matrix remodeling and myofibroblast formation [Citation17]. Following the identification of these pathogenic pathways and implicated mediators, different future potential strategies are being pre-clinically evaluated in SSc-associated ILD (SSc-ILD) and RA-associated ILD (RA-ILD), including the regulation of some relevant overexpressed extracellular matrix proteins such as COL5A2 [Citation18,Citation19].

The clinical course of CTD-ILD is variable, but patients with evidence of pulmonary fibrosis tend to have a worse prognosis. A proportion of fibrotic CTD-ILD patients will develop progressive pulmonary fibrosis (PPF) and rapid deterioration of pulmonary function tests (PFTs), leading to end-stage respiratory failure and high mortality [Citation13,Citation20–23]. Moreover, it has been described that fibrotic ILD is characterized by an increased distal alveolar bacterial burden that might be responsible for both a rapidly deteriorating course of fibrotic disease and deadly exacerbations [Citation24,Citation25]. It has also been shown, that RA-ILD has a higher serious infections risk, especially pneumonia [Citation26,Citation27].

The diagnosis of fibrotic CTD-ILD, especially the early identification of patients with PPF, is complex. In these cases, a comprehensive assessment is essential, including accurate staging of the severity of the disease and extrapulmonary manifestations. In addition, an individualized and comprehensive therapeutic approach is important in the treatment of CTD-ILD to control the autoimmune and inflammatory activity of the underlying disease and to prevent, when necessary, the progression of fibrotic pulmonary changes [Citation28]. For all these reasons, a multidisciplinary approach is worthwhile in the treatment of the different stages in CTD-ILD, for diagnosis and treatment (both initial and during follow-up) [Citation29].

The aim of this article is to describe the most relevant differential characteristics of PPF in CTD and to propose a diagnostic and therapeutic approach based on the published evidence and the clinical experience of a multidisciplinary group of experts. Narrative literature review (PubMed Central) was used to search for the articles published between 2002 and 2022, excluding clinical cases. The terms ‘interstitial lung disease’, ‘progressive fibrosis’, and ‘connective tissue disease’ were used in the literature search.

2. Identification and diagnosis of CTD-ILD

ILD may present as a complication during the course of a known CTD or the clinical onset of a CTD. In both scenarios, a multidisciplinary diagnostic and therapeutic approach is ideally advised [Citation29–31].

In patients with a known CTD, this multidisciplinary model of care has been shown to reduce the time to ILD identification, as well as the time between diagnosis and initiation of treatment [Citation29]. In addition, assessing the severity of the ILD and associated extrapulmonary manifestations is essential for decision making [Citation29].

In patients with new-onset ILD, multidisciplinary assessment allows a specific diagnosis to be established in up to 80.5% of cases [Citation17,Citation29–33]. But, most importantly, it allows early identification of patients with an autoimmune pathogenic substrate, either by reaching the diagnosis of a definite CTD or by establishing the initial orientation of interstitial pneumonia with autoimmune features (IPAF), which has prognostic and therapeutic implications [Citation30,Citation31,Citation34,Citation35].

The recommended composition of a multidisciplinary committee in CTD-ILD should not only include expert ILD pulmonologists, radiologists, and pathologists, but also include experts in CTD (rheumatologists) [Citation31,Citation32].

Notably in CTD, some ILD may be subclinical until advanced stages and the detection of ILD is frequently performed with months or even years of delay [Citation1–12], particularly in RA [Citation4,Citation5,Citation16,Citation36,Citation37]. Increasing data suggest that the diagnostic delay associate a poor outcome and that ILD identification can influence the therapeutic approach [Citation17,Citation29–35]. Therefore, the early detection of this complication is essential. Addressing this concern, regularly monitoring respiratory symptoms and auscultation is essential; ‘velcro-type’ crackles to lung auscultation, correlates very well with the presence of pulmonary fibrosis [Citation38]. Clinically, ILD presents with repetitive dry cough and exertional dyspnea, which may progress to respiratory failure.

lists those patient features that can help to identify fibrotic CTD-ILD patients [Citation17,Citation29–31,Citation35,Citation39]. In case of suspected ILD, a chest X-ray and PFTs including spirometry and carbon monoxide diffusing capacity (DLCO) are usually performed for initial screening. However, chest X-ray is a technique with low sensitivity in the early stages, when only high-resolution computed tomography (HRCT) detects the mild interstitial changes, being the most sensitive imaging tool to detect ILD. The PFTs in fibrotic ILD show a restrictive ventilatory (decrease of total lung capacity [TLC] and forced vital capacity [FVC]) pattern together with a decrease in DLCO. However, in early stages, a decrease in DLCO with preserved FVC may be found. Furthermore, if only a DLCO decrease is present or the severity of DLCO decrease is not associated with a TLC or FVC decline, pulmonary arterial hypertension (PAH) should be considered, especially in SSc patients

Table 1. Patient features and invasive and noninvasive tests useful for identifying an ILD in a CTD.

To confirm ILD and to characterize its pattern, the chest HRCT is essential. Due to the HRCT radiologic-histologic correlation pattern and the association of CTD as a cause of ILD, lung biopsy is not usually required. HRCT is also useful to quantify the extent of parenchymal involvement, the type of interstitial changes (predominant fibrotic or ground-glass) and pattern (usual interstitial pneumonia [UIP] vs. non-UIP), and to assess the disease progression. shows the radiological patterns and signs of fibrosis on chest HRCT [Citation40].

Table 2. Radiological patterns and signs of fibrosis on chest HRCT [Citation40].

Bronchoalveolar lavage would be recommended depending on the clinical context; to rule out infections or to better approach a potential differential diagnosis (if relevant exposures for ILD development are present). Lung biopsy should be reserved for cases in which other conditions, such as hypersensitivity pneumonitis,pneumoconiosis, or malignancies, may be present in a patient with CTD [Citation17,Citation28,Citation32].

PFTs (FVC, TLC and DLCO), 6-minutes walking test (6MWT), and quality of life (QoL) questionnaires are used for monitoring ILD progression. HRCT is recommended during the follow-up in cases that present clinical and/or PFTs worsening, to differentiate disease progression versus acute exacerbation or other respiratory complications such as lung cancer or respiratory infection. In patients with advanced stages of their disease, or in cases with very severe DLCO decline and marked oxyhemoglobin desaturation with exercise, Doppler echocardiography is also useful to detect the development of associated PAH.

Although ILD is a complication described in many CTDs, the prevalence is higher in SSc and RA. The prevalence of ILD ranges from 30% to 60% in SSc, and progressive pulmonary fibrosis is the leading cause of death [Citation41]. The most prevalent fibrotic histologic pattern is nonspecific interstitial pneumonia (NSIP) [Citation2,Citation3,Citation42]. Systematic screening for ILD is recommended in all patients at diagnosis of SSc and, subsequently, on an annual basis during follow-up based on its frequency and mortality [Citation2,Citation3,Citation14,Citation20]. For screening, HRCT is recommended at diagnosis of the disease, since it has been shown that initial screening by PFT has a non-negligible percentage of false negatives [Citation43].

In RA, the prevalence of symptomatic ILD diagnosed by chest HRCT varies from 10% to 29%, being the second cause of death after cardiovascular complications [Citation4,Citation5,Citation16]. The radiological UIP pattern has been more frequently described than NSIP [Citation4,Citation5,Citation16]. Despite a frequent and serious complication, no recommendations on RA-ILD screening have been published until now. In order to respond to this clinical need, a joint collaboration between the Spanish societies of Rheumatology (SER) and Pneumology and Thoracic Surgery (SEPAR) have recently developed a multidisciplinary proposal of selective screening criteria shown in [Citation44]. The main risk factors for ILD development in SSc and RA are shown in .

Table 3. SER-SEPAR proposed screening criteria for diffuse interstitial lung disease in patients diagnosed with rheumatoid arthritis [Citation44].

Table 4. Main risk factors for ILD development in SSc and RA, and prognostic factors for mortality and progression of ILD.

Finally, ILD can be the initial clinical presentation of a CTD. For example, in RA-ILD, ILD precedes joint manifestations by months or years in up to 10–14% of patients [Citation45–47]. Therefore, a complete evaluation for the presence of a CTD, including serological autoimmunity study and physical exam, is recommended in all patients with new-onset ILD () [Citation48]. It is important to keep in mind that anti-Ro or SSA, anti-synthetase, and anti-Th/To antibodies may be positive, even if antinuclear antibodies are negative [Citation29–31].

Table 5. Panel of antibodies to be requested in the evaluation of an ILD to assess its possible association with a CTD.

Repeating the autoimmunity study during the follow-up of ILD patients is relevant because approximately 10–25% of cases that are initially idiopathic evolve into a CTD by clinical-onset or autoimmunity positivity (although there is no consensus on what should be the frequency of serial testing for CTD) [Citation49–51].

3. Progressive pulmonary fibrosis associated with CTD

Different definitions of progressive pulmonary fibrosis have been used, based on expert consensus, definitions used in clinical trials, and recently the definition published in the recent international guide, based on functional, clinical, and radiological parameters [Citation52]. The INBUILD randomized clinical trial [Citation53] used the following criteria to define progression in patients with fibrosing ILD: 1) a decrease in baseline FVC >10%; 2) a decrease in FVC between 5–10% with evidence of fibrosing progression on HRCT of the chest; 3) a decrease in FVC between 5–10% with worsening respiratory symptoms (dyspnea and dry cough); or 4) worsening dyspnea with progression of fibrosis on HRCT in the previous 24 months despite treatment. As a definition of progression, some experts also include a decrease in FVC between 5–10% with worsening DLCO >15% [Citation54].

PPF has been described in many CTDs [Citation13,Citation23], but this progressive feature is more frequent in SSc and RA. In SSc-ILD, 67% of patients will present pulmonary progression at 5 years. Of these, 32–33% have PPF [Citation20]. Different variables associated with an increased risk of progression in SSc-ILD have been identified and are summarized in [Citation55–57]. Two simple algorithms have also been proposed to stratify the prognosis of these patients (Goh et al. and the SPAR model), which are shown in [Citation58,Citation59].

Figure 1. Proposed algorithms to stratify the prognosis of patients with interstitial lung disease associated with systemic sclerosis (SSc-ILD). (a). Algorithm proposed by Goh et al. [Citation58] (b). SPAR model [Citation59] FVC, forced vital capacity; HRCT, high-resolution computed tomography.

Figure 1. Proposed algorithms to stratify the prognosis of patients with interstitial lung disease associated with systemic sclerosis (SSc-ILD). (a). Algorithm proposed by Goh et al. [Citation58] (b). SPAR model [Citation59] FVC, forced vital capacity; HRCT, high-resolution computed tomography.

In RA-ILD, 53% of patients will progress, with an estimated 40% of patients meeting criteria for progressive fibrosing RA-ILD at 5 years of evolution [Citation13,Citation23,Citation60]. The main prognostic factors for pulmonary progression and mortality in RA-ILD are also shown in [Citation61]. The usefulness of the gender, age, physiology (GAP) and composite physiological index (CPI) in predicting the risk of mortality in these patients has been demonstrated [Citation21,Citation62,Citation63].

Knowledge of the prognostic factors in these two entities (SSc-ILD and RA-ILD) allows early identification of patients with a potentially higher severity profile who are candidates for intensive targeted treatment as soon as the diagnosis of ILD is established and for close clinical follow-up.

4. Treatment and follow-up

The treatment of CTD-ILD should be individualized and comprehensive and should be planned considering the underlying CTD, the patient’s comorbidities, and the severity of extrapulmonary manifestations. In any case, the choice of treatment should always seek a balance between the preservation of pulmonary function and the risk of adverse events.

So far, low-dose glucocorticoids are usually used as initial treatment in monotherapy or in combination with immunosuppressants agents. There are no randomized controlled trials (RCTs) that have evaluated the efficacy of glucocorticoids in CTD-ILD and the evidence supporting their use is based primarily on clinical experience and real-life data. Glucocorticoids should be used with caution in SSc since their use, especially at medium or high doses, is an independent risk factor for the development of scleroderma renal crisis [Citation64]. The efficacy of glucocorticoids in fibrotic patterns (UIP and fibrosing NSIP) is inconclusive. On the contrary, there is accumulating evidence about the detrimental effect of steroids as treatment of PPF [Citation65,Citation66].

Cytotoxic immunosuppressive drugs that are commonly used to treat CTD and CTD-ILD include cyclophosphamide, mycophenolate, azathioprine, calcineurin inhibitors (especially in ILD associated with inflammatory myopathies) and leflunomide (in RA). Despite their use in all CTDs, their efficacy has only been demonstrated in RCTs in the case of cyclophosphamide and mycophenolate for the treatment of SSc-ILD [Citation67,Citation68], with none of them having a therapeutic indication.

Methotrexate has classically been discouraged in patients with RA-ILD because of the risk of causing acute pneumonitis. However, the literature confirms that the actual risk of drug-induced pneumonitis is very low (0.3%) [Citation69–71]. There is no confirmation that methotrexate increases the risk of developing ILD in patients with RA, but recent studies suggest that RA control achieved with this drug is associated with a delay in the onset of ILD and a better prognosis [Citation36,Citation72–76].

In case of ILD progression, despite initial treatment with glucocorticoids and immunosuppressants, there are several options: rescue therapy with biologic drugs (rituximab, tocilizumab or abatacept) particularly in ILDs with inflammatory features or patterns. Antifibrotic treatment can be used in CTD-ILD patients with PPF despite previous management . In SSc, the option of autologous hematopoietic stem cell transplantation is also available, although its effect on the lung remains controversial [Citation77–80].

Regarding evidence for the efficacy of biologic agents in the treatment of CTD-ILD, only tocilizumab has confirmed effectiveness in RCTs, specifically in SSc-ILD [Citation81–83]. Although the trials only included patients with mild-to-moderate ILD, tocilizumab has recently been approved by the Food and Drug Administration (FDA) for the treatment of SSc-ILD. There are several ongoing RCTs with rituximab and abatacept (RECITAL and EVER-ILD in SSc-ILD, and APRIL in RA-ILD) [Citation84–86]; currently, however, their use is only supported by real-life observational studies demonstrating their usefulness in stabilizing or improving lung function, particularly in patients with a non-UIP radiological pattern, including cases in which ILD worsened despite previous treatment with glucocorticoids and classic synthetic disease-modifying antirheumatic drugs (DMARDs) or immunosuppressants [Citation87–89].

There are currently two antifibrotic agents on the market: nintedanib and pirfenidone. At present, only nintedanib has been approved by the FDA and the European Medicines Agency (EMA) for the treatment of fibrosing SSc-ILD, based on data from the phase III RCT SENSCIS [Citation90], and for the treatment of other progressive fibrosing ILD, including CTD-ILD, based on results from the phase III RCT INBUILD [Citation53].

Pirfenidone does not have data in CTD-ILD with fibrosis and a progressive phenotype but only in a few disease entities that are under the designation of CTD. The published experience with pirfenidone in CTD-ILD is so far limited to two phase II RCTs (LOTUSS and RELIEF) [Citation91,Citation92]. Ongoing studies include a phase III RCT in SSc-ILD (Scleroderma lung study III) [Citation93] and a phase II in RA-ILD (TRAIL 1) [Citation94].

Equally relevant non-pharmacological measures within the comprehensive approach include smoking cessation, respiratory rehabilitation, prevention and treatment of gastroesophageal reflux, systematic vaccination against respiratory pathogens such as influenza, pneumococcus and SARS-CoV-2, adequate and early treatment of coexisting respiratory infections, psychosocial support, and long-term home oxygen therapy in the presence of chronic respiratory failure.

When PPF does not show stabilization, despite the measures taken, lung transplantation should always be considered in candidate patients and the case should be referred to Lung Transplant Units before reaching an advanced stage of respiratory involvement.

5. Conclusion

CTD-ILD especially those with progressive pulmonary fibrosis have a high morbidity and mortality rate. In order to reverse this situation and improve the prognosis and QoL of patients, a multidisciplinary diagnostic and therapeutic approach is necessary as the best model of care to guarantee early diagnosis and comprehensive and individualized treatment. Antifibrotic drugs are considered for those presenting progressive pulmonary fibrosis.

6. Expert opinion

There are still unmet needs in PPF-CTD, including the need for reliable prognostic biomarkers for early identification and tools to select the most appropriate comprehensive treatment strategy for each patient. It is desirable that, in the coming years, through the identification of biomarkers, genetic polymorphisms and other molecular classification techniques, progress will be made in personalized precision medicine. There are also high expectations for new drugs that could expand the therapeutic arsenal for PPF, such as JAK kinase inhibitors, pentraxin 2, pamrevlumab, and autotaxin inhibitors [Citation95–99].

It is also essential to know the long-term safety and efficacy of combined immunosuppressant and antifibrotic therapy, and to have methods for evaluating the response to treatment that provide a more comprehensive and multifaceted vision, including patient-reported outcomes, because patients should be increasingly empowered in decision-making.

Another area for improvement is the diagnostic delay experienced by these patients, which leads to a delay in treatment initiation. The origin of this delay is usually multifactorial (difficulty of access to an expert multidisciplinary team, waiting lists in the referral circuits, and in the performance of complementary examinations such as HRCT or PFTs, and failure to detect respiratory symptoms or signs early), so its solution seems difficult, since it implies better planning of resources and training programs for clinicians who care for these patients, whether in primary or specialized care.

Article highlights

  • Interstitial lung disease (ILD) is a frequent and severe complication of connective tissue diseases (CTD), such as systemic sclerosis and rheumatoid arthritis.

  • The diagnosis of fibrotic ILD in CTDs, especially the early identification of patients with progressive pulmonary fibrosis, is complex, requiring a comprehensive assessment, including accurate staging of the disease severity and extrapulmonary manifestations.

  • An individualized and comprehensive therapeutic approach is important in the treatment of ILD associated with CTD to control the autoimmune and inflammatory activity of the underlying disease and to prevent, when necessary, the progression of fibrotic pulmonary changes.

  • A multidisciplinary diagnostic and therapeutic approach is required to not only guarantee early diagnosis but also provide comprehensive and individualized treatment.

  • Antifibrotic drugs, such as nintedanib, have been shown in recent clinical trials to attenuate progressive pulmonary fibrosis in patients with CTD and may provide therapeutic benefit.

Conflict of interests

Boehringer Ingelheim had the opportunity to review the content of the manuscript for medical and scientific accuracy, as well as intellectual property considerations.

The authors meet the authorship criteria recommended by the International Committee of Medical Journal Editors (ICMJE) and did not receive payment related to the development of this article.

M. Molina-Molina has received research and conference presentation support from Roche, Boehringer Ingelheim, Esteve-Teijin, Chiesi, Galapagos, Astra-Zeneca, and Pfizer.

I. Castellví has received support for research and presentation at congresses from Roche, Boehringer Ingelheim, Bristol Myers-Squibb, and Janssen-Cilag.

C. Valenzuela has received research and presentation support from Roche, Boehringer Ingelheim, and Bristol Myers Squibb.

J. Ramírez has received research and presentation support from Roche, Boehringer Ingelheim, and Pfizer.

J.A. Rodríguez-Portal has received research and presentation support from Roche, Boehringer Ingelheim, Janssen, and Bristol Myers Squibb.

T. Franquet has received research and presentation support from Roche and Boehringer Ingelheim.

J. Narváez has received funding for meetings and congresses and course attendance from Roche, Bristol Myers Squibb, Kern, Pfizer, Lilly, and Sanofi; speaking fees from Roche, Bristol Myers Squibb, Lilly, AbbVie, Pfizer, Boehringer Ingelheim, Sanofi, GSK, Gebro pharma, Kern, and Sobi; and consulting for a pharmaceutical company/other technologies from Roche, Bristol Myers Squibb, Lilly, AbbVie, Pfizer, Boehringer Ingelheim, Sanofi, GSK, Gebro pharma, Kern, and Sobi.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Geolocation information

This article has been developed by experts from different cities in Spain: L’Hospitalet de Llobregat, Barcelona, Madrid, and Seville.

Acknowledgments

The authors would like to thank Fernando Sánchez Barbero PhD and Content Ed Net (Madrid, Spain) for their help in the preparation of this manuscript.

Additional information

Funding

Boehringer Ingelheim financed the writing services and editorial process.

References

  • Atzeni F, Gerardi MC, Barilaro G, et al. Interstitial lung disease in systemic autoimmune rheumatic diseases: a comprehensive review. Expert Rev Clin Immunol. 2018 Jan;14(1):69–82.
  • Perelas A, Silver RM, Arrossi AV, et al. Systemic sclerosis-associated interstitial lung disease. Lancet Respir Med. 2020 Mar;8(3):304–320.
  • Castelino FV, Dellaripa PF. Recent progress in systemic sclerosis-interstitial lung disease. Curr Opin Rheumatol. 2018 Nov;30(6):570–575.
  • Kadura S, Raghu G. Rheumatoid arthritis-interstitial lung disease: manifestations and current concepts in pathogenesis and management. Eur Respir Rev. 2021 Jun;30;30(160):210011.
  • Spagnolo P, Lee JS, Sverzellati N, et al. The lung in rheumatoid arthritis: focus on interstitial lung disease. Arthritis Rheumatol. 2018 Oct;70(10):1544–1554.
  • Nombel A, Fabien N, Coutant F. Dermatomyositis with anti-MDA5 antibodies: bioclinical features, pathogenesis and emerging therapies. Front Immunol. 2021;12:773352.
  • Selva-O’Callaghan A, Pinal-Fernández I, Trallero-Araguas E, et al. Classification and management of adult inflammatory myopathies. Lancet Neurol. 2018 Sep;17(9):816–828.
  • Opinc AH, Makowska JS. Antisynthetase syndrome - much more than just a myopathy. Semin Arthritis Rheum. 2021 Feb;51(1):72–83.
  • Barba T, Mainbourg S, Nasser M, et al. Lung diseases in inflammatory myopathies. Semin Respir Crit Care Med. 2019 Apr;40(2):255–270.
  • Lee CT, Strek ME. The other connective tissue disease-associated interstitial lung diseases: sjogren’s syndrome, mixed connective tissue disease, and systemic lupus erythematosus. Curr Opin Pulm Med. 2021 Sep 1;27(5):388–395.
  • Narváez J, Borrell H, Sánchez-Alonso F, et al. Primary respiratory disease in patients with systemic lupus erythematosus: data from the Spanish rheumatology society lupus registry (RELESSER) cohort. Arthritis Res Ther. 2018 Dec 19;20(1):280.
  • Sebastiani M, Manfredi A, Vacchi C, et al. Epidemiology and management of interstitial lung disease in ANCA-associated vasculitis. Clin Exp Rheumatol. 2020Mar-Apr; 124(2):221–231. 38 Suppl
  • Spagnolo P, Distler O, Ryerson CJ, et al. Mechanisms of progressive fibrosis in connective tissue disease (CTD)-associated interstitial lung diseases (ILDs). Ann Rheum Dis. 2021 Feb;80(2):143–150.
  • Cappelli S, Bellando Randone S, Camiciottoli G, et al. Interstitial lung disease in systemic sclerosis: where do we stand? Eur Respir Rev. 2015 Sep;24(137):411–419.
  • Herzog EL, Mathur A, Tager AM, et al. Review: interstitial lung disease associated with systemic sclerosis and idiopathic pulmonary fibrosis: how similar and distinct? Arthritis Rheumatol. 2014 Aug;66(8):1967–1978.
  • Johnson C. Recent advances in the pathogenesis, prediction, and management of rheumatoid arthritis-associated interstitial lung disease. Curr Opin Rheumatol. 2017 May;29(3):254–259.
  • Wijsenbeek M, Cottin V. Spectrum of fibrotic lung diseases. N Engl J Med. 2020 Sep 3;383(10):958–968.
  • Christmann RB, Sampaio-Barros P, Stifano G, et al. Association of interferon- and transforming growth factor beta-regulated genes and macrophage activation with systemic sclerosis-related progressive lung fibrosis. Arthritis Rheumatol. 2014 Mar;66(3):714–725.
  • Zhu J, Tu S, Qu Q. lncRNA BZRAP1-AS1 alleviates rheumatoid arthritis by regulating miR-1286/COL5A2 axis. Immun Inflamm Dis. 2022 Feb;10(2):163–174.
  • Hoffmann-Vold AM, Allanore Y, Alves M, et al. Progressive interstitial lung disease in patients with systemic sclerosis-associated interstitial lung disease in the EUSTAR database. Ann Rheum Dis. 2021 Feb;80(2):219–227.
  • Zamora-Legoff JA, Krause ML, Crowson CS, et al. Patterns of interstitial lung disease and mortality in rheumatoid arthritis. Rheumatology (Oxford). 2017 Mar 1;56(3):344–350.
  • Hyldgaard C, Ellingsen T, Hilberg O, et al. Rheumatoid arthritis-associated interstitial lung disease: clinical characteristics and predictors of mortality. Respiration. 2019;98(5):455–460.
  • Olson A, Hartmann N, Patnaik P, et al. Estimation of the prevalence of progressive fibrosing interstitial lung diseases: systematic literature review and data from a physician survey. Adv Ther. 2021 Feb;38(2):854–867.
  • Molyneaux PL, Cox MJ, Willis-Owen SA, et al. The role of bacteria in the pathogenesis and progression of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2014 Oct 15;190(8):906–913.
  • Papiris SA, Manali ED, Kolilekas L, et al. Acute respiratory events in connective tissue disorders. Respiration. 2016;91(3):181–201.
  • Thomas K, Lazarini A, Kaltsonoudis E, et al. Incidence, risk factors and validation of the RABBIT score for serious infections in a cohort of 1557 patients with rheumatoid arthritis. Rheumatology (Oxford). 2021 May 14;60(5):2223–2230.
  • Izumi Y, Akazawa M, Akeda Y, et al. The 23-valent pneumococcal polysaccharide vaccine in patients with rheumatoid arthritis: a double-blinded, randomized, placebo-controlled trial. Arthritis Res Ther. 2017 Jan 25;19(1):15.
  • Cottin V, Hirani NA, Hotchkin DL, et al. Presentation, diagnosis and clinical course of the spectrum of progressive-fibrosing interstitial lung diseases. Eur Respir Rev. 2018 Dec 31;27(150):180076.
  • Wells A, Devaraj A, Renzoni EA, et al. Multidisciplinary evaluation in patients with lung disease associated with connective tissue disease. Semin Respir Crit Care Med. 2019 Apr;40(2):184–193.
  • Sebastiani M, Faverio P, Manfredi A, et al. Interstitial pneumonia with autoimmune features: why rheumatologist-pulmonologist collaboration is essential. Biomedicines. 2020 Dec 26;9(1):17.
  • Geerts S, Wuyts W, Langhe E, et al. Connective tissue disease associated interstitial pneumonia: a challenge for both rheumatologists and pulmonologists. Sarcoidosis Vasc Diffuse Lung Dis. 2017;34(4):326–335.
  • Mathai SC, Danoff SK. Management of interstitial lung disease associated with connective tissue disease. BMJ. 2016 Feb 24;352:h6819
  • De Sadeleer LJ, Meert C, Yserbyt J, et al. Diagnostic ability of a dynamic multidisciplinary discussion in interstitial lung diseases: a retrospective observational study of 938 cases. Chest. 2018 Jun;153(6):1416–1423.
  • Ryerson CJ, Urbania TH, Richeldi L, et al. Prevalence and prognosis of unclassifiable interstitial lung disease. Eur Respir J. 2013 Sep;42(3):750–757.
  • Fischer A, Antoniou KM, Brown KK, et al. An official European respiratory society/American thoracic society research statement: interstitial pneumonia with autoimmune features. Eur Respir J. 2015 Oct;46(4):976–987.
  • Cano-Jiménez E, Vázquez Rodríguez T, Martín-Robles I, et al. Diagnostic delay of associated interstitial lung disease increases mortality in rheumatoid arthritis. Sci Rep. 2021 Apr 28;11(1):9184.
  • Robles-Pérez A, Luburich P, Bolivar S, et al. A prospective study of lung disease in a cohort of early rheumatoid arthritis patients. Sci Rep. 2020 Sep 24;10(1):15640.
  • Sgalla G, Walsh SLF, Sverzellati N, et al. “Velcro-type” crackles predict specific radiologic features of fibrotic interstitial lung disease. BMC Pulm Med. 2018 Jun 18;18(1):103.
  • Kondoh Y, Makino S, Ogura T, et al. 2020 guide for the diagnosis and treatment of interstitial lung disease associated with connective tissue disease. Respir Investig. 2021 Nov;59(6):709–740.
  • Giménez Palleiro A, Franquet T. Radiological patterns in interstitial lung disease. Semin Fund Esp Reumatol. 2013;14(4):97–105.
  • Tyndall AJ, Bannert B, Vonk M, et al. Causes and risk factors for death in systemic sclerosis: a study from the EULAR scleroderma trials and research (EUSTAR) database. Ann Rheum Dis. 2010 Oct;69(10):1809–1815.
  • Bouros D, Wells AU, Nicholson AG, et al. Histopathologic subsets of fibrosing alveolitis in patients with systemic sclerosis and their relationship to outcome. Am J Respir Crit Care Med. 2002 Jun 15;165(12):1581–1586.
  • Suliman YA, Dobrota R, Huscher D, et al. Brief report: pulmonary function tests: high rate of false-negative results in the early detection and screening of scleroderma-related interstitial lung disease. Arthritis Rheumatol. 2015 Dec;67(12):3256–3261.
  • Narváez J, Aburto M, Seoane-Mato D, et al. Criterios de cribado de la enfermedad pulmonar intersticial difusa asociada a la artritis reumatoide: propuesta de expertos basada en metodología Delphi. Reumatol Clin. 2022. [en prensa].
  • Hyldgaard C, Hilberg O, Pedersen AB, et al. A population-based cohort study of rheumatoid arthritis-associated interstitial lung disease: comorbidity and mortality. Ann Rheum Dis. 2017 Oct;76(10):1700–1706.
  • Gizinski AM, Mascolo M, Loucks JL, et al. Rheumatoid arthritis (RA)-specific autoantibodies in patients with interstitial lung disease and absence of clinically apparent articular RA. Clin Rheumatol. 2009 May;28(5):611–613.
  • Singh K, Al-Sadawi M, Ortega RR, et al. Interstitial lung disease as the initial manifestation of rheumatoid arthritis: a case report and review of the literature. Am J Med Case Rep. 2019;7(12):342–347.
  • Raghu G, Remy-Jardin M, Myers JL, et al. Diagnosis of idiopathic pulmonary fibrosis. an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2018 Sep 1;198(5):e44–e68.
  • Hu Y, Wang LS, Wei YR, et al. Clinical characteristics of connective tissue disease-associated interstitial lung disease in 1,044 Chinese patients. Chest. 2016 Jan;149(1):201–208.
  • Ando M, Miyazaki E, Ishii T, et al. Incidence of myeloperoxidase anti-neutrophil cytoplasmic antibody positivity and microscopic polyangitis in the course of idiopathic pulmonary fibrosis. Respir Med. 2013 Apr;107(4):608–615.
  • Kagiyama N, Takayanagi N, Kanauchi T, et al. Antineutrophil cytoplasmic antibody-positive conversion and microscopic polyangiitis development in patients with idiopathic pulmonary fibrosis. BMJ Open Respir Res. 2015;2(1):e000058.
  • Raghu G, Remy-Jardin M, Richeldi L, et al. Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2022 May 1;205(9):e18–e47.
  • Flaherty KR, Wells AU, Cottin V, et al. Nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med. 2019 Oct 31;381(18):1718–1727.
  • George PM, Spagnolo P, Kreuter M, et al. Progressive fibrosing interstitial lung disease: clinical uncertainties, consensus recommendations, and research priorities. Lancet Respir Med. 2020 Sep;8(9):925–934.
  • Vonk MC, Walker UA, Volkmann ER, et al. Natural variability in the disease course of SSc-ILD: implications for treatment. Eur Respir Rev. 2021 Mar 31;30(159):200340.
  • Volkmann ER. Natural history of systemic sclerosis-related interstitial lung disease: how to identify a progressive fibrosing phenotype. J Scleroderma Relat Disord. 2020 Mar;5(2 Suppl):31–40.
  • Distler O, Assassi S, Cottin V, et al. Predictors of progression in systemic sclerosis patients with interstitial lung disease. Eur Respir J. 2020 May;55(5):1902026.
  • Goh NS, Desai SR, Veeraraghavan S, et al. Interstitial lung disease in systemic sclerosis: a simple staging system. Am J Respir Crit Care Med. 2008 Jun 1;177(11):1248–1254.
  • Wu W, Jordan S, Becker MO, et al. Prediction of progression of interstitial lung disease in patients with systemic sclerosis: the SPAR model. Ann Rheum Dis. 2018 Sep;77(9):1326–1332.
  • Zamora-Legoff JA, Krause ML, Crowson CS, et al. Progressive decline of lung function in rheumatoid arthritis-associated interstitial lung disease. Arthritis Rheumatol. 2017 Mar;69(3):542–549.
  • Rodríguez Portal JA, Brito García N, Díaz Del Campo Fontecha P, et al. Recomendaciones SER-SEPAR para el manejo de la enfermedad pulmonar intersticial difusa asociada a la artritis reumatoide. Parte 1: epidemiología, factores de riesgo y pronóstico. Reumatol Clin. 2022; [en prensa].
  • Morisset J, Vittinghoff E, Lee BY, et al. The performance of the GAP model in patients with rheumatoid arthritis associated interstitial lung disease. Respir Med. 2017 Jun;127:51–56.
  • Nurmi HM, Purokivi MK, Karkkainen MS, et al. Are risk predicting models useful for estimating survival of patients with rheumatoid arthritis-associated interstitial lung disease? BMC Pulm Med. 2017 Jan 13;17(1):16.
  • Trang G, Steele R, Baron M, et al. Corticosteroids and the risk of scleroderma renal crisis: a systematic review. Rheumatol Int. 2012 Mar;32(3):645–653.
  • Papiris SA, Kagouridis K, Papadaki G, et al. Treating CTDs related fibrotic ILDs by immunosuppressants: “facts and faults”. Lung. 2014 Apr;192(2):221–223.
  • Papiris SA, Kolilekas L, Kagouridis K, et al. ipf-acute exacerbations: advances and future perspectives. Front Pharmacol. 2022;13:836553.
  • Tashkin DP, Elashoff R, Clements PJ, et al. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med. 2006 Jun 22;354(25):2655–2666.
  • Tashkin DP, Roth MD, Clements PJ, et al. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial. Lancet Respir Med. 2016 Sep;4(9):708–719.
  • Solomon DH, Glynn RJ, Karlson EW, et al. Adverse effects of low-dose methotrexate: a randomized trial. Ann Intern Med. 2020 Mar 17;172(6):369–380.
  • Sparks JA, Dellaripa PF, Glynn RJ, et al. Pulmonary adverse events in patients receiving low-dose methotrexate in the randomized, double-blind, placebo-controlled cardiovascular inflammation reduction trial. Arthritis Rheumatol. 2020 Dec;72(12):2065–2071.
  • Ridker PM, Everett BM, Pradhan A, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 2019 Feb 21;380(8):752–762.
  • Narváez J, Díaz Del Campo Fontecha P, Brito García N, et al. Recomendaciones SER-SEPAR para el manejo de la enfermedad pulmonar intersticial difusa asociada a la artritis reumatoide. Parte 2: tratamiento. Reumatol Clin. 2022; [en prensa]
  • Juge PA, Lee JS, Lau J, et al. Methotrexate and rheumatoid arthritis associated interstitial lung disease. Eur Respir J. 2021 Feb;57(2):2000337.
  • Ibfelt EH, Jacobsen RK, Kopp TI, et al. Methotrexate and risk of interstitial lung disease and respiratory failure in rheumatoid arthritis: a nationwide population-based study. Rheumatology (Oxford). 2021 Jan 5;60(1):346–352.
  • Kiely P, Busby AD, Nikiphorou E, et al. Is incident rheumatoid arthritis interstitial lung disease associated with methotrexate treatment? Results from a multivariate analysis in the ERAS and ERAN inception cohorts. BMJ Open. 2019 May 5;9(5):e028466.
  • Rojas-Serrano J, Herrera-Bringas D, Pérez-Roman DI, et al. Rheumatoid arthritis-related interstitial lung disease (RA-ILD): methotrexate and the severity of lung disease are associated to prognosis. Clin Rheumatol. 2017 July;36(7):1493–1500.
  • van Laar JM, Farge D, Sont JK, et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA. 2014 Jun 25;311(24):2490–2498.
  • Burt RK, Shah SJ, Dill K, et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an open-label, randomised phase 2 trial. Lancet. 2011 Aug 6;378(9790):498–506.
  • Sullivan KM, Goldmuntz EA, Keyes-Elstein L, et al. Myeloablative autologous stem-cell transplantation for severe scleroderma. N Engl J Med. 2018 Jan 4;378(1):35–47.
  • Del Papa N, Onida F, Zaccara E, et al. Autologous hematopoietic stem cell transplantation has better outcomes than conventional therapies in patients with rapidly progressive systemic sclerosis. Bone Marrow Transplant. 2017 Jan;52(1):53–58.
  • Roofeh D, Lin CJF, Goldin J, et al. Tocilizumab prevents progression of early systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol. 2021 July;73(7):1301–1310.
  • Khanna D, Lin CJF, Furst DE, et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2020 Oct;8(10):963–974.
  • Crespo MM, Lease ED, Sole A, et al. ISHLT consensus document on lung transplantation in patients with connective tissue disease: part I: epidemiology, assessment of extrapulmonary conditions, candidate evaluation, selection criteria, and pathology statements. J Heart Lung Transplant. 2021 Nov;40(11):1251–1266.
  • Saunders P, Tsipouri V, Keir GJ, et al. Rituximab versus cyclophosphamide for the treatment of connective tissue disease-associated interstitial lung disease (RECITAL): study protocol for a randomised controlled trial. Trials. 2017 Jun 15;18(1):275.
  • Evaluation of efficacy and safety of rituximab with mycophenolate mofetil in patients with interstitial lung disease (EvER-ILD) [cited 2022 24 enero]. Available from: https://clinicaltrials.gov/ct2/show/NCT02990286
  • April (Abatacept in Rheumatoid arthritis-ILD) [cited 2022 24 enero]. Available from: https://clinicaltrials.gov/ct2/show/NCT03084419
  • Narváez J, Luch J, Molina-Molina M, et al. Rituximab as a rescue treatment added on mycophenolate mofetil background therapy in progressive systemic sclerosis associated interstitial lung disease unresponsive to conventional immunosuppression. Semin Arthritis Rheum. 2020 Oct;50(5):977–987.
  • Narváez J, Robles-Pérez A, Molina-Molina M, et al. Real-world clinical effectiveness of rituximab rescue therapy in patients with progressive rheumatoid arthritis-related interstitial lung disease. Semin Arthritis Rheum. 2020 Oct;50(5):902–910.
  • Vicente-Rabaneda EF, Atienza-Mateo B, Blanco R, et al. Efficacy and safety of abatacept in interstitial lung disease of rheumatoid arthritis: a systematic literature review. Autoimmun Rev. 2021 Jun;20(6):102830.
  • Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J Med. 2019 Jun 27;380(26):2518–2528.
  • Khanna D, Albera C, Fischer A, et al. An open-label, phase II study of the safety and tolerability of pirfenidone in patients with scleroderma-associated interstitial lung disease: the LOTUSS trial. J Rheumatol. 2016 Sep;43(9):1672–1679.
  • Behr J, Prasse A, Kreuter M, et al. Pirfenidone in patients with progressive fibrotic interstitial lung diseases other than idiopathic pulmonary fibrosis (RELIEF): a double-blind, randomised, placebo-controlled, phase 2b trial. Lancet Respir Med. 2021 May;9(5):476–486.
  • Scleroderma Lung Study III - Combining Pirfenidone with Mycophenolate (SLSIII) [cited 2022 24 enero]. Available from: https://clinicaltrials.gov/ct2/show/NCT03221257
  • Phase ll Study of Pirfenidone in Patients with RAILD (TRAIL1) [cited 2022 24 enero]. Available from: https://clinicaltrials.gov/ct2/show/NCT02808871
  • Talotta R. The rationale for targeting the JAK/STAT pathway in scleroderma-associated interstitial lung disease. Immunotherapy. 2021 Feb;13(3):241–256.
  • Raghu G, van den Blink B, Hamblin MJ, et al. Effect of recombinant human pentraxin 2 vs placebo on change in forced vital capacity in patients with idiopathic pulmonary fibrosis: a randomized clinical trial. JAMA. 2018 Jun 12;319(22):2299–2307.
  • Raghu G, van den Blink B, Hamblin MJ, et al. Long-term treatment with recombinant human pentraxin 2 protein in patients with idiopathic pulmonary fibrosis: an open-label extension study. Lancet Respir Med. 2019 Aug;7(8):657–664.
  • Richeldi L, Fernández Pérez ER, Costabel U, et al. Pamrevlumab, an anti-connective tissue growth factor therapy, for idiopathic pulmonary fibrosis (PRAISE): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2020 Jan;8(1):25–33.
  • Maher TM, van der Aar EM, van de Steen O, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of GLPG1690, a novel autotaxin inhibitor, to treat idiopathic pulmonary fibrosis (FLORA): a phase 2a randomised placebo-controlled trial. Lancet Respir Med. 2018 Aug;6(8):627–635.