Next Article in Journal
Multimodal Long-Term Predictors of Outcome in Out of Hospital Cardiac Arrest Patients Treated with Targeted Temperature Management at 36 °C
Next Article in Special Issue
Telomeres in Interstitial Lung Disease
Previous Article in Journal
A Novel Combination of Accelerometry and Ecological Momentary Assessment for Post-Stroke Paretic Arm/Hand Use: Feasibility and Validity
Previous Article in Special Issue
Evaluation of Regional Pulmonary Ventilation in Spontaneously Breathing Patients with Idiopathic Pulmonary Fibrosis (IPF) Employing Electrical Impedance Tomography (EIT): A Pilot Study from the European IPF Registry (eurIPFreg)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progression in the Management of Non-Idiopathic Pulmonary Fibrosis Interstitial Lung Diseases, Where Are We Now and Where We Would Like to Be

1
Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, B-3000 Leuven, Belgium
2
Department of Respiratory Diseases, University Hospitals Leuven, B-3000 Leuven, Belgium
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(6), 1330; https://doi.org/10.3390/jcm10061330
Submission received: 22 January 2021 / Revised: 5 March 2021 / Accepted: 15 March 2021 / Published: 23 March 2021
(This article belongs to the Special Issue The New Perspective in Pulmonary Fibrosis)

Abstract

:
A significant proportion of patients with interstitial lung disease (ILD) may develop a progressive fibrosing phenotype characterized by worsening of symptoms and pulmonary function, progressive fibrosis on chest computed tomography and increased mortality. The clinical course in these patients mimics the relentless progressiveness of idiopathic pulmonary fibrosis (IPF). Common pathophysiological mechanisms such as a shared genetic susceptibility and a common downstream pathway—self-sustaining fibroproliferation—support the concept of a progressive fibrosing phenotype, which is applicable to a broad range of non-IPF ILDs. While antifibrotic drugs became the standard of care in IPF, immunosuppressive agents are still the mainstay of treatment in non-IPF fibrosing ILD (F-ILD). However, recently, randomized placebo-controlled trials have demonstrated the efficacy and safety of antifibrotic treatment in systemic sclerosis-associated F-ILD and a broad range of F-ILDs with a progressive phenotype. This review summarizes the current pharmacological management and highlights the unmet needs in patients with non-IPF ILD.

1. Introduction

In addition to idiopathic pulmonary fibrosis (IPF), which is defined as a relentless progressive disease, a significant proportion of patients with other interstitial lung disease (ILD) subtypes may develop a progressive phenotype characterized by worsening of symptoms, deterioration of pulmonary function, progressive pulmonary fibrosis on chest computed tomography (CT) and increased mortality. Other non-IPF ILD subtypes that may be associated with a progressive fibrosing phenotype include connective tissue disease-associated ILD (CTD-ILD), idiopathic non-specific interstitial pneumonia (iNSIP), sarcoidosis, hypersensitivity pneumonitis (HP), occupational ILD and unclassifiable ILD (uILD) [1,2,3,4]. Based on survey data and cohort studies, it is estimated that 30% of non-IPF ILD patients will develop a progressive fibrosing phenotype [5,6,7]. In this review, we summarize the current pharmacological management and highlight the unmet needs in patients with non-IPF ILD.

2. Historical Context

For decades, the mainstay of treatment in IPF was immunosuppressive therapy, based on the hypothesis that inflammation leads to injury and fibrosis [8]. Thereafter, it was thought that an oxidant–antioxidant imbalance may contribute to this process of fibrosis in IPF, leading to the IFIGENIA-trial in 2005 [9]. The major merit of this trial was that it made the field realize that larger randomized controlled trials (RCTs) in ILD were possible. The management of IPF changed drastically in 2012, as the PANTHER-IPF trial showed that immunosuppressive treatment resulted in increased mortality [10]. Therefore, it became clear that immunosuppressive agents provided no benefit and, on the contrary, were harmful in IPF patients. This observation supported the new hypothesis that IPF was not the result of an exaggerated immune response but should be considered as an aberrant progressive self-sustaining fibrotic process initiated by repeated epithelial injury without an identifiable cause [8]. Indeed, two antifibrotic drugs, pirfenidone and nintedanib, were shown to be effective in IPF by reducing forced vital capacity (FVC) decline and are currently recommended for the treatment of IPF [11,12,13,14].
While antifibrotic drugs became the standard of care in IPF, immunosuppressive agents are still the mainstay of treatment in non-IPF fibrosing ILD (F-ILD). However, it became clear that a subset of patients with non-IPF F-ILD continued to have progressive fibrosis despite conventional immunosuppressive treatment and had an IPF-like outcome [1,15,16,17,18]. Additionally, from a pathophysiological perspective, studies have reported shared mechanisms between IPF and non-IPF ILD, such as a shared genetic susceptibility [19,20,21,22]. Furthermore, the idea grew that when F-ILD has reached a stage of fibrosis that becomes self-sustaining, antifibrotic treatment could be an effective treatment to slow down disease progression. Because of the similarities in disease behavior and pathophysiological mechanisms with a common downstream pathobiological pathway, the non-IPF progressive F-ILDs (PF-ILDs) have been lumped together, and recently, RCTs also demonstrated the efficacy of antifibrotic treatment in systemic sclerosis-associated F-ILD and a broad range of F-ILDs with a progressive phenotype. [23,24,25,26].

3. Immunosuppressive Treatment: Where Are We?

3.1. Connective Tissue Disease-Associated ILD (CTD-ILD)

Patients with Sjogren’s syndrome, systemic lupus erythematosus, mixed connective tissue disease, polymyositis and dermatomyositis, rheumatoid arthritis and systemic sclerosis are at risk of developing ILD. Until recently, RCTs have only been performed in patients with systemic sclerosis associated-ILD (SSc-ILD), and evidence from these clinical trials has been extrapolated to other CTDs.

3.1.1. Systemic Sclerosis-Associated ILD (SSc-ILD)

ILD is a common complication in patients with systemic sclerosis and is the leading cause of disease-related deaths [27]. The extent of fibrosis on chest CT is highly predictive of mortality: in a study by Goh et al., patients with extensive disease (extent of fibrosis on CT >30% or 10–30% plus FVC <70% predicted) had a three-fold risk of mortality (hazard ratio (HR) 3.46) compared to patients with limited disease [28]. In 2016, the EULAR (European League against Rheumatism) recommended treatment with cyclophosphamide (CYC) in SSc-ILD as the Scleroderma Lung Study (SLS)-I and Fibrosing Alveolitis in Scleroderma Trial (FAST) showed a beneficial but modest effect of CYC on FVC decline compared to placebo (FVC change in SLS-I: placebo −2.6%, CYC −1.0%; FAST: placebo −3.0%, CYC +2.4%) [29,30,31]. As the treatment effect was mainly due to the prevention of progression and the efficacy of CYC was lost after 24 months, EULAR recommended the use of CYC in particular in patients with progressive disease [31,32]. In the SLS-II study, similar efficacy was seen with two years of mycophenolate mofetil (MMF) compared to one year of oral CYC, but MMF seemed to be better tolerated, especially with less hematological toxicity [33]. Both MMF and CYC resulted in improvements over 24 months in FVC% predicted (FVC%pred). MMF is now considered a first-line therapy for patients with SSc-ILD. Based on observational studies, azathioprine (AZA) can be considered as an alternative treatment when MMF is poorly tolerated [34,35,36,37]. Rituximab, an anti-CD20 monoclonal antibody, is not routinely used as first-line treatment but can be considered in SSc-ILD patients who are progressive despite standard treatment. In a randomized open-label study, rituximab was more efficacious than CYC (change in FVC%pred: rituximab +6.2%, CYC −1.2%) [38].
SSc-ILD has a heterogeneous and variable disease course and close monitoring is important in considering when to start treatment. In a recent published cohort study of 826 SSc-ILD patients, 27% showed progressive ILD (FVC decline >5%) during the initial 12-month period, and 67% experienced progression any time over the mean five-year follow-up [39]. In current clinical practice, treatment is often initiated after progression has occurred, and novel treatment concepts are needed in patients at risk of developing a progressive fibrosing phenotype that aim for the prevention of progression. In this regard, tocilizumab, an IL-6 receptor antibody, is a promising biological agent as exploratory analysis of the faSScinate phase 2 trial and the focuSSced phase 3 trial suggested that tocilizumab could preserve pulmonary function in very early SSc-ILD patients [40,41].

3.1.2. Rheumatoid Arthritis-Associated ILD (RA-ILD)

The development of ILD in RA is associated with a three-times greater risk of mortality, with a median survival of 3–10 years [15,42,43,44,45,46]. Jacob et al. identified 23% of a RA-ILD cohort with an IPF-like progressive fibrotic phenotype based on the combination of the presence of a modified UIP pattern and the presence of extensive fibrosis, which was defined according to the staging system of Goh et al. in SSc-ILD (see above) [28,47]. The management of RA-ILD is challenging, as so far, no RCTs have been performed. Evidence for the use of MMF and CYC has been extrapolated from RCTs in SSc-ILD, as only small cohort studies have been performed in RA-ILD [48,49]. The biologicals rituximab and abatacept, a cytotoxic T lymphocyte antigen 4 (CTLA-4)-immunoglobulin, had a beneficial effect in small open-label and retrospective cohort studies [50,51,52,53,54]. The effect of immunosuppressive agents in RA-UIP remains unclear, and patients with inflammatory types of ILD, such as NSIP and organizing pneumonia, generally have a better response to immunosuppressive agents. There are historical concerns about methotrexate (MTX)-related hypersensitivity pneumonitis and the association between MTX and RA-ILD, but in a recent study by Juge et al., MTX was not associated with an increased risk of RA-ILD, and ILD detection was delayed by 3.6 years [55].

3.2. Interstitial Pneumonia with Autoimmune Features (IPAF)

A subgroup of patients that develop interstitial pneumonia have clinical characteristics suggestive of an autoimmune process, but do not fulfill the established criteria for a CTD. Several terms and criteria to describe these patients have been used, including “undifferentiated CTD-associated ILD” (UCTD-ILD), “lung-dominant CTD” or “autoimmune-featured ILD” [56,57,58]. In 2015, the European Respiratory Society (ERS) and American Thoracic Society (ATS) “Task Force on Undifferentiated Forms of Connective Tissue Disease-associated Interstitial Lung Disease” proposed the term “interstitial pneumonia with autoimmune features (IPAF)” and provided classification criteria [59]. So far, no prospective studies have been performed on IPAF. In a retrospective cohort study by Odlham et al., IPAF patients with an UIP pattern had an outcome similar to IPF, while IPAF patients with a non-UIP pattern had an outcome similar to CTD-ILD [18].

3.3. Idiopathic Non-Specific Interstitial Pneumonia (iNSIP)

iNSIP has been subcategorized into cellular iNSIP and fibrotic iNSIP, and cellular iNSIP generally responds better to immunosuppressive treatment than fibrotic iNSIP [60,61]. Corticosteroids and other immunosuppressive agents such as AZA, CYC and MMF have always been the cornerstone of management in iNSIP, but no RCTs are available, and only a few retrospective cohort studies have investigated the efficacy of immunosuppressive treatment in iNSIP patients. In a retrospective study by Park et al., the response rate to immunosuppressive treatment—defined as an increase in FVC%pred of ≥10% or diffusing capacity of the lung (DLCO) ≥15% after 1 year—was 53% [62]. Nineteen percent deteriorated despite treatment and had a mortality rate comparable to IPF patients (69% five-year mortality). In a study by Nunes et al., response to treatment was the most robust predictor of mortality [63]. Long-term evolution of pulmonary function tests showed a deterioration in 60% despite immunosuppressive treatment. The response rate to immunosuppressive agents was 26% and was comparable to the previously reported 25% in a study by Danniil et al. and to the reported 29% in a study by Nicholson et al. [64,65]. In summary, the disease course of iNSIP patients is variable, but a proportion of iNSIP patients will deteriorate despite conventional immunosuppressive treatment.

3.4. Sarcoidosis

Sarcoidosis has an unpredictable clinical course ranging from asymptomatic self-limited disease to chronic progressive fibrosing disease that is refractory to treatment [66]. Recently, the American Thoracic Society published first practice guidelines for the diagnosis and detection of sarcoidosis [67]. Walsh et al. provided a simple prognostic staging system for disease staging and for guiding treatment decisions based on the composite physiological index (CPI)—which was developed as a tool by Wells et al. to reflect the extent of fibrosis on CT more accurately than individual pulmonary function test indices—and two HRCT variables: CPI > 40, a main pulmonary artery diameter to ascending aorta diameter ratio of greater than 1 (CPI < 40) or an extent of fibrosis of >20% was highly predictive of mortality (HR 5.89) [68,69]. Most patients do not need any immunosuppressive treatment but in those that do, corticosteroids are considered the cornerstone of treatment [70,71,72]. Second-line therapy includes MTX, leflunomide, MMF and AZA [73,74]. In a retrospective study by Vorselaars et al., MTX and AZA had similar efficacy, but a higher infection rate was observed in the group treated with AZA [75]. TNF-α antagonists are used in cases of refractory disease [76,77]. However, treatment recommendations are based mainly on clinical practice and expert opinion rather than results from RCTs [66].

3.5. Hypersensitivity Pneumonitis (HP)

HP is a an immune-mediated disease caused by sensitization to an antigen. Until recently, HP has been classified as acute or chronic HP (cHP) according to clinical presentation [78]. In addition to antigen avoidance, management of HP classically consists of treatment with corticosteroids and other immunosuppressive agents. However, there is only one randomized trial that assessed the efficacy of corticosteroids in a small cohort of patients with acute farmer’s lung. Patients treated with prednisolone had a more rapid improvement of pulmonary function, but after a follow-up of five years, no differences were observed between both groups [79]. De Sadeleer et al. stratified a retrospective cohort of HP patients according to the presence of fibrosis on high-resolution CT (HRCT) and confirmed former evidence that fibrosis correlates more with disease behavior than symptom duration [80]. A beneficial effect of corticosteroid treatment on FVC and DLCO decline was only seen in non-fibrotic HP, but in fibrotic HP, the data suggested a worse outcome. The treatment effect of AZA and MMF was investigated in a retrospective cohort study by Morrisset et al. [81]. Both drugs significantly increased DLCO in cHP patients but had no effect on FVC. No beneficial effect of treatment with corticosteroids before initiation of MMF or AZA was seen in the entire study cohort. Another study demonstrated that the use of MMF or AZA in addition to corticosteroids may decrease adverse events without worsening lung function decline or survival when compared to prednisolone alone [82]. Of note, patients who received immunosuppressive treatment had worsened survival compared to patients who did not. It is unclear whether this increased mortality was the result of a different clinical course or a consequence of immunosuppressive treatment. Recently, guidelines of the American Thoracic Society supported the classification of HP in fibrotic HP and non-fibrotic HP [83]. This might be the start of new research and prospective RCTs in this field.

3.6. Occupational ILD

The most common occupational ILDs are asbestosis and silicosis. In fibrosing asbestosis, chest CT resembles the UIP-pattern of IPF, and other radiological features such as pleural thickening or plaques are helpful to make the distinction between IPF and asbestosis. The prognosis is generally better than in IPF, but the disease course depends on its extent and the quantity of asbestosis retained in the lungs [2,84,85]. Simple silicosis is characterized radiologically by small round opacities with an upper-lobe predominance which can coalesce, resulting in progressive massive fibrosis. Some patients present with an UIP-pattern without the typical nodular opacities. The development and progression of chronic silicosis is generally slow, but an accelerated form exists [84,86,87,88,89]. As there are no known immunosuppressive agents that are effective in the treatment of occupational ILD, the mainstay of treatment is avoidance of the causative mineral dusts [84].

3.7. Unclassifiable ILD (uILD)

ILD can remain unclassifiable if there are conflicting clinical, radiological or histopathological findings, if the risks of a surgical biopsy do not outweigh the potential benefits or if patients are unable or unwilling to undergo a surgical biopsy [26,90,91,92,93]. Up to 15% of ILD cases are unclassifiable [90]. The prognosis of uILD is variable [92,94]. In a study by Ryerson et al., uILD patients with honeycombing, a UIP or possible UIP pattern, a high fibrosis score and cases in whom a diagnosis of IPF was suspected in the differential diagnosis had an outcome comparable to patients with IPF [92]. However, a proportion of these patients would probably be reclassified as having IPF nowadays, as the current guidelines give more weight to radiological parameters, and recommend a surgical lung biopsy only for cases having a probable or indeterminate UIP pattern on HRCT and for cases with a possible alternative diagnosis [95]. Management of unclassifiable ILD is challenging and should be based on the most probable diagnosis [90,96,97]. There is no direct evidence to guide the decision of immunosuppressive treatment in uILD. However, if the probability of IPF is moderate or high, immunosuppressive agents should be avoided given the increased mortality in IPF patients in the PANTHER-IPF trial [10].

4. Antifibrotic Treatment: Where Are We?

4.1. Antifibrotics in SSc-ILD

Recently, RCTs provided evidence for the use of antifibrotic treatment in non-IPF (P)F-ILD (Table 1) [98]. SENSCIS, a phase 3 clinical trial, investigated the efficacy and safety of antifibrotics in non-IPF F-ILD [24]. Patients with systemic sclerosis and fibrosis affecting at least 10% of the lungs on HRCT were included. This study demonstrated a reduced FVC decline in patients treated with nintedanib compared to placebo (−52.4 mL vs. −93.3 mL) over a one-year period. An acceptable safety profile of pirfenidone in SSc-ILD patients was demonstrated in an open-label phase 2 study (LOTUSS) [99].

4.2. Antifibrotics in PF-ILD

The INBUILD trial included patients with a broad range of non-IPF ILDs [23]. Patients who had fibrotic changes affecting at least 10% of the lungs on HRCT and who were progressive despite conventional treatment were eligible for enrollment. Progression was defined as one of the following criteria within 24 months before screening: (1) a relative decline in FVC%pred of ≥10% or (2) two elements of the following: a relative decline in FVC%pred of ≥5 but <10%, worsening of symptoms or increasing extent of fibrosis on HRCT. This study showed a beneficial effect of nintedanib on FVC decline compared to placebo (−80.8 mL/y vs. −187.8 mL/y). The INBUILD-trial was not powered to provide evidence for the efficacy of nintedanib in the specific ILD entities, but post hoc analysis suggested that nintedanib reduces FVC decline irrespective of the underlying ILD subtype [100]. Results of the RELIEF trial, a phase 2 trial of pirfenidone in non-IPF PF-ILD, so far only have been published in abstract form, but showed a reduction in FVC and DLCO decline in patients treated with pirfenidone compared to placebo [25]. Progression was defined as an annualized absolute decline in FVC%pred of ≥5% within 6–24 months prior to inclusion. A recently published phase 2 trial investigated the efficacy and safety of pirfenidone in progressive uILD [26]. Study participants had F-ILD that was unclassifiable after multidisciplinary discussion and had an absolute decline in FVC%pred of >5% or significant symptomatic worsening six months prior to inclusion, attributable to the underlying uILD. FVC change measured by daily home spirometry was chosen as the primary endpoint, but due to intraindividual variability in this home spirometry, the predetermined statistical model could not be administered to the primary endpoint. However, FVC decline measured by spirometry at study visits, a secondary endpoint, suggested a beneficial effect of pirfenidone in patients with unclassifiable PF-ILD (mean change in FVC at 24 weeks: pirfenidone −17.8 mL vs. placebo −113.0 mL).
Several other clinical trials are planned or ongoing to assess the efficacy and safety of antifibrotics in non-IPF (progressive) F-ILD, as demonstrated in Table 1.

5. Unresolved Key Issues in the Pharmacological Management of Non-IPF F-ILD: Where We Would Like to Be

5.1. The Use of Immunosuppressive Agents: The Need for Prospective Studies

Although immunosuppressive treatment has always been the mainstay of treatment in non-IPF ILD, the evidence basis for immunosuppression in non-IPF ILD is limited, as RCTs have mainly been performed in CTD-ILD, and evidence is mainly based on retrospective studies or small open-label studies. Moreover, a substantial proportion of non-IPF ILD patients will deteriorate despite conventional immunosuppressive treatment. Non-IPF ILD patients, and especially those with a progressive fibrosing phenotype, have shared pathogenetic mechanisms with IPF, such as short telomeres: rare genetic variants in telomere-related genes and reduced telomere length have been identified in familial pulmonary fibrosis, sporadic IPF and more recently, in a variety of other ILD subtypes such as cHP, uILD and CTD-ILD [22,101,102,103]. Short telomeres are associated with a progressive phenotype and a poor prognosis [22]. Newton et al. investigated the association between age-adjusted leukocyte telomere length (LTL) and the harmful effect of immunosuppressive agents in the PANTHER-IPF trial [104]. Patients with LTL < 10th percentile that received triple therapy demonstrated a worse composite endpoint-free survival (death, lung transplantation, hospitalization or FVC decline) compared to placebo. In contrast, there was no difference in composite endpoint-free survival for patients randomized to the triple therapy or placebo arms who had LTL > 10th percentile. If this association between telomere length and the effect of immunosuppressive agents is independent of the ILD diagnosis, then it is possible that immunosuppressive agents have a detrimental effect in non-IPF patients with short telomeres. Prospective RCTs are needed to address this question and to identify those patients that benefit from immunosuppressive treatment.
However, immune dysregulation is also thought to be one of the pathobiological mechanisms, especially in CTD-ILD which generally responds better to immunosuppressive treatment than other ILD entities. This highlights the need of pursuing an accurate ILD diagnosis which carries prognostic and therapeutic implications. Moreover, it should be stressed that immunosuppressive treatment improved FVC in RCTs in SSc-ILD, while antifibrotics have only been shown to reduce FVC decline. Moreover, patients in the SLS-II trial had worse pulmonary-function baseline characteristics compared to patients in the SENSCIS trial [24,33]. Several clinical trials investigating the efficacy of immunosuppressive agents and biologicals are still ongoing (Table 2). Combination treatment with MMF and rituximab is currently being investigated in patients with iNSIP and NSIP associated with CTD or IPAF that do not respond to first-line immunosuppressive treatment (EvER-ILD (NCT02990286)), and a head-to-head comparison between rituximab and CYC is currently being performed in patients with severe and/or progressive CTD-ILD (RECITAL (NCT01862926)) [105,106]. The PULMORA-trial (NCT04311567) will investigate the role of tofacitinib in RA-ILD, and abatacept and basiliximab are currently being investigated in myositis-associated ILD (ATtackMy-ILD-NCT03215927)/RA-ILD (APRIL-NCT03084419) and amyopathic dermatomyositis-associated ILD (NCT03192657), respectively.

5.2. The Use of Antifibrotics: The Need for Adequately Defining Disease Progression

Antifibrotics have been shown to be effective in large, good-quality RCTs on PF-ILD, but to date there is no universally accepted definition of progression [23,25,26]. The eligibility criteria of clinical trials have provided a starting point to establish standardized criteria for disease progression. These criteria are based on the occurrence of one or more of the following recognized clinical parameters of progression within a certain time period: worsening of respiratory symptoms or lung function and increasing extent of fibrosis on HRCT. The duration of the time period and thresholds for defining “worsening” and “increasing extent” are not standardized and vary between studies. Another concern is that all definitions of progression imply that patients should wait for clinical deterioration and loss of lung capacity before a progressive fibrosing phenotype can be designated. Further research to identify and validate predictors of progression is needed with the aim of initiating antifibrotic treatment before progression has effectively occurred.
Furthermore, it is worth nothing that antifibrotic treatment is much more expensive than the existing immunosuppressive agents and should therefore be used in a well-selected population (e.g., PF-ILD patients) to guarantee the cost-benefit ratio.

5.3. The Choice between Antifibrotics, Immunosuppressive Agents and Combination Treatment: The Need for Biomarkers

In the future, pharmacological treatment options will consist of no pharmacological treatment, immunosuppressive agents or antifibrotics or even a combination of these treatments. The question as to which drug(s) to use in a particular patient has not been answered yet, as no head-to-head comparisons have been performed. However, soon, clinicians will need to make the decision regarding which therapy to initiate. The identification and validation of biomarkers such as clinical features and genetic and molecular profiles that reflect upregulated pathways, predict disease behavior and identify patients at increased risk of harmful effects of treatment may help to initiate appropriate treatment early in the disease course. Based on current knowledge, it appears likely that patients with an UIP pattern on histopathology or CT or patients with extensive fibrotic disease will benefit from antifibrotic treatment. However, it should be stressed that the INBUILD trial and RELIEF trial selected patients who were progressive despite conventional immunosuppressive treatment and that antifibrotic drugs as a first-line treatment in non-IPF PF-ILD has not yet been investigated.
In the near future, combination treatment with immunosuppressive agents and antifibrotics will be a potential option. The tolerability and safety of combination treatment has been demonstrated in SSc-ILD, but the role of antifibrotic treatment as add-on treatment has not yet been investigated. In the SENSCIS trial, the use of immunosuppressive agents was not randomized, but the FVC decline in the placebo group was lower in patients receiving MMF, suggesting a potential beneficial effect of MMF in fibrosing SSc-ILD. Moreover, patients receiving both therapies had the slowest rate of FVC decline (MMF + nintedanib: −40.2 mL/y; MMF + placebo: −66.5 mL/y; nintedanib: −63.9 mL/y; placebo: 119.3 mL/y), suggesting a potential role for combination treatment in fibrosing SSc-ILD [24].
However, the difference between patients receiving MMF + placebo and those receiving MMF + nintedanib was only small (26.3 mL) and it is unclear whether this small difference is clinically significant. The safety of combination therapy with pirfenidone and MMF has also been demonstrated in progressive fibrosing uILD, but assessment of the effect of MMF on FVC decline was not possible in this study due to small sample sizes [26]. The SLS-3 trial (NCT03221257) is ongoing and investigates the role of combination treatment in SSc-ILD: 150 patients will be randomized to MMF plus pirfenidone or MMF monotherapy, with the primary outcome being the change in FVC%pred from baseline over a time period of 18 months.

6. Conclusions

The clinical course in patients with PF-ILD mimicks the relentless progressiveness of idiopathic pulmonary fibrosis (IPF) and recently, RCTs demonstrated the efficacy of antifibrotic treatment in non-IPF ILD. However, several questions remain and need to be addressed in future studies: which patients benefit from immunosuppressive therapy? Should antifibrotics be restricted to PF-ILD or should it be used in any F-ILD patient? What is the role of combination treatment? Which antifibrotic and immunosuppressive treatment can be combined safely? Further research is needed to address these questions and to identify and validate biomarkers that will help to initiate appropriate treatment early in the disease course.

Author Contributions

Conceptualization, T.G., L.J.D.S. and W.A.W.; methodology, T.G.; writing—original draft preparation, T.G.; writing—review and editing, T.G., L.J.D.S., J.Y., G.M.V., M.V., S.E.V. and W.A.W.; supervision, W.A.W.; project administration, W.A.W.; funding acquisition, W.A.W. All authors have read and agreed to the published version of the manuscript.

Funding

The research reported in this publication was supported by the Research Foundation—Flanders and the University Hospitals Leuven (1.8.325.12N-1S73921N-T001520N) and grants from Roche and Boehringer-Ingelheim. The funding sources were not involved in study design, data collection, data analysis, data interpretation, manuscript writing or the decision to submit the article for publication.

Conflicts of Interest

W.A.W. reports grants from Roche, Boehringer-Ingelheim, Research Foundation—Flanders, all paid to the institution (University Hospitals Leuven), outside the submitted work. L.J.D.S. reports non-financial support from Roche and Boehringer-Ingelheim, outside the submitted work. T.G., J.Y., G.E.V., M.V. and S.E.V. have nothing to disclose.

References

  1. Wells, A.U.; Brown, K.K.; Flaherty, K.R.; Kolb, M.; Thannickal, V.J. What’s in a name? That which we call IPF, by any other name would act the same. Eur. Respir. J. 2018, 51, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cottin, V.; Hirani, N.A.; Hotchkin, D.L.; Nambiar, A.M.; Ogura, T.; Otaola, M.; Skowasch, D.; Park, J.S.; Poonyagariyagorn, H.K.; Wuyts, W.; et al. Presentation, diagnosis and clinical course of the spectrum of progressive-fibrosing interstitial lung diseases. Eur. Respir. Rev. 2018, 27. [Google Scholar] [CrossRef] [Green Version]
  3. Cottin, V.; Wollin, L.; Fischer, A.; Quaresma, M.; Stowasser, S.; Harari, S. Fibrosing interstitial lung diseases: Knowns and unknowns. Eur. Respir. Rev. 2019, 28, 1–9. [Google Scholar] [CrossRef]
  4. Kolb, M.; Vašáková, M. The natural history of progressive fibrosing interstitial lung diseases. Respir. Res. 2019, 20, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Wijsenbeek, M.; Kreuter, M.; Olson, A.; Fischer, A.; Bendstrup, E.; Wells, C.D.; Denton, C.P.; Mounir, B.; Zouad-Lejour, L.; Quaresma, M.; et al. Progressive fibrosing interstitial lung diseases: Current practice in diagnosis and management. Curr. Med. Res. Opin. 2019, 35, 2015–2024. [Google Scholar] [CrossRef] [Green Version]
  6. Nasser, M.; Larrieu, S.; Si-Mohamed, S.; Ahmad, K.; Boussel, L.; Brevet, M.; Chalabreysse, L.; Fabre, C.; Marque, S.; Revel, D.; et al. Progressive fibrosing interstitial lung disease: A clinical cohort (the PROGRESS® study). Eur. Respir. J. 2020, 57, 2002718. [Google Scholar] [CrossRef]
  7. Faverio, P.; Cassandro, R.; Piluso, M.; De Giacomi, F.; Della Zoppa, M.; Harari, S.A.; Luppi, F.; Pesci, A. Progressive fibrosing interstitial lung diseases (PF-ILDs): Prevalence and characterization in two Italian referral centers. Respiration 2020, 99, 838–845. [Google Scholar] [CrossRef]
  8. Gross, T.; Gary, H. Idiopathic pulmonary ibrosis. N. Engl. J. Med. 2001, 345, 517–525. [Google Scholar] [CrossRef] [PubMed]
  9. Demedts, M.; Behr, J.; Buhl, R.; Costabel, U.; Dekhuijzen, R.; Jansen, H.M.; MacNee, W.; Thomeer, M.; Wallaert, B.; Laurent, F.; et al. High-dose acetylcysteine in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2005, 353, 2229–2242. [Google Scholar] [CrossRef] [Green Version]
  10. Idiopathic Pulmonary Fibrosis Clinical Research Network; Raghu, G.; Anstrom, K.J.; King, T.E., Jr.; Lasky, J.A.; Martinez, F.J. Prednisone, Azathioprine, and N-Acetylcysteine for Pulmonary Fibrosis. N. Engl. J. Med. 2012, 366, 1968–1977. [Google Scholar] [CrossRef]
  11. Noble, P.W.; Albera, C.; Bradford, W.Z.; Costabel, U.; Glassberg, M.K.; Kardatzke, D.; King, T.E.; Lancaster, L.; Sahn, S.A.; Szwarcberg, J.; et al. Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): Two randomised trials. Lancet 2011, 377, 1760–1769. [Google Scholar] [CrossRef]
  12. Richeldi, L.; Kreuter, M.; Selman, M.; Crestani, B.; Kirsten, A.M.; Wuyts, W.A.; Xu, Z.; Bernois, K.; Stowasser, S.; Quaresma, M.; et al. Long-term treatment of patients with idiopathic pulmonary fibrosis with nintedanib: Results from the TOMORROW trial and its open-label extension. Thorax 2018, 73, 581–583. [Google Scholar] [CrossRef] [Green Version]
  13. King, T.E.; Bradford, W.Z.; Castro-Bernardini, S.; Fagan, E.A.; Glaspole, I.; Glassberg, M.K.; Gorina, E.; Hopkins, P.M.; Kardatzke, D.; Lancaster, L.; et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2083–2092. [Google Scholar] [CrossRef] [Green Version]
  14. Richeldi, L.; Du Bois, R.M.; Raghu, G.; Azuma, A.; Brown, K.K.; Costabel, U.; Cottin, V.; Flaherty, K.R.; Hansell, D.M.; Inoue, Y.; et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2071–2082. [Google Scholar] [CrossRef] [Green Version]
  15. Tsuchiya, Y.; Takayanagi, N.; Sugiura, H.; Miyahara, Y.; Tokunaga, D.; Kawabata, Y.; Sugita, Y. Lung diseases directly associated with rheumatoid arthritis and their relationship to outcome. Eur. Respir. J. 2011, 37, 1411–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Walsh, S.L.F.; Sverzellati, N.; Devaraj, A.; Wells, A.U.; Hansell, D.M. Chronic hypersensitivity pneumonitis: High resolution computed tomography patterns and pulmonary function indices as prognostic determinants. Eur. Radiol. 2012, 22, 1672–1679. [Google Scholar] [CrossRef]
  17. Gaxiola, M.; Buendía-Roldán, I.; Mejía, M.; Carrillo, G.; Estrada, A.; Navarro, M.C.; Rojas-Serrano, J.; Selman, M. Morphologic diversity of chronic pigeon breeder’s disease: Clinical features and survival. Respir. Med. 2011, 105, 608–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Oldham, J.M.; Adegunsoye, A.; Valenzi, E.; Lee, C.; Witt, L.; Chen, L.; Husain, A.N.; Montner, S.; Chung, J.H.; Cottin, V.; et al. Characterisation of patients with interstitial pneumonia with autoimmune features. Eur. Respir. J. 2016, 47, 1767–1775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ley, B.; Liu, S.; Elicker, B.M.; Henry, T.S.; Vittinghoff, E.; Golden, J.A.; Jones, K.D.; Wolters, P.J. Telomere length in patients with unclassifiable interstitial lung disease: A cohort study. Eur. Respir. J. 2020, 2000268. [Google Scholar] [CrossRef] [PubMed]
  20. Ley, B.; Newton, C.A.; Arnould, I.; Elicker, B.M.; Henry, T.S.; Vittinghoff, E.; Golden, J.A.; Jones, K.D.; Batra, K.; Torrealba, J.; et al. The MUC5B promoter polymorphism and telomere length in patients with chronic hypersensitivity pneumonitis: An observational cohort-control study. Lancet Respir. Med. 2017, 5, 639–647. [Google Scholar] [CrossRef]
  21. Juge, P.A.; Lee, J.S.; Ebstein, E.; Furukawa, H.; Dobrinskikh, E.; Gazal, S.; Kannengiesser, C.; Ottaviani, S.; Oka, S.; Tohma, S.; et al. MUC5B promoter variant and rheumatoid arthritis with interstitial lung disease. N. Engl. J. Med. 2018, 379, 2209–2219. [Google Scholar] [CrossRef] [PubMed]
  22. Newton, C.A.; Batra, K.; Torrealba, J.; Kozlitina, J.; Glazer, C.S.; Aravena, C.; Meyer, K.; Raghu, G.; Collard, H.R.; Garcia, C.K. Telomere-related lung fibrosis is diagnostically heterogeneous but uniformly progressive. Eur. Respir. J. 2016, 48, 1710–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Flaherty, K.R.; Wells, A.U.; Cottin, V.; Devaraj, A.; Walsh, S.L.F.; Inoue, Y.; Richeldi, L.; Kolb, M.; Tetzlaff, K.; Stowasser, S.; et al. Nintedanib in progressive fibrosing interstitial lung diseases. N. Engl. J. Med. 2019, 381, 1718–1727. [Google Scholar] [CrossRef] [Green Version]
  24. Distler, O.; Highland, K.B.; Gahlemann, M.; Azuma, A.; Fischer, A.; Mayes, M.D.; Raghu, G.; Sauter, W.; Girard, M.; Alves, M.; et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N. Engl. J. Med. 2019, 380, 2518–2528. [Google Scholar] [CrossRef]
  25. Guenther, A.; Prasse, A.; Kreuter, M.; Neuser, P.; Rabe, K.; Bonella, F.; Bonnet, R.; Grohe, C.; Held, M.; Wilkens, H.; et al. Late Breaking Abstract—Exploring Efficacy and Safety of oral Pirfenidone for progressive, non-IPF Lung Fibrosis (RELIEF). Eur. Respir. J. 2019, 54, RCT1879. [Google Scholar] [CrossRef]
  26. Maher, T.M.; Corte, T.J.; Fischer, A.; Kreuter, M.; Lederer, D.J.; Molina-Molina, M.; Axmann, J.; Kirchgaessler, K.U.; Samara, K.; Gilberg, F.; et al. Pirfenidone in patients with unclassifiable progressive fibrosing interstitial lung disease: A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Respir. Med. 2020, 8, 147–157. [Google Scholar] [CrossRef]
  27. Tyndall, A.J.; Bannert, B.; Vonk, M.; Airò, P.; Cozzi, F.; Carreira, P.E.; Bancel, D.F.; Allanore, Y.; Müller-Ladner, U.; Distler, O.; 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, 69, 1809–1815. [Google Scholar] [CrossRef] [Green Version]
  28. Goh, N.S.L.; Desai, S.R.; Veeraraghavan, S.; Hansell, D.M.; Copley, S.J.; Maher, T.M.; Corte, T.J.; Sander, C.R.; Ratoff, J.; Devaraj, A.; et al. Interstitial lung disease in systemic sclerosis: A simple staging system. Am. J. Respir. Crit. Care Med. 2008, 177, 1248–1254. [Google Scholar] [CrossRef]
  29. Tashkin, D.P.; Elashoff, R.; Clements, P.J.; Goldin, J.; Roth, M.D.; Furst, D.E.; Arriola, E.; Silver, R.; Strange, C.; Bolster, M.; et al. Cyclophosphamide versus placebo in scleroderma lung disease. N. Engl. J. Med. 2006, 354, 2655–2666. [Google Scholar] [CrossRef] [Green Version]
  30. Hoyles, R.K.; Ellis, R.W.; Wellsbury, J.; Lees, B.; Newlands, P.; Goh, N.S.L.; Roberts, C.; Desai, S.; Herrick, A.L.; McHugh, N.J.; et al. A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma. Arthritis Rheum. 2006, 54, 3962–3970. [Google Scholar] [CrossRef]
  31. Kowal-Bielecka, O.; Fransen, J.; Avouac, J.; Becker, M.; Kulak, A.; Allanore, Y.; Distler, O.; Clements, P.; Cutolo, M.; Czirjak, L.; et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann. Rheum. Dis. 2017, 76, 1327–1339. [Google Scholar] [CrossRef] [Green Version]
  32. Tashkin, D.P.; Elashoff, R.; Clements, P.J.; Roth, M.D.; Furst, D.E.; Silver, R.M.; Goldin, J.; Arriola, E.; Strange, C.; Bolster, M.B.; et al. Effects of 1-year treatment with cyclophosphamide on outcomes at 2 years in scleroderma lung disease. Am. J. Respir. Crit. Care Med. 2007, 176, 1026–1034. [Google Scholar] [CrossRef]
  33. Tashkin, D.P.; Roth, M.D.; Clements, P.J.; Furst, D.E.; Khanna, D.; Kleerup, E.C.; Goldin, J.; Arriola, E.; Volkmann, E.R.; Kafaja, S.; 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, 4, 708–719. [Google Scholar] [CrossRef] [Green Version]
  34. Boerner, E.B.; Cuyas, M.; Theegarten, D.; Ohshimo, S.; Costabel, U.; Bonella, F. Azathioprine for Connective Tissue Disease-Associated Interstitial Lung Disease. Respiration 2020, 99, 628–636. [Google Scholar] [CrossRef] [PubMed]
  35. Oldham, J.M.; Lee, C.; Valenzi, E.; Witt, L.J.; Adegunsoye, A.; Hsu, S.; Chen, L.; Montner, S.; Chung, J.H.; Noth, I.; et al. Azathioprine Response in Patients with Fibrotic Connective Tissue. Respir. Med. 2016, 121, 117–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Paone, C.; Chiarolanza, I.; Cuomo, G.; Ruocco, L.; Vettori, S.; Menegozzo, M.; La Montagna, G.; Valentini, G. Twelve-month azathioprine as maintenance therapy in early diffuse systemic sclerosis patients treated for 1-year with low dose cyclophosphamide pulse therapy. Clin. Exp. Rheumatol. 2007, 25, 613–616. [Google Scholar]
  37. Bérezné, A.; Ranque, B.; Valeyre, D.; Brauner, M.; Allanore, Y.; Launay, D.; Le Guern, V.; Kahn, J.E.; Couderc, L.J.; Constans, J.; et al. Therapeutic strategy combining intravenous cyclophosphamide followed by oral azathioprine to treat worsening interstitial lung disease associated with systemic sclerosis: A retrospective multicenter open-label study. J. Rheumatol. 2008, 35, 1064–1072. [Google Scholar]
  38. Sircar, G.; Goswami, R.P.; Sircar, D.; Ghosh, A.; Ghosh, P. Intravenous cyclophosphamide vs rituximab for the treatment of early diffuse scleroderma lung disease: Open label, randomized, controlled trial. Rheumatology 2018, 57, 2106–2113. [Google Scholar] [CrossRef]
  39. Hoffmann-Vold, A.M.; Allanore, Y.; Alves, M.; Brunborg, C.; Airó, P.; Ananieva, L.P.; Czirják, L.; Guiducci, S.; Hachulla, E.; Li, M.; et al. Progressive interstitial lung disease in patients with systemic sclerosis-associated interstitial lung disease in the EUSTAR database. Ann. Rheum. Dis. 2020, 1–9. [Google Scholar] [CrossRef]
  40. Khanna, D.; Denton, C.P.; Jahreis, A.; van Laar, J.M.; Frech, T.M.; Anderson, M.E.; Baron, M.; Chung, L.; Fierlbeck, G.; Lakshminarayanan, S.; et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): A phase 2, randomised, controlled trial. Lancet 2016, 387, 2630–2640. [Google Scholar] [CrossRef]
  41. Khanna, D.; Lin, C.J.F.; Furst, D.E.; Goldin, J.; Kim, G.; Kuwana, M.; Allanore, Y.; Matucci-Cerinic, M.; Distler, O.; Shima, Y.; et al. Tocilizumab in systemic sclerosis: A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 2020, 8, 963–974. [Google Scholar] [CrossRef]
  42. Yunt, Z.X.; Chung, J.H.; Hobbs, S.; Fernandez-Perez, E.R.; Olson, A.L.; Huie, T.J.; Keith, R.C.; Janssen, W.J.; Goldstein, B.L.; Lynch, D.A.; et al. High resolution computed tomography pattern of usual interstitial pneumonia in rheumatoid arthritis-associated interstitial lung disease: Relationship to survival. Respir. Med. 2017, 126, 100–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bongartz, T.; Nannini, C.; Medina-Velasquez, Y.F.; Achenbach, S.J.; Crowson, C.S.; Ryu, J.H.; Vassallo, R.; Gabriel, S.E.; Matteson, E.L. Incidence and Mortality of Inter-stitial Lung Disease in Rheumatoid Arthritis: A Population Based Study. Arthritis Rheum. 2010, 62, 1583–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Solomon, J.J.; Ryu, J.H.; Tazelaar, H.D.; Myers, J.L.; Tuder, R.; Cool, C.D.; Curran-Everett, D.; Fischer, A.; Swigris, J.J.; Brown, K.K. Fibrosing interstitial pneumonia predicts survival in patients with rheumatoid arthritis-associated interstitial lung disease (RA-ILD). Respir. Med. 2013, 107, 1247–1252. [Google Scholar] [CrossRef] [Green Version]
  45. Solomon, J.J.; Chung, J.H.; Cosgrove, G.P.; Demoruelle, M.K.; Fernandez-Perez, E.R.; Fischer, A.; Frankel, S.K.; Hobbs, S.B.; Huie, T.J.; Ketzer, J.; et al. Predictors of mortality in rheumatoid arthritis-associated interstitial lung disease. Eur. Respir. J. 2016, 47, 588–596. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, E.J.; Collard, H.R.; King, T.E. Rheumatoid arthritis-associated interstitial lung disease: The relevance of histopathologic and radiographic pattern. Chest 2009, 136, 1397–1405. [Google Scholar] [CrossRef] [Green Version]
  47. Jacob, J.; Hirani, N.; Van Moorsel, C.H.M.; Rajagopalan, S.; Murchison, J.T.; Van Es, H.W.; Bartholmai, B.J.; Van Beek, F.T.; Struik, M.H.L.; Stewart, G.A.; et al. Predicting outcomes in rheumatoid arthritis related interstitial lung disease. Eur. Respir. J. 2019, 53. [Google Scholar] [CrossRef]
  48. Fischer, A.; Brown, K.K.; Du Bois, R.M.; Frankel, S.K.; Cosgrove, G.P.; Fernandez-Perez, E.R.; Huie, T.J.; Krishnamoorthy, M.; Meehan, R.T.; Olson, A.L.; et al. Mycophenolate mofetil improves lung function in connective tissue disease-associated interstitial lung disease. J. Rheumatol. 2013, 40, 640–646. [Google Scholar] [CrossRef]
  49. Saketkoo, L.; Espinoza, A.L.R. Rheumatoid Arthritis Interstitial Lung Disease: Mycophenolate Mofetil as an Antifibrotic and Disease-Modifying Antirheumatic Drug. Arch. Intern. Med. 2014, 168, 1718–1719. [Google Scholar] [CrossRef]
  50. Cassone, G.; Manfredi, A.; Atzeni, F.; Venerito, V.; Vacchi, C.; Picerno, V.; Furini, F.; Erre, G.L.; Tomietto, P.; Fedele, A.L.; et al. Safety of Abatacept in Italian Patients with Rheumatoid Arthritis and Interstitial Lung Disease: A Multicenter Retrospective Study. J. Clin. Med. 2020, 9, 277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lopez-Olivo, M.A.; Amezaga Urruela, M.; Mcgahan, L.; Pollono, E.N.; Suarez-Almazor, M.E. Rituximab for rheumatoid arthritis. Cochrane Database Syst. Rev. 2015, 2017. [Google Scholar] [CrossRef]
  52. Fernández-Díaz, C.; Loricera, J.; Castañeda, S.; López-Mejías, R.; Ojeda-García, C.; Olivé, A.; Rodríguez-Muguruza, S.; Carreira, P.E.; Pérez-Sandoval, T.; Retuerto, M.; et al. Abatacept in patients with rheumatoid arthritis and interstitial lung disease: A national multicenter study of 63 patients. Semin. Arthritis Rheum. 2018, 48, 22–27. [Google Scholar] [CrossRef] [Green Version]
  53. Matteson, E.L.; Bongartz, T.; Ryu, J.H.; Crowson, C.S.; Hartman, T.E.; Dellaripa, P.F. Open-Label, Pilot Study of the Safety and Clinical Effects of Rituximab in Patients with Rheumatoid Arthritis-Associated Interstitial Pneumonia. Open J. Rheumatol. Autoimmune Dis. 2012, 2, 53–58. [Google Scholar] [CrossRef] [Green Version]
  54. Md Yusof, M.Y.; Kabia, A.; Darby, M.; Lettieri, G.; Beirne, P.; Vital, E.M.; Dass, S.; Emery, P. Effect of rituximab on the progression of rheumatoid arthritis-related interstitial lung disease: 10 years’ experience at a single centre. Rheumatology (UK) 2017, 56, 1348–1357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Juge, P.-A.; Lee, J.S.; Lau, J.; Kawano-Dourado, L.; Rojas Serrano, J.; Sebastiani, M.; Koduri, G.; Matteson, E.; Bonfiglioli, K.; Sawamura, M.; et al. Methotrexate and rheumatoid arthritis associated interstitial lung disease. Eur. Respir. J. 2020, 2000337. [Google Scholar] [CrossRef]
  56. Fischer, A.; West, S.G.; Swigris, J.J.; Brown, K.K.; Du Bois, R.M. Connective tissue disease-associated interstitial lung disease: A call for clarification. Chest 2010, 138, 251–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kinder, B.W.; Collard, H.R.; Koth, L.; Daikh, D.I.; Wolters, P.J.; Elicker, B.; Jones, K.D.; King, T.E. Idiopathic nonspecific interstitial pneumonia: Lung manifestation of undifferentiated connective tissue disease? Am. J. Respir. Crit. Care Med. 2007, 176, 691–697. [Google Scholar] [CrossRef] [Green Version]
  58. Vij, R.; Noth, I.; Strek, M.E. Autoimmune-featured interstitial lung disease: A distinct entity. Chest 2011, 140, 1292–1299. [Google Scholar] [CrossRef] [Green Version]
  59. Fischer, A.; Antoniou, K.M.; Brown, K.K.; Cadranel, J.; Corte, T.J.; Du Bois, R.M.; Lee, J.S.; Leslie, K.O.; Lynch, D.A.; Matteson, E.L.; et al. An official European Respiratory Society/American Thoracic Society research statement: Interstitial pneumonia with autoimmune features. Eur. Respir. J. 2015, 46, 976–987. [Google Scholar] [CrossRef] [Green Version]
  60. Tomassetti, S.; Ryu, J.H.; Piciucchi, S.; Chilosi, M.; Poletti, V. Nonspecific Interstitial Pneumonia: What Is the Optimal Approach to Management? Semin. Respir. Crit. Care Med. 2016, 37, 378–394. [Google Scholar] [CrossRef]
  61. Travis, W.D.; Hunninghake, G.; King, T.E.; Lynch, D.A.; Colby, T.V.; Galvin, J.R.; Brown, K.K.; Man, P.C.; Cordier, J.F.; Du Bois, R.M.; et al. Idiopathic nonspecific interstitial pneumonia: Report of an American Thoracic Society Project. Am. J. Respir. Crit. Care Med. 2008, 177, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
  62. Park, I.N.; Jegal, Y.; Kim, D.S.; Do, K.H.; Yoo, B.; Shim, T.S.; Lim, C.M.; Lee, S.D.; Koh, Y.; Kim, W.S.; et al. Clinical course and lung function change of idiopathic nonspecific interstitial pneumonia. Eur. Respir. J. 2009, 33, 68–76. [Google Scholar] [CrossRef] [Green Version]
  63. Nunes, H.; Schubel, K.; Piver, D.; Magois, E.; Feuillet, S.; Uzunhan, Y.; Carton, Z.; Tazi, A.; Levy, P.; Brillet, P.Y.; et al. Nonspecific interstitial pneumonia: Survival is influenced by the underlying cause. Eur. Respir. J. 2015, 45, 746–755. [Google Scholar] [CrossRef] [Green Version]
  64. Daniil, Z.D.; Gilchrist, F.C.; Nicholson, A.G.; Hansell, D.M.; Harris, J.; Colby, T.V.; Du Bois, R.M. A histologic pattern of nonspecific interstitial pneumonia is associated with a better prognosis than usual interstitial pneumonia in patients with cryptogenic fibrosing alveolitis. Am. J. Respir. Crit. Care Med. 1999, 160, 899–905. [Google Scholar] [CrossRef] [PubMed]
  65. Nicholson, A.G.; Colby, T.V.; Dubois, R.M.; Hansell, D.M.; Wells, A.U. The prognostic significance of the histologic pattern of interstitial pneumonia in patients presenting with the clinical entity of cryptogenic fibrosing alveolitis. Am. J. Respir. Crit. Care Med. 2000, 162, 2213–2217. [Google Scholar] [CrossRef] [PubMed]
  66. Spagnolo, P.; Rossi, G.; Trisolini, R.; Sverzellati, N.; Baughman, R.P.; Wells, A.U. Pulmonary sarcoidosis. Lancet Respir. Med. 2018, 2600, 1–14. [Google Scholar] [CrossRef]
  67. Crouser, E.D.; Maier, L.A.; Baughman, R.P.; Abston, E.; Bernstein, R.C.; Blankstein, R.; Bonham, C.A.; Chen, E.S.; Culver, D.A.; Drake, W.; et al. Diagnosis and Detection of Sarcoidosis An Official American Thoracic Society Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2020, 201, E26–E51. [Google Scholar] [CrossRef] [PubMed]
  68. Wells, A.U.; Desai, S.R.; Rubens, M.B.; Goh, N.S.L.; Cramer, D.; Nicholson, A.G.; Colby, T.V.; Du Bois, R.M.; Hansell, D.M. Idiopathic pulmonary fibrosis: A composite physiologic index derived from disease extent observed by computed tomography. Am. J. Respir. Crit. Care Med. 2003, 167, 962–969. [Google Scholar] [CrossRef]
  69. Walsh, S.L.F.; Wells, A.U.; Sverzellati, N.; Keir, G.J.; Calandriello, L.; Antoniou, K.M.; Copley, S.J.; Devaraj, A.; Maher, T.M.; Renzoni, E.; et al. An integrated clinicoradiological staging system for pulmonary sarcoidosis: A case-cohort study. Lancet Respir. Med. 2014, 2, 123–130. [Google Scholar] [CrossRef] [Green Version]
  70. Costabel, U.; Hunninghake, G.W.; Statement, S. EDITORIAL ATS / ERS / WASOG statement on sarcoidosis. Eur. Respir. J. 1999, 14, 735–737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Paramothayan, N.S.; Lasserson, T.J.; Jones, P.W. Corticosteroids for pulmonary sarcoidosis. Cochrane Database Syst. Rev. 2005, 2, CD001114. [Google Scholar] [CrossRef]
  72. Paramothayan, S.; Jones, P.W. Corticosteroid therapy in pulmonary sarcoidosis: A systematic review. JAMA 2002, 287, 1301–1307. [Google Scholar] [CrossRef] [PubMed]
  73. Müller-Quernheim, J.; Kienast, K.; Held, M.; Pfeifer, S.; Costabel, U. Treatment of chronic sarcoidosis with an azathioprine/prednisolone regimen. Eur. Respir. J. 1999, 14, 1117–1122. [Google Scholar] [CrossRef] [Green Version]
  74. Baughman, R.P.; Lower, E.E. A clinical approach to the use of methotrexate for sarcoidosis. Thorax 1999, 54, 742–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Vorselaars, A.D.M.; Wuyts, W.A.; Vorselaars, V.M.M.; Zanen, P.; Deneer, V.H.M.; Veltkamp, M.; Thomeer, M.; Van Moorsel, C.H.M.; Grutters, J.C. Methotrexate vs azathioprine in second-line therapy of sarcoidosis. Chest 2013, 144, 805–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Baughman, R.P.; Drent, M.; Kavuru, M.; Judson, M.A.; Costabel, U.; du Bois, R.; Albera, C.; Brutsche, M.; Davis, G.; Donohue, J.F.; et al. Infliximab therapy in patients with chronic sarcoidosis and pulmonary involvement. Am. J. Respir. Crit. Care Med. 2006, 174, 795–802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Rahaghi, F.F.; Baughman, R.P.; Saketkoo, L.A.; Sweiss, N.J.; Barney, J.B.; Birring, S.S.; Costabel, U.; Crouser, E.D.; Drent, M.; Gerke, A.K.; et al. Delphi consensus recommendations for a treatment algorithm in pulmonary sarcoidosis. Eur. Respir. Rev. 2020, 29. [Google Scholar] [CrossRef] [PubMed]
  78. Vasakova, M.; Morell, F.; Walsh, S.; Leslie, K.; Raghu, G. Hypersensitivity pneumonitis: Perspectives in diagnosis and management. Am. J. Respir. Crit. Care Med. 2017, 196, 680–689. [Google Scholar] [CrossRef]
  79. Kokkarinen, J.I.; Tukiainen, H.O.; Terho, E.O. Effect of corticosteroid treatment on the recovery of pulmonary function in farmer’s lung. Am. Rev. Respir. Dis. 1992, 145, 3–5. [Google Scholar] [CrossRef]
  80. De Sadeleer, L.; Hermans, F.; De Dycker, E.; Yserbyt, J.; Verschakelen, J.; Verbeken, E.; Verleden, G.; Wuyts, W. Effects of Corticosteroid Treatment and Antigen Avoidance in a Large Hypersensitivity Pneumonitis Cohort: A Single-Centre Cohort Study. J. Clin. Med. 2018, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  81. Morisset, J.; Johannson, K.A.; Vittinghoff, E.; Aravena, C.; Elicker, B.M.; Jones, K.D.; Fell, C.D.; Manganas, H.; Dubé, B.P.; Wolters, P.J.; et al. Use of Mycophenolate Mofetil or Azathioprine for the Management of Chronic Hypersensitivity Pneumonitis. Chest 2017, 151, 619–625. [Google Scholar] [CrossRef]
  82. Adegunsoye, A.; Oldham, J.M.; Fernández Pérez, E.R.; Hamblin, M.; Patel, N.; Tener, M.; Bhanot, D.; Robinson, L.; Bullick, S.; Chen, L.; et al. Outcomes of immunosuppressive therapy in chronic hypersensitivity pneumonitis. ERS Monogr. 2017, 3, 1–11. [Google Scholar] [CrossRef]
  83. Raghu, G.; Remy-jardin, M.; Ryerson, C.J.; Myers, J.L.; Kreuter, M.; Vasakova, M.; Bargagli, E.; Chung, J.H.; Collins, B.F.; Bendstrup, E.; et al. Diagnosis of Hypersensitivity Pneumonitis in Adults. An Official ATS/JRS/ALAT Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2020, 202, e36–e69. [Google Scholar] [CrossRef] [PubMed]
  84. Glazer, C.S.; Newman, L.S. Occupational interstitial lung disease. Clin. Chest Med. 2004, 25, 467–478. [Google Scholar] [CrossRef] [PubMed]
  85. Oksa, P.; Huuskonen, M.S.; Järvisalo, J.; Klockars, M.; Zitting, A.; Suoranta, H.; Tossavainen, A.; Vattulainen, K.; Laippala, P. Follow-up of asbestosis patients and predictors for radiographic progression. Int. Arch. Occup. Environ. Health 1998, 71, 465–471. [Google Scholar] [CrossRef] [PubMed]
  86. Castranova, V.; Vallyathan, V. Silicosis and coal workers’ pneumoconiosis. Environ. Health Perspect. 2000, 108, 675–684. [Google Scholar] [CrossRef] [Green Version]
  87. Lee, H.S.; Phoon, W.H.; Ng, T.P. Radiological progression and its predictive risk factors in silicosis. Occup. Environ. Med. 2001, 58, 467–471. [Google Scholar] [CrossRef] [Green Version]
  88. Arakawa, H.; Fujimoto, K.; Honma, K.; Suganuma, N.; Morikubo, H.; Saito, Y.; Shida, H.; Kaji, Y. Progression from near-normal to end-stage lungs in chronic interstitial pneumonia related to silica exposure: Long-term CT observations. Am. J. Roentgenol. 2008, 191, 1040–1045. [Google Scholar] [CrossRef]
  89. Arakawa, H.; Johkoh, T.; Honma, K.; Saito, Y.; Fukushima, Y.; Shida, H.; Suganuma, N. Chronic interstitial pneumonia in silicosis and mix-dust pneumoconiosis: Its prevalence and comparison of CT findings with idiopathic pulmonary fibrosis. Chest 2007, 131, 1870–1876. [Google Scholar] [CrossRef]
  90. Guler, S.A.; Ellison, K.; Algamdi, M.; Collard, H.R.; Ryerson, C.J. Heterogeneity in unclassifiable interstitial lung disease a systematic review and meta-analysis. Ann. Am. Thorac. Soc. 2018, 15, 854–863. [Google Scholar] [CrossRef]
  91. Leung, S.C.; Churg, A.M.; Leipsic, J.A.; Levy, R.D.; Wilcox, P.G.; Ryerson, C.J. Unclassifiable interstitial lung disease: An unresolved diagnostic dilemma. Respirol. Case Rep. 2015, 3, 85–88. [Google Scholar] [CrossRef]
  92. Ryerson, C.J.; Urbania, T.H.; Richeldi, L.; Mooney, J.J.; Lee, J.S.; Jones, K.D.; Elicker, B.M.; Koth, L.L.; King, T.E.; Wolters, P.J.; et al. Prevalence and prognosis of unclassifiable interstitial lung disease. Eur. Respir. J. 2013, 42, 750–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Travis, W.D.; Costabel, U.; Hansell, D.M.; King, T.E.; Lynch, D.A.; Nicholson, A.G.; Ryerson, C.J.; Ryu, J.H.; Selman, M.; Wells, A.U.; et al. An official American Thoracic Society/European Respiratory Society statement: Update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am. J. Respir. Crit. Care Med. 2013, 188, 733–748. [Google Scholar] [CrossRef]
  94. Hyldgaard, C.; Bendstrup, E.; Wells, A.U.; Hilberg, O. Unclassifiable interstitial lung diseases: Clinical characteristics and survival. Respirology 2017, 22, 494–500. [Google Scholar] [CrossRef] [PubMed]
  95. Raghu, G.; Remy-Jardin, M.; Myers, J.L.; Richeldi, L.; Ryerson, C.J.; Lederer, D.J.; Behr, J.; Cottin, V.; Danoff, S.K.; Morell, F.; et al. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2018, 198, e44–e68. [Google Scholar] [CrossRef] [PubMed]
  96. Oldham, J.M. Interstitial lung disease: Perhaps unclassifiable, but not untreatable. Lancet Respir. Med. 2020, 8, 126–127. [Google Scholar] [CrossRef]
  97. Guler, S.A.; Ryerson, C.J. Unclassifiable interstitial lung disease: From phenotyping to possible treatments. Curr. Opin. Pulm. Med. 2018, 24, 461–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Collins, B.F.; Raghu, G. Antifibrotic therapy for fibrotic lung disease beyond idiopathic pulmonary fibrosis. Eur. Respir. Rev. 2019, 28. [Google Scholar] [CrossRef]
  99. Khanna, D.; Albera, C.; Fischer, A.; Khalidi, N.; Raghu, G.; Chung, L.; Chen, D.; Schiopu, E.; Tagliaferri, M.; Seibold, J.R.; 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, 43, 1672–1679. [Google Scholar] [CrossRef] [Green Version]
  100. Solomon, J.J.; Danoff, S.K.; Goldberg, H.J.; Woodhead, F.; Kolb, M.; Chambers, D.C.; DiFranco, D.; Spino, C.; Haynes-Harp, S.; Hurwitz, S.; et al. The Design and Rationale of the Trail1 Trial: A Randomized Double-Blind Phase 2 Clinical Trial of Pirfenidone in Rheumatoid Arthritis-Associated Interstitial Lung Disease. Adv. Ther. 2019, 36, 3279–3287. [Google Scholar] [CrossRef]
  101. Snetselaar, R.; Van Moorsel, C.H.M.; Kazemier, K.M.; Van Der Vis, J.J.; Zanen, P.; Van Oosterhout, M.F.M.; Grutters, J.C. Telomere length in interstitial lung diseases. Chest 2015, 148, 1011–1018. [Google Scholar] [CrossRef]
  102. Ley, B.; Torgerson, D.G.; Oldham, J.M.; Adegunsoye, A.; Liu, S.; Li, J.; Elicker, B.M.; Henry, T.S.; Golden, J.A.; Jones, K.D.; et al. Rare protein-altering telomere-related gene variants in patients with chronic hypersensitivity pneumonitis. Am. J. Respir. Crit. Care Med. 2019, 200, 1154–1163. [Google Scholar] [CrossRef] [PubMed]
  103. Molina-Molina, M. Telomere shortening is behind the harm of immunosuppressive therapy in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2019, 200, 274–275. [Google Scholar] [CrossRef] [PubMed]
  104. Newton, C.A.; Zhang, D.; Oldham, J.M.; Kozlitina, J.; Ma, S.F.; Martinez, F.J.; Raghu, G.; Noth, I.; Garcia, C.K. Telomere length and use of immunosuppressive medications in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2019, 200, 336–347. [Google Scholar] [CrossRef] [PubMed]
  105. Bejan-Angoulvant, T.; Naccache, J.M.; Caille, A.; Borie, R.; Nunes, H.; Ferreira, M.; Cadranel, J.; Crestani, B.; Cottin, V.; Marchand-Adam, S. Evaluation of efficacy and safety of rituximab in combination with mycophenolate mofetil in patients with nonspecific interstitial pneumonia non-responding to a first-line immunosuppressive treatment (EVER-ILD): A double-blind placebo-controlled randomize. Respir. Med. Res. 2020, 78, 100770. [Google Scholar] [CrossRef]
  106. Saunders, P.; Tsipouri, V.; Keir, G.J.; Ashby, D.; Flather, M.D.; Parfrey, H.; Babalis, D.; Renzoni, E.A.; Denton, C.P.; Wells, A.U.; et al. Rituximab versus cyclophosphamide for the treatment of connective tissue diseaseassociated interstitial lung disease (RECITAL): Study protocol for a randomised controlled trial. Trials 2017, 18, 1–11. [Google Scholar] [CrossRef] [Green Version]
Table 1. Randomized clinical trials with antifibrotics among patients with (progressive) fibrosing ILD.
Table 1. Randomized clinical trials with antifibrotics among patients with (progressive) fibrosing ILD.
Study/Phase
Study Identifier
Patients (n)
Duration (Weeks)
Criteria for Defining Fibrosis and ProgressionUse of Immunosuppressive AgentsPrimary Outcome
Published RCTs
Ssc-ILD
SENSCIS: Nintedanib in SSc-ILD [24]
Phase 3
(NCT02597933)
n = 576
52 weeks
- fibrosis: fibrosis affecting >10% of the lungs on HRCT
- progression:/
A stable dose of prednisone (<10 mg/day), MMF and/or MTX for at least six months before randomization was permitted.Annual rate of FVC decline
LOTUSS: Pirfenidone in SSc-ILD [99]
Phase 2
(NCT01933334)
n = 63
20 weeks
- fibrosis:/
- progression:/
Stable doses of oral CYC or MMF for at least three months before randomization was permitted. % patients with TE AEs or TE SAEs
PF-ILD
INBUILD: Nintedanib in PF-ILD [23]
Phase 3
(NCT02999178)
n = 663
52 weeks
- fibrosis: reticular abnormality with traction bronchiectasis with or without honeycombing, with disease extent of >10% on HRCT
- progression: one of the following criteria within 24 months before screening despite standard treatment:
• Relative decline in FVC%pred of ≥10%
• 2 elements of the following: relative decline in FVC%pred ≥5% but <10%, symptom worsening or an increasing extent of fibrosis on HRCT
Glucocorticoids <20 mg/day was permitted.Annual rate of FVC decline
RELIEF: pirfenidone in PF-ILD [25]
Phase 2
(DRKS00009822)
n = 127
48 weeks
- fibrosis: fibrotic lung disease on HRCT
- progression: annualized absolute FVC decline ≥5% within 6–24 months before screening
A stable dose of prednisolone (≤15 mg/day) or stable dose of other immunosuppressive therapy for at least three months before randomization was permitted.Absolute change in FVC%pred from baseline
Progressive uILD
pirfenidone in uILD
Phase 2
(NCT03099187)
n = 253
24 weeks
- fibrosis: fibrosis affecting ≥10% of the lungs on HRCT
- progression: One of the following criteria within six months before inclusion:
• absolute decline in FVC of > 5%pred
• significant symptomatic worsening not due to cardiac, pulmonary, vascular or other causes.
A stable dose of MMF for at least three months before randomization was permitted.FVC decline assessed by daily home spirometry during the 24-week study period
Planned and ongoing clinical trials in non-IPF F-ILD
CTD-ILD
SLS-III: Combining pirfenidone with MMF in SSc-ILD
Phase 2
(NCT03221257)
n = 150
18 months
- fibrosis:/
- progression:/
Prednisone <10 mg/day and the use of MMF is permitted if the responsible physician indicates that continued use is in the best clinical interest of the patient. Change in FVC%pred from baseline
Pirfenidone in SSc-ILD
Phase 3
(NCT03856853)
n = 144
52 weeks
- fibrosis:/
- progression:/
Treatment with prednisone <10 mg/day and stable doses of MMF and/or MTX for at least six months before inclusion is permitted. Relative change in FVC% from baseline
TRAIL-1: pirfenidone in RA-ILD
Phase 2 [100]
(NCT02808871)
n = 270
52 weeks
- fibrosis: reticular abnormalities affecting >10% of the lungs on HRCT
- progression:/
Prednisone <20 mg/day and a stable dose of immunosuppressive agents for at least three months before randomization is permitted.incidence of the composite endpoint of decline in FVC%pred of >10% or death
Pirfenidone in progressive ILD associated With Clinically Amyopathic Dermatomyositis
Phase 4
(NCT02821689)
n = 57
52 weeks
- fibrosis:/
- progression: increase in level of dyspnea and worsening of fibrosis on HRCT with >10% increase of HRCT score and/or absolute decrease in FVC%pred of >10% within 3–6 months after diagnosis.
Patients ever treated with biologicals, including basiliximab, were excluded. 12-month survival from the onset of ILD
Pirfenidone in DM-ILD
Phase 3
(NCT03857854)
n = 152
52 weeks
- fibrosis:/
- progression:/
Stable dose of prednisolone <15 mg/day for more than one month and of other immunosuppressive agents for more than three months before inclusion is permitted.Relative change from baseline of FVC
Sarcoidosis
PiRFS: Pirfenidone for Progressive Fibrotic Sarcoidosis
Phase 4
(NCT03260556)
n = 60
104 weeks
- fibrosis: fibrosis affecting >20% of the lungs on HRCT
- progression:/
A stable dose of prednisone <20 mg/day and other immunosuppressive agents for at least two months before inclusion is permitted.Time until clinical worsening
HP
Pirfenidone in Fibrotic HP
Phase: NA
(NCT02958917)
n = 44
52 weeks
- fibrosis: reticular abnormality, and/or traction bronchiectasis, and/or architectural distortion and/or honeycombing on HRCT
- progression:/
Not specified.Mean change from baseline in %FVC
F-ILD: fibrosing interstitial lung diseases; PF-ILD: progressive fibrosing ILD; SSc-ILD: systemic sclerosis-associated interstitial lung disease; cHP: chronic hypersensitivity pneumonitis; DM-ILD: dermatomyositis-associated interstitial lung disease; RA-ILD: rheumatoid arthritis-associated interstitial lung disease; uILD unclassifiable interstitial lung disease; MMF: mycophenolate mofetil; MTX: methotrexate; CYC cyclophosphamide; TE AEs: treatment-emergent adverse events; TE SAEs: treatment-emergent severe adverse events; HRCT: high-resolution computed tomography; FVC: forced vital capacity.
Table 2. Ongoing and unpublished randomized clinical trials with immunosuppressive agents in CTD-ILD.
Table 2. Ongoing and unpublished randomized clinical trials with immunosuppressive agents in CTD-ILD.
Study/Phase
Study Identifier
Patients (n)
Duration (Weeks)
Criteria for Defining CTD, ILD and Severity/ProgressionUse of Immunosuppressive AgentsPrimary Outcome
PULMORA
Effects of Tofacitinib vs. MTX on RA-ILD
Phase 4
NCT04311567
n = 48
24 weeks
- CTD: diagnosis of seropositive RA within 24 months prior to inclusion
- ILD: pulmonary abnormalities suggestive of RA-ILD
-severity/progression:/
Previous treatment with DMARDs is not allowed. History of prednisone use is allowed but should have been discontinued two weeks before baseline visit.Change in total interstitial disease score of pulmonary abnormalities by HRCT
APRIL
AbatacePt in RA-ILD
Phase 2
NCT03084419
28 weeks
n = 30
- CTD: diagnosis of RA
- ILD: ILD associated with RA with supportive findings on PFT and HRCT
- Progression: a decrease in FVC >5% within 24 months prior to inclusion or progression of lung fibrosis on HRCT as reported by a chest radiologist.
Treatment with other immunosuppressive agents, e.g., MMF—unless this has been discontinued with an adequate washout period—is not allowed. A stable dose of MTX and hydroxychloroquine for at least six weeks prior to baseline visit is permitted. Treatment with >10 mg prednisolone daily within six weeks or rituximab within 24 weeks prior to baseline visit is not allowed.Change in FVC
SCLEROCYCCYC
in SSc-ILD
Phase 3
NCT01570764
n = 40
52 weeks
- CTD-ILD: SSc-ILD
- Progression: worsening of ILD on HRCT and worsening of FVC and/or TLC ≥10% and/or worsening of DLCO ≥ 15% as compared to values obtained within 3–18 months preceding inclusion
Treatment with CS >15 mg/d during the last three months, CYC during the last 12 months or rituximab during the last six months prior to inclusion is not allowed. Treatment with MTX or MMF at inclusion is not allowed.Change in FVC
EvER-ILD
Evaluation of Efficacy and Safety of Rituximab With MMF in Patients With ILD [105]
Phase 3
NCT02990286
n = 122
26 weeks
- (CTD-)ILD: diagnosis of NSIP on histopathology or HRCT (basal predominant reticular abnormalities with traction bronchiectasis, peri-bronchovascular extension and subpleural sparing, frequently associated with ground-glass attenuation) and associated with differentiated CTD of IPAF or idiopathic ILD
- Progression: patients who did not respond, relapsed or were not able to continue at least one first-line immunosuppressive treatment of ILD. The absence of response was defined as either a decrease or an increase, but < 10% in FVC%pred.
Treatment with immunosuppressive agents other than CS (AZA, CYC, MTX, cyclosporine, tacrolimus, leflunomide) within two weeks prior to inclusion or IVIG, hydroxychloroquine or other monoclonal antibody therapies within six months prior to inclusion are not allowed. Change in FVC%pred
RECITALR
ituximab Versus CYC in CTD-ILD [106]
Phase2/3
NCT01862926
n = 116
48 weeks
- CTD-ILD: ILD associated with SSc, idiopathic interstitial myopathy and MCTD-severe and/or progressive ILD: intention of the caring physician to treat the ILD with IV CYC (deteriorating symptoms attributable to ILD, deteriorating PFTs, worsening gas exchange or extent of ILD) and when there is a reasonable expectation that immunosuppressive treatment will stabilize or improve CTD-ILD. In individuals with SSc, it is anticipated that subjects will fulfil the criteria for extensive disease defined by Goh et al.Immunosuppressive therapy (other than CS) received within two weeks prior to inclusion is not allowed. Previous treatment with rituximab and/or intravenous CYC is not allowed.Change in FVC
ATtackMy-ILD
Abatacept in Myositis-associated ILD
Phase 2
NCT03215927
n = 20
24 weeks
- CTD: diagnosis of anti-synthetase syndrome
- ILD: reticulation, honeycombing or ground glass opacities (GGO)
- Severe and/or progressive ILD (a) baseline FVC <80% or (b) FVC 80–100% with ≥10% decline in FVC in the last 12 months prior to inclusion
Inclusion criterium is the use of a stable dose of steroids, one of the other immunosuppressive agents (MMF or AZA) or a combination of steroid and an immunosuppressive agent. Other immunosuppressive agents, including MTX, cyclosporine, IVIG, tacrolimus, CYC or tofacitinib, are not allowed. Biologicals, i.e., rituximab, anti-TNF agents, tocilizumab, are not allowed.Change in FVC%pred
CATR-PAT
CYC and AZA vs. Tacrolimus in Anti-synthetase Syndrome-related ILD Phase 3 (open label) NCT03770663
n = 76
52 weeks
- CTD-ILD: ILD associated with anti-synthetase syndrome
- Moderate to severe ILD: FVC <80% and/or DLCO <70%
Previous use of CYC, AZA or tacrolimus in the last six months prior to inclusion is not allowed. Previous use of three daily IV steroids <3 months before inclusion is not allowed. Patients with worsening or relapse under prednisone >0.5 mg/kg/day are excluded. Progression-free survival
Basiliximab as a Treatment of Interstitial Pneumonia in Clinical Amyopathic Dermatomyositis Patients Phase 2NCT03192657n = 100
52 weeks
- CTD-ILD + severity/progression: Dermatomyositis and interstitial pneumonia and meeting at least two of four criteria: (1) interstitial pneumonia images on HRCT, (2) DLCO ≤ 60%, (3) elevated serum KL-6, (4) elevated serum anti-MDA5 (+).Previous application of immunosuppressive agents or any target treatment for dermatomyositis is not allowed.Survival
ILD: interstitial lung disease; CTD-ILD: connective tissue disease-associated ILD; NSIP: non-specific interstitial pneumonia; IPAF: idiopathic pneumonia with autoimmune features; SSc-ILD: systemic sclerosis-associated interstitial lung disease; RA-ILD: rheumatoid arthritis-associated interstitial lung disease; MCTD: mixed connective tissue disease; CS: corticosteroids; MMF: mycophenolate mofetil; MTX: methotrexate; DMARD: disease modifying anti-rheumatic drugs; CYC cyclophosphamide; AZA: azathioprine; HRCT: high-resolution computed tomography; PFT: pulmonary function test; FVC: forced vital capacity; DLCO: diffusing capacity for carbon monoxide; TLC: total lung capacity; IV: intravenous; IVIG: intravenous immunoglobulin.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Goos, T.; De Sadeleer, L.J.; Yserbyt, J.; Verleden, G.M.; Vermant, M.; Verleden, S.E.; Wuyts, W.A. Progression in the Management of Non-Idiopathic Pulmonary Fibrosis Interstitial Lung Diseases, Where Are We Now and Where We Would Like to Be. J. Clin. Med. 2021, 10, 1330. https://doi.org/10.3390/jcm10061330

AMA Style

Goos T, De Sadeleer LJ, Yserbyt J, Verleden GM, Vermant M, Verleden SE, Wuyts WA. Progression in the Management of Non-Idiopathic Pulmonary Fibrosis Interstitial Lung Diseases, Where Are We Now and Where We Would Like to Be. Journal of Clinical Medicine. 2021; 10(6):1330. https://doi.org/10.3390/jcm10061330

Chicago/Turabian Style

Goos, Tinne, Laurens J. De Sadeleer, Jonas Yserbyt, Geert M. Verleden, Marie Vermant, Stijn E. Verleden, and Wim A. Wuyts. 2021. "Progression in the Management of Non-Idiopathic Pulmonary Fibrosis Interstitial Lung Diseases, Where Are We Now and Where We Would Like to Be" Journal of Clinical Medicine 10, no. 6: 1330. https://doi.org/10.3390/jcm10061330

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop