Next Article in Journal
Special Issue “Computer-Aided Drug Discovery and Treatment”
Next Article in Special Issue
Functional MICA Variants Are Differentially Associated with Immune-Mediated Inflammatory Diseases
Previous Article in Journal
MiR-290 Family Maintains Pluripotency and Self-Renewal by Regulating MAPK Signaling Pathway in Intermediate Pluripotent Stem Cells
Previous Article in Special Issue
The Association between Inefficient Repair of DNA Double-Strand Breaks and Common Polymorphisms of the HRR and NHEJ Repair Genes in Patients with Rheumatoid Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development

by
Eunji Jeong
1,†,
Hyunseok Hong
2,3,†,
Yeon-Ah Lee
4 and
Kyoung-Soo Kim
1,3,5,*
1
Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
2
Yale College, Yale University, New Haven, CT 06520, USA
3
Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
4
Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea
5
East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(5), 2682; https://doi.org/10.3390/ijms25052682
Submission received: 22 January 2024 / Revised: 19 February 2024 / Accepted: 20 February 2024 / Published: 26 February 2024
(This article belongs to the Special Issue Molecular and Therapeutic Research in Rheumatoid Arthritis)

Abstract

:
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by swelling in at least one joint. Owing to an overactive immune response, extra-articular manifestations are observed in certain cases, with interstitial lung disease (ILD) being the most common. Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is characterized by chronic inflammation of the interstitial space, which causes fibrosis and the scarring of lung tissue. Controlling inflammation and pulmonary fibrosis in RA-ILD is important because they are associated with high morbidity and mortality. Pirfenidone and nintedanib are specific drugs against idiopathic pulmonary fibrosis and showed efficacy against RA-ILD in several clinical trials. Immunosuppressants and disease-modifying antirheumatic drugs (DMARDs) with anti-fibrotic effects have also been used to treat RA-ILD. Immunosuppressants moderate the overexpression of cytokines and immune cells to reduce pulmonary damage and slow the progression of fibrosis. DMARDs with mild anti-fibrotic effects target specific fibrotic pathways to regulate fibrogenic cellular activity, extracellular matrix homeostasis, and oxidative stress levels. Therefore, specific medications are required to effectively treat RA-ILD. In this review, the commonly used RA-ILD treatments are discussed based on their molecular mechanisms and clinical trial results. In addition, a computational approach is proposed to develop specific drugs for RA-ILD.

1. Introduction

Rheumatoid arthritis (RA) is an autoimmune disease with a systemic inflammatory response characterized by joint inflammation and swelling. Overactive immune systems caused by RA commonly result in extra-articular manifestations, such as cardiovascular and respiratory dysfunction [1]. Approximately 66% of patients with RA have lung involvement in the form of interstitial, airway, pleural, vascular, and parenchymal diseases [2,3]. The most prevalent form of lung involvement in patients with RA is interstitial lung disease (ILD), which is the chronic inflammation of the interstitial space that causes fibrosis and scarring of the lung tissue [4]. Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) constitutes 13% of the total deaths of patients with RA, and patients with RA-ILD show a significantly low 5-year survival rate of 35–39% [5]. Overall, patients with RA-ILD have a death risk three times higher than those with RA without ILD [6].
Pulmonary fibrosis is the fundamental cause of the high mortality and morbidity rates in patients with RA-ILD. The progression of fibrosis can be observed in wide fibrotic areas in the lungs using computed tomography scans with histological patterns, such as usual or nonspecific interstitial pneumonia (abbreviated as UIP and NSIP, respectively) [7]. Aggravated lung function caused by fibrosis results in a decline in forced vital capacity (FVC) and gas exchange (diffusing capacity of the lungs for carbon monoxide [DLCO]) while reducing exercise capacity and the health-related quality of life of patients [1]. If the condition worsens during successive fibrotic events, acute exacerbation, infection, or malignancy, death can occur.
The control of fibrosis is important for determining the prognosis of RA-ILD. However, predicting and diagnosing the onset of fibrotic phases in RA-ILD remain challenging. Genetic factors, such as the rs35705950 variant in the MUC5B gene and short telomere length, or environmental factors, such as smoking, sex, age, underlying diseases, and a high rheumatoid factor, increase the risk of ILD; however, a distinct correlation remains lacking [8,9]. Although genetic biomarkers, such as KL-6, SP-D, and HLA-DR2 alleles, or antibodies in the serum of patients with RA, such as anti-cyclic citrullinated or anti-citrullinated protein antibodies, were suggested as biomarkers to detect the onset of fibrosis, their detection accuracy is limited [10]. Owing to difficulties in identifying the emergence of fibrosis, the rudimentary inhibition of immune responses and hindrance of fibrosis progression are of vital importance in RA-ILD treatment.
Currently, the first-line treatment for RA-ILD primarily relies on drugs used for idiopathic pulmonary fibrosis (IPF) because the two diseases share similar clinical, radiological, and molecular patterns. Two major drugs, namely, pirfenidone and nintedanib, are FDA-approved anti-fibrotic agents for IPF that are commonly used as treatments for RA-ILD. In the case of nintedanib, the treatment recently received FDA approval as the first treatment for chronic fibrosing ILD with a progressive trait, supporting its use in RA-ILD. These anti-fibrotic drugs can directly interfere with specific signaling pathways to block or slow the progression of fibrosis and reverse the morphological changes induced by fibrosis. Several immunosuppressants and disease-modifying antirheumatic drugs (DMARDs), which are the primary treatments for RA, were also shown to slow fibrotic progression in RA-ILD by reducing the autoimmune response, which is the causative factor of pulmonary fibrosis.
However, the limitations of current drugs and the need for basic inhibition of the autoimmune-induced fibrosis phase necessitate the development of new drugs for RA-ILD. While continuous research and development is in progress, drug repositioning is one of the primary strategies to be explored. Because multi-organ fibrotic diseases share similar clinical and pathological characteristics, these drugs can be repurposed to benefit the fibrotic manifestation of RA-ILD. With adequate targets that can hinder the progression of fibrosis in RA-ILD and a set of drug candidates, computational analysis can aid and accelerate the process by predicting the drug–target interactions (DTIs) between them to recommend favorable pairs for further confirmation.

2. Anti-Fibrotic Agents for ILD

To date, the most widely used drugs for the treatment of RA-ILD are pirfenidone and nintedanib, which were initially developed and approved by the Food and Drug Administration (FDA) for IPF. Their anti-fibrotic and anti-inflammatory potentials stem from the strong resemblance of RA-ILD and IPF in terms of pulmonary fibrotic patterns and prognosis. RA-UIP, which is a prevalent subtype of RA-ILD, is considered the most analogous to IPF because both show identical UIP fibrotic patterns and similar prognoses. RA-ILD and IPF are associated with pulmonary fibrosis and acute exacerbations, respectively. In addition, the upregulation of several genetic markers, such as eotaxin, IL-8, MMP-2, MMP-7, MMP-9, and Flt-3L, and genetic mutations in TERT, RTEL1, PARN, and SFTPC were observed in patients with RA-ILD and IPF [11,12]. These similarities support the implementation of therapeutic strategies for IPF in patients with RA-ILD.
However, RA-ILD and IPF differ in several aspects that can cause adverse effects during treatment. RA-ILD is a multi-system disease that stems from an autoimmune response. In contrast, IPF includes only lung-limited fibrosis without a specific cause. Therefore, their responses to treatment may differ based on their contrasting pathogenic pathways. For instance, an immunosuppressive approach to IPF therapy is contraindicated due to its inefficiency and increased mortality and morbidity [13]. However, immunosuppression is the standard of care for patients with RA-ILD because the disease is derived from an autoimmune disorder [14]. Moreover, while IPF exclusively presents UIP histological patterns in pulmonary fibrosis, RA-ILD can include other patterns, such as NSIP, organizing pneumonia (OP), and mixed or indeterminate forms [15]. Different histological patterns affect the disease prognosis and treatment effectiveness. RA-ILD with NSIP or OP responds better than RA-UIP to immunosuppressive treatment while also showing a preferable disease prognosis [16,17]. Thus, while the similarities between RA-ILD and IPF can guide the discovery of novel treatments for RA-ILD, their differences should be considered in the process of drug development. This section provides an overview of the anti-fibrotic mechanisms and clinical trials of pirfenidone and nintedanib in patients with RA-ILD.

2.1. Nintedanib

Nintedanib is an FDA-approved anti-fibrotic agent used for the treatments of IPF, systemic sclerosis-ILD, and chronic fibrosing ILD with a progressive phenotype, regardless of the underlying cause [18]. Using nintedanib for the treatment of RA-ILD has attracted increased attention. Nintedanib functions as a tyrosine kinase inhibitor (TKI) for several receptors that contribute to fibrogenesis, including PDGFR α and β, FGFR 1–3, vascular endothelial growth factor receptor (VEGFR) 1–3, colony-stimulating factor 1 receptor (CSF1R), and lymphocyte-specific tyrosine protein kinase (Lck) [19]. Thus, it exerts its anti-fibrotic effect via a multifaceted inhibitory mechanism. It also regulates the activity of Lck, which is essential for T cell proliferation and the release of pro-fibrotic mediators, such as IL-2, IL-4, IL-5, IL-10, IL-12p70, IL-13, and interferon gamma (IFN-γ) [20,21]. By specifically targeting PDGF, FGF2, VEGF-A, and other growth factors, nintedanib suppresses the function of fibrocytes, consequently inhibiting fibrocyte migration, differentiation, and proliferation [22]. The study also mentioned the ability of nintedanib to suppress transforming growth factor-beta (TGF-β)-induced fibroblast–myofibroblast transition and TGF-β-stimulated collagen secretion [22]. In pulmonary fibrosis, nintedanib significantly decreases certain markers that indicate M2 macrophage polarization. M2 macrophage polarization is a major contributor to pulmonary fibrogenesis because it releases fibrotic factors, such as TGF-β and gelectin-3, which accordingly stimulate the epithelial–mesenchymal transition (EMT), endothelial–mesenchymal transition (EndoMT), and fibroblast proliferation [23,24].
Several clinical trials were conducted using nintedanib in patients with RA-ILD. The INBUILD trial was a phase 3 clinical trial conducted to evaluate the efficacy of nintedanib in patients with progressive fibrosing ILD [25]. Of the 663 patients enrolled in the trial, 89 had RA-ILD, and a retrospective subgroup analysis was performed [26]. Compared with the placebo, nintedanib demonstrated an improvement in patients with RA-ILD by significantly lowering the average rate of FVC decline by 59% over 52 weeks and reducing the risk of acute exacerbation or death. The most common adverse effect associated with nintedanib was diarrhea, which occurred in 61.9% of patients in the nintedanib group; however, the symptoms were still manageable. In addition, its concomitant use with other DMARDs showed no differences in effectiveness or safety profile. Thus, nintedanib demonstrated sufficient efficacy and safety as a potential treatment option for patients with RA-ILD. A recent retrospective cohort study from Portugal suggested the importance of the early initiation of nintedanib treatment in halting ILD progression. Patients treated with nintedanib within 3 years of their initial ILD diagnosis exhibited significantly improved respiratory function within 24 months of treatment [27].
Nintedanib also affects arthritis in murine models. In a zymosan-induced arthritis SKG mouse model, nintedanib treatment reversed joint swelling and decreased the arthritis score compared with a placebo saline treatment. Moreover, while the talus and calcaneus bones of the nintedanib-treated mice were negligibly changed, those of the saline-treated mice were irregular and deformed, suggesting severe erosion [28]. Based on these in vivo results, future clinical trials are required to determine the efficacy of nintedanib against arthritis involvement.

2.2. Pirfenidone

Pirfenidone has been FDA-approved for IPF treatment since 2014. Phase 3 trials, such as ASCEND in 2014 and CAPACITY 1 and 2, supported its efficacy and safety for IPF treatment as an anti-fibrotic agent. Thus, several attempts have been made to use pirfenidone to treat diseases similar to pulmonary fibrosis, including RA-ILD.
Pirfenidone is a pyridine derivative with anti-fibrotic, anti-inflammatory, and anti-oxidant properties, which collectively contribute to the inhibition of pulmonary fibrosis. The anti-fibrotic activity of pirfenidone originates from suppressing TGF-β1, which is a master regulator of fibrosis. Through TGF-β1, pirfenidone reduces fibroblast proliferation, myofibroblast differentiation, extracellular matrix (ECM) production, and TGF-β1-induced SMAD3 phosphorylation [29]. Anti-inflammatory effects of pirfenidone can be observed via a translational level decrease of inflammatory cytokines, such as TNF-α, IL-6, and IFN-γ, and the upregulation of anti-inflammatory cytokines, such as IL-10 [30]. By controlling the balance between pro-inflammatory cytokines, pirfenidone regulates tissue injury and the subsequent restoration process of wound healing. Regulating oxidative stress is also crucial for pulmonary fibrosis treatment because reactive oxygen species participate in the diverse molecular mechanisms of fibrogenesis [31]. By eliminating hydroxyl radicals and controlling oxidative stress, pirfenidone establishes itself as a potent anti-fibrotic agent [32].
In addition to its strong anti-fibrotic capacity, pirfenidone has been repurposed as an antirheumatic drug. Gan et al. [33] demonstrated a reduction in joint synovial inflammation and swelling in a collagen-induced arthritis rat model treated with pirfenidone. In vitro and bioinformatic analyses supported the antirheumatic effect of pirfenidone via the inhibition of inflammatory cytokines and cell migration, rendering it more suitable for RA-ILD treatment.
Multiple clinical trials demonstrated the promising potential of pirfenidone for the treatment of RA-ILD. Recently, a phase 2 trial (TRAIL-1) using pirfenidone in patients with RA-ILD was attempted but was terminated early due to the COVID-19 pandemic. However, certain data indicated that pirfenidone was effective in the treatment of RA-ILD. Pirfenidone lowered the rate of FVC decline compared with the placebo without presenting any new adverse effects. In addition, this study demonstrated that pirfenidone combined with DMARD therapy had an acceptable safety profile and tolerability [34]. The RELIEF trial, which was another phase 2 study with 127 non-IPF patients with ILD, included 17 patients with RA-ILD. In the trial, patients treated with pirfenidone experienced a lower decline in FVC, indicating decelerated pulmonary aggravation, compared with those who received the placebo, showing that pirfenidone alleviated RA-ILD symptoms [35].
A prospective controlled cohort study was conducted to investigate the efficacy and safety of pirfenidone combined with immunosuppressant therapy in patients with connective-tissue-disease-associated interstitial lung disease (CTD-ILD). Of the 111 patients, 17 were diagnosed with RA-ILD. While all patients in the study were prescribed immunosuppressants, those in the test group concomitantly received pirfenidone. Pirfenidone-treated patients with RA-ILD exhibited an enhancement in DLCO%, which is a measure of gas exchange, by 7.40% compared with the baseline, while the control group showed a 5.5% decrease [36].
A phase 2 study of pirfenidone for the treatment of unclassifiable progressive fibrosing ILD was also conducted. Although patients diagnosed with RA-ILD were not included in the study population, pirfenidone enhanced pulmonary function and exercise capacity compared with a placebo without presenting extensive adverse effects [37]. When an ongoing randomized controlled trial for patients with CTD-ILD with a subpopulation of patients with RA-ILD is completed as expected, we will obtain a clearer picture of the potential of pirfenidone for RA-ILD treatment [38].

3. Conventional Synthetic DMARDs for RA-ILD

Immunosuppressants are early-stage drugs used to treat patients with RA. Immunosuppressive agents can hinder the progression of RA to RA-ILD by reducing the autoimmune response that is responsible for lung fibrosis (Figure 1). Although there are currently no FDA-approved immunosuppressants for improving pulmonary function in patients with RA-ILD, several drugs that have shown efficacy against similar or related diseases are being tested. Thus, several immunosuppressants should be considered as plausible treatments for RA-ILD.

3.1. Methotrexate (MTX)

MTX, which is a folate derivative, is considered the first-line DMARD for RA. However, its use for RA-ILD treatment is limited owing to MTX-related pulmonary adverse effects, including hypersensitivity pneumonitis, that are presumed to be associated with RA-ILD progression [39]. Several studies have since overturned the original beliefs of the clinicians. In a retrospective multicenter study conducted by Kiely et al. [40], MTX exposure did not result in RA-ILD but rather delayed its onset. Juge et al. [41] further advocated for the positive effect of MTX using a case–control study that revealed a lower risk of RA-ILD in MTX ever-users compared with MTX never-users, with an average delay of 3.6 years in ILD detection. MTX was also suggested to be a protective factor against lung function decline and mortality in cohorts of patients with RA-ILD when provided after RA-ILD incidence [42]. A phase 4 randomized controlled trial (PULMORA) is in progress to compare the effects of MTX with those of the Janus kinase (JAK) inhibitor tofacitinib [43]. Through this clinical trial, we expect to gain a better understanding of the effects of MTX on RA-ILD.

3.2. Cyclophosphamide (Cyc)

Cyc is a DNA alkylating immunosuppressive agent that hydrolyzes into phosphor mustard when activated, consequently resulting in DNA damage and the inhibition of cell proliferation [44]. Although the underlying mechanism is not fully understood, the immunosuppressive effect of Cyc was shown to benefit patients with RA-ILD [45]. A 10-year retrospective study in Germany by Schupp et al. [46] demonstrated the ability of Cyc to stabilize lung function in patients with both inflammatory and fibrotic ILD. Randomized controlled trials also support the ability of Cyc to improve lung conditions in patients with CTD-ILD with better FVC results compared with a placebo [47,48]. Cyc also relieved tender and swollen joints in patients with RA, which confirms its antirheumatic effects [49]. However, certain studies have reported that the mortality rate of patients with acute exacerbation of RA-ILD treated with Cyc did not significantly differ from that of the control group [50]. Moreover, the administration of Cyc alone is not recommended because of its high toxicity and adverse effects [4].

3.3. Mycophenolate Mofetil (MMF)

MMF is less toxic than Cyc and inhibits inosine-5′-monophosphate dehydrogenase and de novo guanine syntheses, which are critical elements for DNA synthesis and the proliferation of T and B lymphocytes [51]. In addition to its immunosuppressive effects, studies supported the anti-fibrotic capability of MMF by suppressing fibroblast proliferation and the expression of fibrosing cytokines, such as TGF-β and IL-6 [52,53]. Because MMF can control both the inflammatory response and pulmonary fibrosis, it is considered a promising option for the treatment of RA-ILD. Although controlled studies of MMF for RA-ILD are still needed, MMF has shown positive results in several studies, with improved or stabilized pulmonary physiology in patients with CTD-ILD, including subgroups of patients with RA-ILD [54,55]. While the frequent adverse events of MMF include gastrointestinal symptoms, bone marrow suppression, and infection due to immunosuppression, it remains an appealing option compared with the safety profiles of other immunosuppressive drugs [56]. However, MMF is known to have no benefit in treating the articular manifestations of RA [57]. Thus, it is recommended to be used in combination with a different immunosuppressant that can target joint involvement.

3.4. Azathioprine (AZA)

AZA is an antimetabolite that acts as an immunosuppressive agent through the inhibition of purine synthesis and DNA replication of lymphocytes [58]. AZA already demonstrated its advantage on pulmonary function for similar ILDs, such as systemic-sclerosis-associated interstitial lung disease (SSc-ILD), sarcoidosis, and chronic hypersensitivity pneumonitis [59,60,61]. Therefore, AZA is commonly used along with other immunosuppressants for early RA-ILD treatment. A multicenter retrospective cohort study performed by Matson et al. [62] supported the notion of using AZA for RA-ILD. A total of 212 patients with RA-ILD on immunosuppressive therapy were enrolled in the study, including 92 patients who were treated with AZA. Regardless of the type of immunosuppressive agents, patients treated with immunosuppression exhibited a 3.9% increase in FVC and a 4.5% increase in DLCO compared with the predicted value if the pretreatments had continued. Also, a retrospective study of 56 patients with CTD-ILD treated with AZA concluded that AZA can stabilize or improve CTD-ILD as long as the patient can tolerate the drug [63]. However, there are also concerns about AZA’s safety profile. PANTHER-IPF trial was a randomized, double-blind, placebo-controlled trial for IPF with concomitant treatment using prednisone, AZA, and N-acetylcysteine, but it was terminated early due to a high mortality rate [13]. Oldham et al. also noted that while AZA and MMF exhibited comparable efficacy on pulmonary stability, AZA had a higher rate of adverse effects and drug intolerance when treating CTD-ILD. [64]. Therefore, the prescription of AZA to patients, especially with RA-UIP, which is a subtype of RA-ILD similar to IPF, should be done with caution.

3.5. Tacrolimus (TAC)

Tacrolimus is a type of calcineurin inhibitor that inhibits calcium-dependent events, including T lymphocyte signal transduction and IL-2 transcription [65]. There have been several attempts to treat ILD using TAC. For instance, a retrospective study was conducted with 49 patients with ILD associated with polymyositis or dermatomyositis [66]. In the study, the patients treated with TAC along with conventional therapy showed significantly longer event-free and disease-free survival compared with the group only treated with conventional therapy, suggesting the potential of TAC to improve the prognosis of ILD. Similar research was done with antisynthetase-associated ILD. A cohort study of patients with antisynthetase-associated ILD revealed that 13 out of 15 patients who received a calcineurin inhibitor showed stable or improved FVC [67]. To gain a better understanding of TAC’s efficacy against CTD-ILD, Yamano et al. [68] conducted a retrospective study of patients with CTD-ILD who received combination therapy of TAC and corticosteroid. Among the study population of 28 patients, 11 patients were diagnosed with RA-ILD. After 1 year of TAC therapy, patients showed significantly improved lung physiology and exercise capacity without severe adverse effects. Recently, a Korean RA-ILD cohort also revealed that tacrolimus delayed the progression of RA-ILD compared with a non-treated group, but with non-statistical significance [69].

4. Biologic DMARDs

Antibodies can disable specific cells and the immune system, primarily during organ rejection or in autoimmune diseases (Figure 1). Rituximab (RTX) is an anti-B-cell antibody widely used to alleviate joint inflammation in RA. Recently, clinical trials showed their auxiliary efficacy, not only in joint inflammation but also in delaying fibrosis. Anti-tumor necrosis factor alpha (TNF-α) agents have limited clinical evidence against RA-ILD, but are widely used in RA treatments.

4.1. Rituximab (RTX)

RTX is a chimeric monoclonal antibody that targets the CD20 surface marker on both B cell precursors and mature B cells [70]. RTX binding can result in B cell depletion via several mechanisms, including complement-dependent cytotoxicity, cell-mediated cytotoxicity, and apoptosis [71]. With its anti-arthritic effect, RTX treatment showed clinically significant improvements with articular involvement and was approved by the FDA as a biologic DMARD for RA in the REFLEX and DANCER trials [72,73,74]. Promising results were also observed in the lung function of patients with CTD-ILD and RA-ILD, showing improved or maintained pulmonary function and slower progression of the disease [75,76,77]. Recently, a randomized controlled trial was conducted to compare the efficacy of RTX and Cyc in patients with CTD-ILD (RECITAL). This study demonstrated the ability of RTX and Cyc to improve the pulmonary function and quality of life of patients without significant differences, with RTX causing fewer adverse effects [48].

4.2. TNF Inhibitors

Although few clinical studies have been conducted, three TNF-α inhibitors, namely, infliximab, adalimumab, and etanercept, exhibit positive effects on patients with RA-ILD. The intravenous infusion of infliximab showed the radiographic stabilization of pulmonary functions, such as dyspnea, cough, and exercise tolerance, and improvement in joints [78]. However, a retrospective study of 100 patients with RA-ILD demonstrated that the anti-TNF-α therapy of adalimumab and infliximab should be used with caution in older patients with RA-ILD because lung complications can occur within months of initial anti-TNF-α treatment [79]. Etanercept was also suggested to be used with caution because the exacerbation of ILD was observed in a specific case [80].

4.3. Abatacept (ABA)

ABA is a co-stimulation blocker comprising the extracellular domain of human cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) and a modified Fc portion of human immunoglobulin G1. By binding to CD80/CD86, ABA hinders the CD80/86-CD28 interaction, consequently inhibiting co-stimulatory signals essential for T cell activation [81]. Although ABA is an FDA-approved drug for the treatment of RA, studies showed that it also has favorable effects on the treatment of RA-ILD. In vivo studies demonstrated the potency of ABA in inhibiting lung inflammation and fibrosis in murine models [82,83]. In an observational study of 263 patients with RA-ILD, approximately 80% of patients treated with ABA maintained stable or improved pulmonary function with better FVC and DLCO, as well as a reduced DAS28esr score, indicating RA disease activity [84]. Other studies and systematic reviews also supported the efficacy and safety of ABA in patients with RA-ILD [85,86,87]. Although it was terminated because of the COVID-19 pandemic, a phase II study of ABA in patients with RA-ILD (APRIL) indicated an acceptable safety profile with no serious adverse reactions [88]. Thus, randomized controlled trials are necessary to further investigate the potential of ABA as a treatment for RA-ILD.

4.4. Tocilizumab (TCZ)

Tocilizumab is an FDA-approved IL-6 inhibitor for SSc-ILD treatment based on the focuSSced and faSScinate trials [89,90]. As SSc-ILD and RA-ILD share similarities as members of the CTD-ILD family, tocilizumab is expected to demonstrate similar efficacy toward RA-ILD. Several studies strengthened this idea with positive results. In a national multicenter retrospective study from Italy, 19 out of 28 with RA-ILD showed improved or stabilized FVC and DLCO after TCZ therapy without significant adverse effects [91]. Furthermore, in the study conducted by Otsuji et al. [92], 34 patients with RA-ILD were treated with TCZ for 6 months to evaluate the safety and efficacy of the treatment. Patients administered with TCZ displayed a significant decrease in both MMP-3 and KL-6, indicating the efficacy of the drug toward both RA and ILD. Patients’ chest CTs also indicated the attenuation of ILD progression after TCZ treatment.

5. Corticosteroid (Glucocorticoid)

Glucocorticoids, also known as corticosteroids or steroids, are conventional treatments for autoimmune diseases. They function via multiple mechanisms, including blocking the synthesis of cytokines and cell surface molecules necessary for the activation of the immune system via the inhibition of NF-κB [93]. Owing to their anti-inflammatory and immunosuppressive effects, glucocorticoids are favored for treating inflammatory RA-ILD with NSIP or OP [94,95]. Positive results were also observed with steroid treatments for fibrotic RA-ILD with UIP. In a retrospective study conducted by Song et al. [96], approximately 50% of patients with RA-UIP showed improvement or stabilization following glucocorticoid treatment. In contrast, glucocorticoid treatment in patients with IPF resulted in higher morbidity and mortality; therefore, it is contraindicated in IPF [97,98]. Despite the high histopathological similarity between IPF and RA-ILD, the same treatment might have disputed pharmacological effects. Moreover, the use of glucocorticoids in patients with RA-ILD carries potential risks, such as an increased risk of infection or osteoporosis development; therefore, additional caution is needed [99,100].

6. Targeted Synthetic DMARDs (JAK Inhibitors)

JAKs are a group of intracellular tyrosine kinases that participate in signaling pathways that regulate several physiological cell processes, including differentiation and metabolism [101]. Transcription factors, called STATS, which is an oncoprotein family encoded by tumor-associated genes, jointly form a signaling pathway with JAK. JAK and STATS have several subtypes, but JAK2/STAT3 is the most prevalent in ILD [10,102,103]. STAT3 transduces peptide hormone signals from the cell surface to the nucleus and is activated by cytokines, growth factors, and several peptide hormones. While RA-ILD with UIP shares almost identical clinical patterns with IPF, RNA transcriptomic data show that RA-UIP has a nuclear form of phosphorylated JAK2, whereas most patients with IPF have a cytoplasmic form of phosphorylated JAK2 [104]. Such variations imply that applying therapeutic approaches for IPF in patients with RA-ILD may not yield the same effects and may even cause unexpected adverse effects. The JAK/STAT pathway is activated by several pro-fibrotic or pro-inflammatory cytokines, such as IL-6, IL-11, and IL-13, or growth factors, such as platelet-derived growth factors (PDGFs), TGF-β1, and fibroblast growth factors (FGFs) [102]. Blocking these signals inactivates the JAK/STAT pathway, which consequently arrests fibrosis. JAK inhibitors also exhibit anti-fibrotic effects by directly affecting fibroblast activation [105].
Tofacitinib, ruxolitinib, baricitinib, and upadacitinib are FDA-approved JAK inhibitors used for the treatment of RA [106,107,108,109]. They show significant improvement in joints by controlling the plasma levels of pro-inflammatory cytokines, reducing joint destruction, and exerting anti-fibrotic effects by directly affecting fibroblasts. Research on macrophages has revealed that they can be effective in controlling diseases characterized by concomitant inflammation and fibrotic manifestations, which also supports their efficacy against RA-ILD [96,97]. Figure 2 summarizes the treatment options of RA-ILD we have reviewed so far, including JAK inhibitors.

6.1. Tofacitinib

Tofacitinib is a JAK 3/2 inhibitor with strong regulatory effects on IL-17A. IL-17A is secreted by T helper 17 cells and functions cooperatively with IL-17RA and IL-17RC during fibrosis. Large areas of IL-17RA-induced fibroblast accumulation and fibrosis were observed in patients with RA-ILD. IL-17A plays a distinct role in RA-ILD compared with other normal lung diseases or even IPF [110]. The suppression of IL-17A ameliorates the activity of inflammatory T helper 17 cells, thereby alleviating fibrosis. In vivo research using SKG mice showed that tofacitinib slowed the progression of RA-ILD by increasing the number of myeloid-derived suppressor cells [111]. A phase 3 randomized controlled clinical trial indicated that 5 and 10 mg of tofacitinib administered twice a day demonstrated maximal healing effects at 6 months. Signs, symptoms, and pain caused by RA were reduced, physical lung function improved, and the progression of fibrosis or structural damage decreased. However, 28% of the patients discontinued the treatment because of adverse events [112]. Tofacitinib can be applied either alone or in combination with immunosuppressants, such as MTX, without affecting its efficacy [113]. For instance, tofacitinib was administered to a 52-year-old man after signs of ILD were diagnosed without immunosuppressants. Tofacitinib successfully controlled frequent inflammation and improved respiratory functions [114]. Multiple studies showed that tofacitinib retains or slows the progression of RA-ILD, reduces the risk of RA leading to ILD, and exceeds other biological DMARDs (bDMARDs) in restoring pulmonary function. The administration of tofacitinib over an 8–12 month period to two patients with RA-ILD successfully controlled inflammation in the joints without exacerbations or hospitalizations due to lung disease [115]. In another study, among 31 patients with RA-ILD, 13 were treated with tofacitinib, and 83.9% exhibited overall stability or improvement in RA-ILD symptoms [116]. Tofacitinib exceeded other bDMARDS in reducing the risk of ILD, and another post hoc analysis of 21 clinical trials supported this finding by concluding that the incidence rate of ILD following tofacitinib treatment was extremely low (0.18).

6.2. Ruxolitinib

Ruxolitinib, which is a specific JAK2 inhibitor, regulates the activity of pro-fibrotic M2 and pro-inflammatory M1 macrophages [117]. IL-13- and IFN-γ-activated macrophages followed a signaling pathway that involved JAK2, to which ruxolitinib indicated direct repression. Polarization markers, such as MHC II and Toll-like receptor 4 (TLR4), which indicate the differentiation processes of M1 and M2, respectively, were decreased, as well as the secretion of pro-inflammatory cytokines, such as CXCL10, IL-6, and TNF-α [117].
Although clinical trials of ruxolitinib for patients with RA-ILD are absent, few case reports support ruxolitinib’s anti-fibrotic capacity for ILD. In the case report by Manuel et al. [118], ruxolitinib administration improved the pulmonary function of two patients with ILD from STAT3 gain-of-function mutation without major adverse events. The administration of ruxolitinib showed similar CT scan changes and improvement in clinical symptoms, while the FEV rates significantly increased. Another case study suggested ruxolitinib’s efficacy and safety in treating a patient with refractory systemic idiopathic juvenile-arthritis-associated ILD [119]. Ruxolitinib was well tolerated and when concomitantly administered with corticosteroid, the corticosteroid administration dosage tapered.

6.3. Baricitinib

Baricitinib is a JAK1/JAK2 inhibitor suggested as a treatment for patients with RA-ILD who do not respond to the initial treatment with MTX [120]. An analysis of 3770 patients with RA from eight randomized clinical trials and one long-term extension study on baricitinib showed promising effects in lowering the risk of ILD development [121]. When 11 patients with RA and 4 patients with RA-ILD were treated with baricitinib for 6 months, a considerable decrease in inflammation was observed in patients with RA-ILD via a reduction in KL-6, which is a biomarker of RA-ILD. Moreover, improvements in DLCO and diffusion coefficient percentages confirmed that baricitinib restored pulmonary function [122].

6.4. Upadacitinib

Upadacitinib is a second-generation JAK inhibitor selective for the JAK1 subtype. A patient with RA-ILD usually receives initial treatment with a csDMARD, mostly MTX monotherapy. However, when the patient is resistant to MTX, a combination of MTX with other csDMARDs or bDMARDs is recommended [123]. tsDMARDs, especially updacitinib, are recommended when a patient still does not show an adequate response to a combination of MTX and TNF inhibitor, which is a type of bDMARD [124]. A recent case report from Nishii et al. illustrated a case of a 69-year-old man with RA-ILD who previously did not respond to several treatments including MTX, TAC, ABA, and Cyc. However, upadacitinib therapy improved the patient’s pulmonary condition, which showed better DLCO and FVC. These results were consistent with a retrospective study of 43 patients with RA-ILD in Italy [125]. All three patients who were treated with upadacitinib exhibited stable high-resolution computed tomography (HRCT) results of pulmonary fibrosis. However, due to the insufficiency of clinical data, further investigations are anticipated to confirm the suitability of upadacitinib as an RA-ILD treatment.

7. Computational Methods for Further RA-ILD Drug Development

To prevent fibrosis and the progression of RA-ILD, and because there are limitations in applying IPF treatment to patients with RA-ILD, a novel drug for RA-ILD is necessary. However, considering that diseases with fibrotic characteristics are numerous and that their mechanisms share common molecules and genes, another solution for RA-ILD drug development could be drug repositioning or repurposing.
The first basis for drug repositioning is the phenotypic similarity between diseases, which has already been confirmed by studies that demonstrated shared molecules, genes, and signaling pathways between multiple organ fibrotic diseases [126]. Thus, computational drug repositioning is a favorable approach for identifying additional drugs with positive effects on RA-ILD. The drugs currently suggested for RA-ILD treatment also follow this notion because they were initially implemented for IPF or other CTD-ILDs.
Although they were demonstrated only by in vitro assessments, certain clinical drugs for distinct diseases show potential for RA-ILD treatment. Cyclosporine, which is a well-known immunosuppressant, downregulates immune- and fibrosis-related genes, such as insulin-like growth factor binding protein 2 (IGFBP2), inhibitor of DNA binding protein 1 (ID1), and peroxisome proliferator-activated receptor gamma (PPARγ) [127]. Theophylline, which is a chronic obstructive pulmonary disease and asthma drug, is a pan-phosphodiesterase inhibitor that exhibits anti-inflammatory and anti-fibrotic effects by interfering with the TGF-β1 signaling pathway [128]. Fluvoxamine, which is a well-known antidepressant drug, showed equivalent anti-fibrotic effects with only one-third the dosage of pirfenidone [129]. These results suggested that multiple drugs that have already been approved for clinical safety can be used to control fibrosis.
For drug repositioning, potential targets that can effectively impede disease progression must be identified, as discussed in the following section. A computational platform for specifying the optimal targeting drug is introduced in the next section. In the forthcoming therapeutic development, the limitations of implementing RA-ILD in animal models and the great variety of patients with RA-ILD render the computational approach a more favorable option than traditional methods (Figure 3).

7.1. Molecular Targets Suggested for RA-ILD Fibrosis

Fibrosis is regulated by the transition, migration, and metabolic activity of fibroblasts and myofibroblasts. Excessive immune reactions in the lung tissue trigger cytokines and chemokines to be released and fibroblasts to be activated, which then differentiate into α-smooth-muscle-actin-expressing myofibroblasts. The invasive nature of pulmonary myofibroblasts stimulates cytoskeletal synthesis and upregulates the expression of contractile proteins. This contracts the lung tissue and substantial numbers of ECM proteins are produced [130,131,132,133].
Multiple studies suggested three molecular targets that can restrict fibroblast activity: TGF-β, CTGF, and MMP. These three targets inhibit and stimulate pulmonary fibrosis via multiple mechanisms and complex interactions. Genetic analysis demonstrated that they are deeply involved in multiple fibrotic diseases in different organs (eyes, heart, lungs, pancreas, and kidneys) [131,134].
TGF-β is a multifunctional cytokine secreted by bronchial structural cells or inflammatory cells that maintain tissue homeostasis. It causes the transition of epithelial and endothelial cells to mesenchymal cells (EMT and EndoMT), activates pericytes and smooth muscle cells related to blood vessels, and induces ECM deposition. Targeting TGF-β hinders signaling pathways that include SMAD 2/3, Wnt/β-catenin, MAPK, and PI3K [135,136,137]. The complex interplay between TGF-β and CTGF has been reported in pulmonary and renal fibrosis [131]. Different subtypes of MMPs have dual-purpose properties in pulmonary fibrosis. Dismantling specific collagen via MMP activation may delay fibrotic progression, whereas regulating MMP activity with a tissue inhibitor of metalloproteinase reduces inflammatory effects. Among these, MMP-1, -2, and -9 are marked with pulmonary fibrosis [134]. Targeting MMP-1 controls the local epithelial–mesenchymal interaction, MMP-2 plays a significant role in the EMT via the Wnt/β-catenin signaling pathway, and MMP-9 enhances recovery by decreasing IGFBP-3 expression [138,139,140].
Additional target molecules, including Syndecan-2, which promotes internalization and the degradation of the TGF-β receptor, are related to citrullinated peptides found in more than half of patients with RA and to the prevalence and exacerbation of ILD in patients with RA [141,142,143]. Additional targets can be found in other CTD-ILDs, including various autoimmune diseases, such as systemic lupus erythematosus, RA, Sjogren’s syndrome, polymyositis/dermatomyositis, systemic sclerosis (SSc), and mixed connective tissue disease invading the interstitial compartment of the lungs [144]. TGF-β, PDGF, and FGF are key growth factors that participate in the fibrotic process of most CTD-ILD diseases. In particular, the FGF and PDGF receptors and downstream signaling proteins, such as YAP/TAZ, are considered target molecules of SSc-associated lung fibrosis and RA-ILD [145].

7.2. Computational Approach for Drug Repurposing

Three primary components must be considered in the drug repositioning process: diseases, drugs, and their targets. By understanding their interconnections, researchers can gain insight into the efficacy of plausible treatments and identify potential side effects. Numerous data, such as chemical structures, transcriptional profiles, and phenotypic information, are already available. Thus, holistic data analysis is essential to determine the capacity of drug repositioning, an area in which in silico models have already stood out.
The computational drug repositioning of anti-fibrotic agents is currently in progress. Wu et al. [146] developed Dr AFC, which is a drug repositioning platform for anti-fibrotic characteristics, using two different models: structural and biological profile prediction models. As their names imply, each model uses chemical structures and compound-induced gene profiles to predict the anti-fibrotic efficacy of the given compound. Dr AFC demonstrated high predictability for the test sets and proposed several candidates as novel anti-fibrotic agents.
Similar to Dr AFC, similarity-based virtual screening using 2D and 3D structures of possible target molecules of RA-ILD and drug candidates can be performed to predict their affinity. Drugs showing a higher affinity for the target molecule can be selected and evaluated on the cell lines of SKG mouse models to further confirm their effectiveness in RA-ILD. The drug repositioning process can be streamlined by sorting drugs with high structural and biological probabilities using computational methods before proceeding to the next step.
Since diseases share molecular pathways and targets, multiple screening analyses on the molecular targets of RA-ILD have already been conducted, even though the studies’ primary goal was not RA-ILD. For instance, several possible drugs targeting PDGFRa were screened among 7200 drugs and natural compounds through structure-based virtual screening and molecular docking analysis. Specifically, three-dimensional quantitative structure–activity relationship (3D-QSAR) and absorption, distribution, metabolism, excretion, and toxicity analyses were performed [147]. Gimeno et al. [148] virtually screened 20 promising broad-spectrum MMP inhibitors from the Specs library and 18 of them showed their MMP inhibition ability through a bioactivity assay. Multiple targets can also be simultaneously considered in computational screening. Wang et al. [149] screened multi-targeting inhibitors of VEFGRs, VEGFRs, FGFRs, PDGFRs, and TGFβ1R as a potential IPF prodrug using virtual screening and machine learning models. Twenty compounds were selected from the first screening and went through a further verification process. Subsequently, two promising drug candidates were found with anti-fibrotic activity on mouse IPF models with low cellular toxicity. These studies demonstrate that computational screening gives valuable and reliable outcomes for further drug development processes.
Such DTI prediction not only suggests a new drug but can also increase our knowledge about its potential side effects. For instance, Lounkine et al. [150] created a drug-target-adverse drug reaction network using a similarity ensemble approach, which predicts target binding based on chemical structure similarity with known ligands. New off-target and adverse events can be predicted using the network and can elucidate unknown mechanisms of adverse effects. One of the greatest obstacles to treating RA-ILD is the presence of multiple factors, such as joint, lung, and autoimmunity factors, that should be considered concurrently. If a drug benefits from one factor, but aggravates others, the drug is ineffective. Thus, we can predict off-target and adverse effects of the drugs in the early stages using these programs and filter out unsuitable options to accelerate the development process.
Computational analysis can help drug development to a further extent by identifying target molecules and signaling pathways that are involved in either the healing or adverse effects of drugs. A study by Zhu et al. [151] performed drug target analysis to understand the inexplicable healing effects of triptolide on CTD-ILDs. Using AutoDock Vina, they first discovered the top 10 target molecules of triptolide and further identified the signaling pathways. Another study aimed to understand the ILD that occurs in patients receiving TKI treatment. This treatment targeted EGFR, which is a key molecule of fibrosis. They analyzed the protein–protein interaction of EGFR and other proteins based on the String-bd database and discovered 37 molecules and 12 signaling pathways involved in EGFR-TKI-induced ILD [152].
Scholars are advancing in silico models with better algorithms and strategies to achieve higher prediction accuracy. In particular, the emergence and development of AI have changed the paradigm of drug discovery and drug repositioning. Traditional computational predictive approaches have relied on QSAR models utilizing machine learning techniques. These methods quickly calculated simple physicochemical properties from large amounts of data but were insufficient to predict complex properties, such as drug efficacy and adverse effects. Furthermore, these models face challenges with handling the variability of the dataset and their performance depends on the condition of the training datasets, such as their size or contamination with experimental errors [153]. However, with the development of AI, these issues are resolved as the computational screening techniques now encompass new models, such as deep learning (DL) and deep neural networks (DNN). For instance, DeepDTI, which was the first DL-based DTI prediction model developed by Wen et al. [154], outperformed other traditional machine learning techniques. In addition, the multitasking DNN-based model showed better predictability of drug toxicity compared with traditional QSAR models [155]. These advancements have also provided possibilities for precision medicine, which is suitable for RA-ILD because different biomarkers and pathogenic pathways are observed depending on the condition of the individual. Therefore, if these models can consider individual factors and help to prescribe customized drugs to each patient, this approach will be the optimal direction for future drug development.

8. Conclusions

RA-ILD is one of the most common lung manifestations of systemic inflammatory autoimmune disease, as roughly 10% of the patients with RA experience clinically significant RA-ILD [156]. Considering its high morbidity and mortality rates, the control of inflammation and fibrosis is of significant importance. This paper reviews the currently used agents for RA-ILD and describes a computational drug repositioning strategy that can benefit future drug developments for RA-ILD.
Nintedanib and pirfenidone are the most commonly used drugs for RA-ILD. The tyrosine kinase inhibitor, namely, nintedanib, controls the activity of fibrogenic cells (T cells, M2 macrophages, fibroblasts, and myofibroblasts) and decreases oxidative stress. Pirfenidone functions comprehensively via its anti-fibrotic, anti-inflammatory, and anti-oxidant effects. Multiple clinical trials were conducted using pirfenidone and nintedanib. They were shown to restore pulmonary function by slowing FVC decline and improving DLCO% and exercise capacity. Multiple studies demonstrated that immunosuppressants can stabilize or improve pulmonary function in patients with RA-ILD. However, the specific mechanism has not been clarified. Although more randomized controlled trials are required, regulating the autoimmune response can systematically affect multiple symptoms, including lung and arthritic manifestations. Similarly, several DMARDs exert auxiliary anti-fibrotic effects by controlling specific pathways involved in the progression of fibrogenesis. Drugs target signaling pathways, such as JAK/STAT, Hippo/Yap, Wnt/β-catenin, cGAS-STING, cAMP-PKA, and PI3K/AKT, to regulate cellular activity (differentiation, metabolic activity, or morphology), ECM homeostasis, and oxidative stress levels. While they differ only in the type of specific enzymes involved, these signaling pathways commonly regulate ECM homeostasis.
Although several drugs have been suggested and investigated, additional research in this area is required. This review summarizes three potent molecular targets that can impede fibrosis and a computational method that can determine the optimal drug to target.
Little is known about the pathogenesis of RA-ILD. Research conducted thus far has identified certain genetic backgrounds, environmental risk factors that trigger RA-ILD, and histologic evidence [8,157,158]. Thus, further research on the mechanism of RA-ILD, particularly focusing on the onset of the fibrotic stage and molecular correlations between joint inflammation and progressive fibrosis, is urgently needed. If the onset of the fibrotic stage can be clearly identified, preemptive actions, such as aggressive immunosuppressive treatment, can be administered in advance to inhibit subsequent fibrotic progression. Moreover, most drugs against RA are either predetermined to target joint inflammation or partially target fibrosis.
In contrast, anti-fibrotic agents inhibit joint inflammation. This implies that fibrosis and joint inflammation are interrelated in RA-ILD. In addition, deeper insights into the pathogenesis can resolve adverse events that frequently occur with current therapeutic treatments. Ultimately, the pathogenesis of RA-ILD must be elucidated to develop new drugs and reverse adverse effects.

Author Contributions

Conceptualization, K.-S.K.; writing—original draft preparation, E.J. and H.H.; writing—review and editing, E.J., H.H., Y.-A.L. and K.-S.K.; supervision, K.-S.K.; funding acquisition, K.-S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF), which is funded by the Ministry of Education, Science, and Technology (grant number 2021R1F1A1048536) and was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant HI17C0658).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABAAbatacept
AZAAzathioprine
bDMARDBiological disease-modifying antirheumatic drug
CSF1RColony-stimulating factor 1 receptor
CTD-ILDConnective tissue disease-interstitial lung disease
CTLA4Cytotoxic T-lymphocyte-associated antigen 4
CYCCyclophosphamide
DLDeep learning
DLCODiffusing capacity of lungs for carbon monoxide
DMARDDisease-modifying antirheumatic drug
DNNDeep neural network
DTIDrug–target interaction
ECMExtracellular matrix
EMTEpithelial–mesenchymal transition
EndoMTEndothelial–mesenchymal transition
FDAFood and Drug Administration
FGFFibroblast growth factor
FVCForced vital capacity
HRCTHigh-resolution computed tomography
ID1Inhibitor of DNA binding protein 1
IFN-γInterferon gamma
IGFBP2Insulin-like growth factor binding protein 2
ILDInterstitial lung disease
IPFIdiopathic pulmonary fibrosis
JAKJanus kinase
LckLymphocyte-specific tyrosine protein kinase
MMFMycophenolate mofetil
MTXMethotrexate
NSIPNonspecific interstitial pneumonia
OPOrganizing pneumonia
PDGFPlatelet-derived growth factor
PPARγPeroxisome proliferator-activated receptor gamma
QSARQuantitative structure–activity relationship
RARheumatoid arthritis
RA-ILDRheumatoid arthritis-associated interstitial lung disease
RTXRituximab
SScSystemic sclerosis
SSc-ILDSystemic sclerosis-associated interstitial lung disease
TACTacrolimus
TCZTocilizumab
TGF-βTumor growth factor beta
TKITyrosine kinase inhibitor
TLR4Toll-like receptor 4
TNF-αTumor necrosis factor alpha
UIPUsual interstitial pneumonia
VEGFRVascular growth factor receptor

References

  1. Liang, M.; Matteson, E.L.; Abril, A.; Distler, J.H.W. The role of antifibrotics in the treatment of rheumatoid arthritis–associated interstitial lung disease. Ther. Adv. Musculoskelet. Dis. 2022, 14, 1759720X2210744. [Google Scholar] [CrossRef]
  2. Norton, S.; Koduri, G.; Nikiphorou, E.; Dixey, J.; Williams, P.; Young, A. A study of baseline prevalence and cumulative incidence of comorbidity and extra-articular manifestations in RA and their impact on outcome. Rheumatology 2013, 52, 99–110. [Google Scholar] [CrossRef]
  3. Wilsher, M.; Voight, L.; Milne, D.; Teh, M.; Good, N.; Kolbe, J.; Williams, M.; Pui, K.; Merriman, T.; Sidhu, K.; et al. Prevalence of airway and parenchymal abnormalities in newly diagnosed rheumatoid arthritis. Respir. Med. 2012, 106, 1441–1446. [Google Scholar] [CrossRef]
  4. Huang, S.; Kronzer, V.L.; Dellaripa, P.F.; Deane, K.D.; Bolster, M.B.; Nagaraja, V.; Khanna, D.; Doyle, T.J.; Sparks, J.A. Rheumatoid arthritis-associated interstitial lung disease: Current update on prevalence, risk factors, and pharmacologic treatment. Curr. Treat. Options Rheumatol. 2020, 6, 337–353. [Google Scholar] [CrossRef] [PubMed]
  5. Yamakawa, H.; Sato, S.; Nishizawa, T.; Kawabe, R.; Oba, T.; Kato, A.; Horikoshi, M.; Akasaka, K.; Amano, M.; Sasaki, H.; et al. Impact of radiological honeycombing in rheumatoid arthritis-associated interstitial lung disease. BMC Pulm. Med. 2020, 20, 25. [Google Scholar] [CrossRef] [PubMed]
  6. 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 interstitial lung disease in rheumatoid arthritis: A population-based study. Arthritis Rheum. 2010, 62, 1583–1591. [Google Scholar] [CrossRef] [PubMed]
  7. Fragoulis, G.E.; Nikiphorou, E.; Larsen, J.; Korsten, P.; Conway, R. Methotrexate-Associated Pneumonitis and Rheumatoid Arthritis-Interstitial Lung Disease: Current Concepts for the Diagnosis and Treatment. Front. Med. 2019, 6, 238. [Google Scholar] [CrossRef] [PubMed]
  8. Fazeli, M.S.; Khaychuk, V.; Wittstock, K.; Han, X.; Crocket, G.; Lin, M.; Hill, S.L.; Ferri, L. Rheumatoid arthritis-associated interstitial lung disease: Epidemiology, risk/prognostic factors, and treatment landscape. Clin. Exp. Rheumatol. 2021, 39, 1108–1118. [Google Scholar] [CrossRef] [PubMed]
  9. Iqbal, K.; Kelly, C. Treatment of rheumatoid arthritis-associated interstitial lung disease: A perspective review. Ther. Adv. Musculoskelet. Dis. 2015, 7, 247–267. [Google Scholar] [CrossRef] [PubMed]
  10. Furukawa, H.; Oka, S.; Higuchi, T.; Shimada, K.; Hashimoto, A.; Matsui, T.; Tohma, S. Biomarkers for interstitial lung disease and acute-onset diffuse interstitial lung disease in rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X211022506. [Google Scholar] [CrossRef] [PubMed]
  11. Matson, S.; Lee, J.; Eickelberg, O. Two sides of the same coin? A review of the similarities and differences between idiopathic pulmonary fibrosis and rheumatoid arthritis-associated interstitial lung disease. Eur. Respir. J. 2021, 57, 2002533. [Google Scholar] [CrossRef]
  12. Kass, D.J.; Nouraie, M.; Glassberg, M.K.; Ramreddy, N.; Fernandez, K.; Harlow, L.; Zhang, Y.; Chen, J.; Kerr, G.S.; Reimold, A.M.; et al. Comparative Profiling of Serum Protein Biomarkers in Rheumatoid Arthritis-Associated Interstitial Lung Disease and Idiopathic Pulmonary Fibrosis. Arthritis Rheumatol. 2020, 72, 409–419. [Google Scholar] [CrossRef]
  13. Idiopathic Pulmonary Fibrosis Clinical Research Network; Raghu, G.; Anstrom, K.J.; King, T.E.; 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] [PubMed]
  14. Papiris, S.A.; Kagouridis, K.; Papadaki, G.; Kolilekas, L.; Manali, E.D. Treating CTDs related fibrotic ILDs by immunosuppressants: “facts and faults”. Lung 2014, 192, 221–223. [Google Scholar] [CrossRef]
  15. Cipriani, N.A.; Strek, M.; Noth, I.; Gordon, I.O.; Charbeneau, J.; Krishnan, J.A.; Krausz, T.; Husain, A.N. Pathologic quantification of connective tissue disease-associated versus idiopathic usual interstitial pneumonia. Arch. Pathol. Lab. Med. 2012, 136, 1253–1258. [Google Scholar] [CrossRef]
  16. Cassone, G.; Manfredi, A.; Vacchi, C.; Luppi, F.; Coppi, F.; Salvarani, C.; Sebastiani, M. Treatment of Rheumatoid Arthritis-Associated Interstitial Lung Disease: Lights and Shadows. J. Clin. Med. 2020, 9, 1082. [Google Scholar] [CrossRef]
  17. Kim, E.J.; Elicker, B.M.; Maldonado, F.; Webb, W.R.; Ryu, J.H.; Van Uden, J.H.; Lee, J.S.; King, T.E.; Collard, H.R. Usual interstitial pneumonia in rheumatoid arthritis-associated interstitial lung disease. Eur. Respir. J. 2010, 35, 1322–1328. [Google Scholar] [CrossRef]
  18. Antoniou, K.M.; Wuyts, W.; Wijsenbeek, M.; Wells, A.U. Medical Therapy in Idiopathic Pulmonary Fibrosis. Semin. Respir. Crit. Care Med. 2016, 37, 368–377. [Google Scholar] [CrossRef]
  19. Hilberg, F.; Roth, G.J.; Krssak, M.; Kautschitsch, S.; Sommergruber, W.; Tontsch-Grunt, U.; Garin-Chesa, P.; Bader, G.; Zoephel, A.; Quant, J.; et al. BIBF 1120: Triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res. 2008, 68, 4774–4782. [Google Scholar] [CrossRef]
  20. Wollin, L.; Distler, J.H.W.; Redente, E.F.; Riches, D.W.H.; Stowasser, S.; Schlenker-Herceg, R.; Maher, T.M.; Kolb, M. Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur. Respir. J. 2019, 54, 1900161. [Google Scholar] [CrossRef]
  21. Wollin, L.; Ostermann, A.; Williams, C. Nintedanib inhibits pro-fibrotic mediators from T cells with relevance to connective tissue disease-associated interstitial lung disease. Eur. Respir. J. 2017, 50, PA903. [Google Scholar] [CrossRef]
  22. Sato, S.; Shinohara, S.; Hayashi, S.; Morizumi, S.; Abe, S.; Okazaki, H.; Chen, Y.; Goto, H.; Aono, Y.; Ogawa, H.; et al. Anti-fibrotic efficacy of nintedanib in pulmonary fibrosis via the inhibition of fibrocyte activity. Respir. Res. 2017, 18, 172. [Google Scholar] [CrossRef]
  23. Braga, T.T.; Agudelo, J.S.H.; Camara, N.O.S. Macrophages During the Fibrotic Process: M2 as Friend and Foe. Front. Immunol. 2015, 6, 602. [Google Scholar] [CrossRef]
  24. Tandon, K.; Herrmann, F.; Ayaub, E.; Parthasarathy, P.; Ackermann, M.; Inman, M.D.; Kolb, M.R.J.; Wollin, L.; Ask, K. Nintedanib Attenuates the Polarization of Profibrotic Macrophages Through the Inhibition of Tyrosine Phosphorylation on CSF1 Receptor. In A72. MECHANISMS DRIVING FIBROSIS; American Thoracic Society International Conference Abstracts; American Thoracic Society: New York, NY, USA, 2017; p. A2397. [Google Scholar]
  25. 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]
  26. Matteson, E.L.; Aringer, M.; Burmester, G.R.; Mueller, H.; Moros, L.; Kolb, M. Effect of nintedanib in patients with progressive pulmonary fibrosis associated with rheumatoid arthritis: Data from the INBUILD trial. Clin. Rheumatol. 2023, 42, 2311–2319. [Google Scholar] [CrossRef]
  27. Silva, A.J.S.G.; Martins, A.; Ferreira, C.C.; Oliveira, C.P.; Guimarães, F.; Gago, L.; Duarte, A.C.; Costa, C.; Salvador, M.J.; Teixeira, V.; et al. Pos1307 Nintedanib in Rheumatic Disease-Associated Interstitial Lung Disease—A Multicentre Nationwide Cohort Study. Ann. Rheum. Dis. 2023, 82 (Suppl. S1), 1001–1002. [Google Scholar] [CrossRef]
  28. Redente, E.F.; Aguilar, M.A.; Black, B.P.; Edelman, B.L.; Bahadur, A.N.; Humphries, S.M.; Lynch, D.A.; Wollin, L.; Riches, D.W.H. Nintedanib reduces pulmonary fibrosis in a model of rheumatoid arthritis-associated interstitial lung disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 314, L998–L1009. [Google Scholar] [CrossRef]
  29. Conte, E.; Gili, E.; Fagone, E.; Fruciano, M.; Iemmolo, M.; Vancheri, C. Effect of pirfenidone on proliferation, TGF-β-induced myofibroblast differentiation and fibrogenic activity of primary human lung fibroblasts. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2014, 58, 13–19. [Google Scholar] [CrossRef]
  30. Nakazato, H.; Oku, H.; Yamane, S.; Tsuruta, Y.; Suzuki, R. A novel anti-fibrotic agent pirfenidone suppresses tumor necrosis factor-alpha at the translational level. Eur. J. Pharmacol. 2002, 446, 177–185. [Google Scholar] [CrossRef]
  31. Cheresh, P.; Kim, S.-J.; Tulasiram, S.; Kamp, D.W. Oxidative stress and pulmonary fibrosis. Biochim. Biophys. Acta 2013, 1832, 1028–1040. [Google Scholar] [CrossRef] [PubMed]
  32. Misra, H.P.; Rabideau, C. Pirfenidone inhibits NADPH-dependent microsomal lipid peroxidation and scavenges hydroxyl radicals. Mol. Cell. Biochem. 2000, 204, 119–126. [Google Scholar] [CrossRef] [PubMed]
  33. Gan, D.; Cheng, W.; Ke, L.; Sun, A.R.; Jia, Q.; Chen, J.; Lin, J.; Li, J.; Xu, Z.; Zhang, P. Repurposing of Pirfenidone (Anti-Pulmonary Fibrosis Drug) for Treatment of Rheumatoid Arthritis. Front. Pharmacol. 2021, 12, 631891. [Google Scholar] [CrossRef]
  34. Solomon, J.J.; Danoff, S.K.; Woodhead, F.A.; Hurwitz, S.; Maurer, R.; Glaspole, I.; Dellaripa, P.F.; Gooptu, B.; Vassallo, R.; Cox, P.G.; et al. Safety, tolerability, and efficacy of pirfenidone in patients with rheumatoid arthritis-associated interstitial lung disease: A randomised, double-blind, placebo-controlled, phase 2 study. Lancet Respir. Med. 2023, 11, 87–96. [Google Scholar] [CrossRef]
  35. Behr, J.; Prasse, A.; Kreuter, M.; Johow, J.; Rabe, K.F.; Bonella, F.; Bonnet, R.; Grohe, C.; Held, M.; Wilkens, H.; 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, 9, 476–486. [Google Scholar] [CrossRef]
  36. Wang, J.; Wang, X.; Qi, X.; Sun, Z.; Zhang, T.; Cui, Y.; Shu, Q. The Efficacy and Safety of Pirfenidone Combined With Immunosuppressant Therapy in Connective Tissue Disease-Associated Interstitial Lung Disease: A 24-Week Prospective Controlled Cohort Study. Front. Med. 2022, 9, 871861. [Google Scholar] [CrossRef]
  37. 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]
  38. Shu, Q. Efficacy, Safety, Immune Function of Pirfenidone in the Treatment of Connetive Tissue Disease-Related Interstitial Lung Disease(CTD-LID). Clinical Trial Registration, 2022. Available online: clinicaltrials.gov (accessed on 2 January 2024).
  39. Fragoulis, G.E.; Conway, R.; Nikiphorou, E. Methotrexate and interstitial lung disease: Controversies and questions. A narrative review of the literature. Rheumatology 2019, 58, 1900–1906. [Google Scholar] [CrossRef]
  40. Kiely, P.; Busby, A.D.; Nikiphorou, E.; Sullivan, K.; Walsh, D.A.; Creamer, P.; Dixey, J.; Young, A. 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, 9, e028466. [Google Scholar] [CrossRef]
  41. 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. 2021, 57, 2000337. [Google Scholar] [CrossRef]
  42. Kim, K.; Woo, A.; Park, Y.; Yong, S.H.; Lee, S.H.; Lee, S.H.; Leem, A.Y.; Kim, S.Y.; Chung, K.S.; Kim, E.Y.; et al. Protective effect of methotrexate on lung function and mortality in rheumatoid arthritis-related interstitial lung disease: A retrospective cohort study. Ther. Adv. Respir. Dis. 2022, 16, 17534666221135314. [Google Scholar] [CrossRef] [PubMed]
  43. Vastra Gotaland Region. Effects of Tofacitinib vs Methotrexate on Clinical and Molecular Disease Activity Markers in Joints and Lungs in Early Rheumatoid Arthritis (PULMORA)—A Randomized, Controlled, Open-label, Assessor-blinded, Phase IV Trial. Clinical Trial Registration, 2022. Available online: clinicaltrials.gov (accessed on 2 January 2024).
  44. Ahmed, A.R.; Hombal, S.M. Cyclophosphamide (Cytoxan). A review on relevant pharmacology and clinical uses. J. Am. Acad. Dermatol. 1984, 11, 1115–1126. [Google Scholar] [CrossRef]
  45. Ahlmann, M.; Hempel, G. The effect of cyclophosphamide on the immune system: Implications for clinical cancer therapy. Cancer Chemother. Pharmacol. 2016, 78, 661–671. [Google Scholar] [CrossRef]
  46. Schupp, J.C.; Köhler, T.; Müller-Quernheim, J. Usefulness of Cyclophosphamide Pulse Therapy in Interstitial Lung Diseases. Respir. Int. Rev. Thorac. Dis. 2016, 91, 296–301. [Google Scholar] [CrossRef]
  47. Barnes, H.; Holland, A.E.; Westall, G.P.; Goh, N.S.; Glaspole, I.N. Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst. Rev. 2018, 1, CD010908. [Google Scholar] [CrossRef]
  48. Maher, T.M.; Tudor, V.A.; Saunders, P.; Gibbons, M.A.; Fletcher, S.V.; Denton, C.P.; Hoyles, R.K.; Parfrey, H.; Renzoni, E.A.; Kokosi, M.; et al. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): A double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir. Med. 2023, 11, 45–54. [Google Scholar] [CrossRef] [PubMed]
  49. Suarez-Almazor, M.E.; Belseck, E.; Shea, B.; Wells, G.; Tugwell, P. Cyclophosphamide for treating rheumatoid arthritis. Cochrane Database Syst. Rev. 2000, 2000, CD001157. [Google Scholar] [CrossRef] [PubMed]
  50. Nakamura, K.; Ohbe, H.; Ikeda, K.; Uda, K.; Furuya, H.; Furuta, S.; Nakajima, M.; Sasabuchi, Y.; Matsui, H.; Fushimi, K.; et al. Intravenous cyclophosphamide in acute exacerbation of rheumatoid arthritis-related interstitial lung disease: A propensity-matched analysis using a nationwide inpatient database. Semin. Arthritis Rheum. 2021, 51, 977–982. [Google Scholar] [CrossRef] [PubMed]
  51. Ritter, M.L.; Pirofski, L. Mycophenolate mofetil: Effects on cellular immune subsets, infectious complications, and antimicrobial activity. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2009, 11, 290–297. [Google Scholar] [CrossRef] [PubMed]
  52. Morath, C.; Schwenger, V.; Beimler, J.; Mehrabi, A.; Schmidt, J.; Zeier, M.; Muranyi, W. Antifibrotic actions of mycophenolic acid. Clin. Transplant. 2006, 20 (Suppl. S17), 25–29. [Google Scholar] [CrossRef]
  53. Saketkoo, L.A.; Espinoza, L.R. Rheumatoid Arthritis Interstitial Lung Disease: Mycophenolate Mofetil as an Antifibrotic and Disease-Modifying Antirheumatic Drug. Arch. Intern. Med. 2008, 168, 1718–1719. [Google Scholar] [CrossRef] [PubMed]
  54. 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]
  55. Omair, M.A.; Alhamad, E.H. Mycophenolate mofetil is an effective therapy for connective tissue disease-associated interstitial lung disease. Int. J. Clin. Rheumatol. 2017, 12, 67. [Google Scholar]
  56. Cassone, G.; Sebastiani, M.; Vacchi, C.; Erre, G.L.; Salvarani, C.; Manfredi, A. Efficacy and safety of mycophenolate mofetil in the treatment of rheumatic disease-related interstitial lung disease: A narrative review. Drugs Context 2021, 10, 2020-8-8. [Google Scholar] [CrossRef]
  57. Schiff, M.; Beaulieu, A.; Scott, D.L.; Rashford, M. Mycophenolate mofetil in the treatment of adults with advanced rheumatoid arthritis: Three 24-week, randomized, double-blind, placebo- or ciclosporin-controlled trials. Clin. Drug Investig. 2010, 30, 613–624. [Google Scholar] [CrossRef] [PubMed]
  58. Maltzman, J.S.; Koretzky, G.A. Azathioprine: Old drug, new actions. J. Clin. Investig. 2003, 111, 1122–1124. [Google Scholar] [CrossRef]
  59. 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]
  60. 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]
  61. Terras Alexandre, A.; Martins, N.; Raimundo, S.; Melo, N.; Catetano Mota, P.; Novais E Bastos, H.; Pereira, J.M.; Cunha, R.; Guimarães, S.; Souto Moura, C.; et al. Impact of Azathioprine use in chronic hypersensitivity pneumonitis patients. Pulm. Pharmacol. Ther. 2020, 60, 101878. [Google Scholar] [CrossRef] [PubMed]
  62. Matson, S.M.; Baqir, M.; Moua, T.; Marll, M.; Kent, J.; Iannazzo, N.S.; Boente, R.D.; Donatelli, J.M.; Dai, J.; Diaz, F.J.; et al. Treatment Outcomes for Rheumatoid Arthritis-Associated Interstitial Lung Disease: A Real-World, Multisite Study of the Impact of Immunosuppression on Pulmonary Function Trajectory. Chest 2023, 163, 861–869. [Google Scholar] [CrossRef] [PubMed]
  63. Boerner, E.B.; Cuyas, M.; Theegarten, D.; Ohshimo, S.; Costabel, U.; Bonella, F. Azathioprine for Connective Tissue Disease-Associated Interstitial Lung Disease. Respir. Int. Rev. Thorac. Dis. 2020, 99, 628–636. [Google Scholar] [CrossRef]
  64. 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 disease-associated interstitial lung disease. Respir. Med. 2016, 121, 117–122. [Google Scholar] [CrossRef] [PubMed]
  65. Broen, J.C.A.; van Laar, J.M. Mycophenolate mofetil, azathioprine and tacrolimus: Mechanisms in rheumatology. Nat. Rev. Rheumatol. 2020, 16, 167–178. [Google Scholar] [CrossRef] [PubMed]
  66. Kurita, T.; Yasuda, S.; Oba, K.; Odani, T.; Kono, M.; Otomo, K.; Fujieda, Y.; Oku, K.; Bohgaki, T.; Amengual, O.; et al. The efficacy of tacrolimus in patients with interstitial lung diseases complicated with polymyositis or dermatomyositis. Rheumatology 2015, 54, 39–44. [Google Scholar] [CrossRef] [PubMed]
  67. Labirua-Iturburu, A.; Selva-O’Callaghan, A.; Martínez-Gómez, X.; Trallero-Araguás, E.; Labrador-Horrillo, M.; Vilardell-Tarrés, M. Calcineurin inhibitors in a cohort of patients with antisynthetase-associated interstitial lung disease. Clin. Exp. Rheumatol. 2013, 31, 436–439. [Google Scholar] [PubMed]
  68. Yamano, Y.; Taniguchi, H.; Kondoh, Y.; Ando, M.; Kataoka, K.; Furukawa, T.; Johkoh, T.; Fukuoka, J.; Sakamoto, K.; Hasegawa, Y. Multidimensional improvement in connective tissue disease-associated interstitial lung disease: Two courses of pulse dose methylprednisolone followed by low-dose prednisone and tacrolimus. Respirology 2018, 23, 1041–1048. [Google Scholar] [CrossRef]
  69. Kim, J.-W.; Chung, S.W.; Pyo, J.Y.; Chang, S.H.; Kim, M.U.; Park, C.H.; Lee, J.S.; Lee, J.S.; Ha, Y.-J.; Kang, E.H.; et al. Methotrexate, leflunomide and tacrolimus use and the progression of rheumatoid arthritis-associated interstitial lung disease. Rheumatology 2023, 62, 2377–2385. [Google Scholar] [CrossRef]
  70. Pescovitz, M.D. Rituximab, an anti-cd20 monoclonal antibody: History and mechanism of action. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2006, 6, 859–866. [Google Scholar] [CrossRef]
  71. Smith, M.R. Rituximab (monoclonal anti-CD20 antibody): Mechanisms of action and resistance. Oncogene 2003, 22, 7359–7368. [Google Scholar] [CrossRef]
  72. Ansari, M.A.; Nadeem, A.; Attia, S.M.; Bakheet, S.A.; Alasmari, A.F.; Alomar, H.A.; Al-Mazroua, H.A.; Alhamed, A.S.; Shahid, M.; Alqinyah, M.; et al. Rituximab exerts its anti-arthritic effects via inhibiting NF-κB/GM-CSF/iNOS signaling in B cells in a mouse model of collagen-induced arthritis. Heliyon 2023, 9, e16673. [Google Scholar] [CrossRef]
  73. Cohen, S.B.; Emery, P.; Greenwald, M.W.; Dougados, M.; Furie, R.A.; Genovese, M.C.; Keystone, E.C.; Loveless, J.E.; Burmester, G.-R.; Cravets, M.W.; et al. Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: Results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum. 2006, 54, 2793–2806. [Google Scholar] [CrossRef]
  74. Emery, P.; Fleischmann, R.; Filipowicz-Sosnowska, A.; Schechtman, J.; Szczepanski, L.; Kavanaugh, A.; Racewicz, A.J.; van Vollenhoven, R.F.; Li, N.F.; Agarwal, S.; et al. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: Results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial. Arthritis Rheum. 2006, 54, 1390–1400. [Google Scholar] [CrossRef]
  75. Fui, A.; Bergantini, L.; Selvi, E.; Mazzei, M.A.; Bennett, D.; Pieroni, M.G.; Rottoli, P.; Bargagli, E. Rituximab therapy in interstitial lung disease associated with rheumatoid arthritis. Intern. Med. J. 2020, 50, 330–336. [Google Scholar] [CrossRef]
  76. Vadillo, C.; Nieto, M.A.; Romero-Bueno, F.; Leon, L.; Sanchez-Pernaute, O.; Rodriguez-Nieto, M.J.; Freites, D.; Jover, J.A.; Álvarez-Sala, J.L.; Abasolo, L. Efficacy of rituximab in slowing down progression of rheumatoid arthritis-related interstitial lung disease: Data from the NEREA Registry. Rheumatology 2020, 59, 2099–2108. [Google Scholar] [CrossRef]
  77. 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 2017, 56, 1348–1357. [Google Scholar] [CrossRef]
  78. Vassallo, R.; Matteson, E.; Thomas, C.F. Clinical response of rheumatoid arthritis-associated pulmonary fibrosis to tumor necrosis factor-alpha inhibition. Chest 2002, 122, 1093–1096. [Google Scholar] [CrossRef]
  79. Koo, B.S.; Hong, S.; Kim, Y.J.; Kim, Y.-G.; Lee, C.-K.; Yoo, B. Mortality in patients with rheumatoid arthritis-associated interstitial lung disease treated with an anti-tumor necrosis factor agent. Korean J. Intern. Med. 2015, 30, 104–109. [Google Scholar] [CrossRef]
  80. Hagiwara, K.; Sato, T.; Takagi-Kobayashi, S.; Hasegawa, S.; Shigihara, N.; Akiyama, O. Acute exacerbation of preexisting interstitial lung disease after administration of etanercept for rheumatoid arthritis. J. Rheumatol. 2007, 34, 1151–1154. [Google Scholar]
  81. Korhonen, R.; Moilanen, E. Abatacept, a novel CD80/86-CD28 T cell co-stimulation modulator, in the treatment of rheumatoid arthritis. Basic Clin. Pharmacol. Toxicol. 2009, 104, 276–284. [Google Scholar] [CrossRef]
  82. Israël-Assayag, E.; Fournier, M.; Cormier, Y. Blockade of T cell costimulation by CTLA4-Ig inhibits lung inflammation in murine hypersensitivity pneumonitis. J. Immunol. Baltim. Md 1950 1999, 163, 6794–6799. [Google Scholar] [CrossRef]
  83. Boleto, G.; Guignabert, C.; Pezet, S.; Cauvet, A.; Sadoine, J.; Tu, L.; Nicco, C.; Gobeaux, C.; Batteux, F.; Allanore, Y.; et al. T-cell costimulation blockade is effective in experimental digestive and lung tissue fibrosis. Arthritis Res. Ther. 2018, 20, 197. [Google Scholar] [CrossRef]
  84. Fernández-Díaz, C.; Castañeda, S.; Melero-González, R.B.; Ortiz-Sanjuán, F.; Juan-Mas, A.; Carrasco-Cubero, C.; Casafont-Solé, I.; Olivé, A.; Rodríguez-Muguruza, S.; Almodóvar-González, R.; et al. Abatacept in interstitial lung disease associated with rheumatoid arthritis: National multicenter study of 263 patients. Rheumatology 2020, 59, 3906–3916. [Google Scholar] [CrossRef]
  85. Tardella, M.; Di Carlo, M.; Carotti, M.; Giovagnoni, A.; Salaffi, F. Abatacept in rheumatoid arthritis-associated interstitial lung disease: Short-term outcomes and predictors of progression. Clin. Rheumatol. 2021, 40, 4861–4867. [Google Scholar] [CrossRef]
  86. Vicente-Rabaneda, E.F.; Atienza-Mateo, B.; Blanco, R.; Cavagna, L.; Ancochea, J.; Castañeda, S.; González-Gay, M.Á. Efficacy and safety of abatacept in interstitial lung disease of rheumatoid arthritis: A systematic literature review. Autoimmun. Rev. 2021, 20, 102830. [Google Scholar] [CrossRef]
  87. Mena-Vázquez, N.; Rojas-Gimenez, M.; Fuego-Varela, C.; García-Studer, A.; Perez-Gómez, N.; Romero-Barco, C.M.; Godoy-Navarrete, F.J.; Manrique-Arija, S.; Gandía-Martínez, M.; Calvo-Gutiérrez, J.; et al. Safety and Effectiveness of Abatacept in a Prospective Cohort of Patients with Rheumatoid Arthritis-Associated Interstitial Lung Disease. Biomedicines 2022, 10, 1480. [Google Scholar] [CrossRef]
  88. Gudu, T.; Stober, C.; Fifield, M.; Babar, J.; Ostor, A.; Parfrey, H.; Hall, F. P147 Safety of AbatacePt in Rheumatoid arthritis associated Interstitial Lung disease (APRIL). Rheumatology 2021, 60, keab247.143. [Google Scholar] [CrossRef]
  89. 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]
  90. 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 Lond. Engl. 2016, 387, 2630–2640. [Google Scholar] [CrossRef] [PubMed]
  91. Manfredi, A.; Cassone, G.; Furini, F.; Gremese, E.; Venerito, V.; Atzeni, F.; Arrigoni, E.; Della Casa, G.; Cerri, S.; Govoni, M.; et al. Tocilizumab therapy in rheumatoid arthritis with interstitial lung disease: A multicentre retrospective study. Intern. Med. J. 2020, 50, 1085–1090. [Google Scholar] [CrossRef] [PubMed]
  92. Otsuji, N.; Sugiyama, K.; Arifuku, H.; Nakano, K.; Watanabe, H.; Wakayama, T.; Koyama, K.; Hirata, H.; Fukushima, Y. Effect of tocilizumab on interstitial lung disease (ILD) in patients with rheumatoid arthritis (RA). Eur. Respir. J. 2020, 56, 747. [Google Scholar] [CrossRef]
  93. Coutinho, A.E.; Chapman, K.E. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol. Cell. Endocrinol. 2011, 335, 2–13. [Google Scholar] [CrossRef]
  94. Lee, J.Y.; Jin, S.-M.; Lee, B.J.; Chung, D.H.; Jang, B.-G.; Park, H.S.; Lee, S.-M.; Yim, J.-J.; Yang, S.-C.; Yoo, C.-G.; et al. Treatment response and long term follow-up results of nonspecific interstitial pneumonia. J. Korean Med. Sci. 2012, 27, 661–667. [Google Scholar] [CrossRef]
  95. O’Dwyer, D.N.; Armstrong, M.E.; Cooke, G.; Dodd, J.D.; Veale, D.J.; Donnelly, S.C. Rheumatoid Arthritis (RA) associated interstitial lung disease (ILD). Eur. J. Intern. Med. 2013, 24, 597–603. [Google Scholar] [CrossRef]
  96. Song, J.W.; Lee, H.-K.; Lee, C.K.; Chae, E.J.; Jang, S.J.; Colby, T.V.; Kim, D.S. Clinical course and outcome of rheumatoid arthritis-related usual interstitial pneumonia. Sarcoidosis Vasc. Diffuse Lung Dis. Off. J. WASOG 2013, 30, 103–112. [Google Scholar]
  97. Bradley, B.; Branley, H.M.; Egan, J.J.; Greaves, M.S.; Hansell, D.M.; Harrison, N.K.; Hirani, N.; Hubbard, R.; Lake, F.; Millar, A.B.; et al. Interstitial lung disease guideline: The British Thoracic Society in collaboration with the Thoracic Society of Australia and New Zealand and the Irish Thoracic Society. Thorax 2008, 63 (Suppl. S5), v1–v58. [Google Scholar] [CrossRef]
  98. Idiopathic Pulmonary Fibrosis Clinical Research Network; Martinez, F.J.; de Andrade, J.A.; Anstrom, K.J.; King, T.E.; Raghu, G. Randomized trial of acetylcysteine in idiopathic pulmonary fibrosis. N. Engl. J. Med. 2014, 370, 2093–2101. [Google Scholar] [CrossRef]
  99. Zamora-Legoff, J.A.; Krause, M.L.; Crowson, C.S.; Ryu, J.H.; Matteson, E.L. Risk of serious infection in patients with rheumatoid arthritis-associated interstitial lung disease. Clin. Rheumatol. 2016, 35, 2585–2589. [Google Scholar] [CrossRef]
  100. Canalis, E.; Mazziotti, G.; Giustina, A.; Bilezikian, J.P. Glucocorticoid-induced osteoporosis: Pathophysiology and therapy. Osteoporos. Int. 2007, 18, 1319–1328. [Google Scholar] [CrossRef]
  101. Liu, J.; Wang, F.; Luo, F. The Role of JAK/STAT Pathway in Fibrotic Diseases: Molecular and Cellular Mechanisms. Biomolecules 2023, 13, 119. [Google Scholar] [CrossRef]
  102. Montero, P.; Milara, J.; Roger, I.; Cortijo, J. Role of JAK/STAT in Interstitial Lung Diseases; Molecular and Cellular Mechanisms. Int. J. Mol. Sci. 2021, 22, 6211. [Google Scholar] [CrossRef] [PubMed]
  103. Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, S.; Liu, M.; Li, X.; Zhang, J.; Wang, F.; Zhang, C.; Roden, A.; Ryu, J.H.; Warrington, K.J.; Sun, J.; et al. Canonical and noncanonical regulatory roles for JAK2 in the pathogenesis of rheumatoid arthritis-associated interstitial lung disease and idiopathic pulmonary fibrosis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2022, 36, e22336. [Google Scholar] [CrossRef]
  105. Zhang, Y.; Liang, R.; Chen, C.-W.; Mallano, T.; Dees, C.; Distler, A.; Reich, A.; Bergmann, C.; Ramming, A.; Gelse, K.; et al. JAK1-dependent transphosphorylation of JAK2 limits the antifibrotic effects of selective JAK2 inhibitors on long-term treatment. Ann. Rheum. Dis. 2017, 76, 1467–1475. [Google Scholar] [CrossRef]
  106. Strand, V.; van Vollenhoven, R.F.; Lee, E.B.; Fleischmann, R.; Zwillich, S.H.; Gruben, D.; Koncz, T.; Wilkinson, B.; Wallenstein, G. Tofacitinib or adalimumab versus placebo: Patient-reported outcomes from a phase 3 study of active rheumatoid arthritis. Rheumatology 2016, 55, 1031–1041. [Google Scholar] [CrossRef]
  107. Kremer, J.M.; Bloom, B.J.; Breedveld, F.C.; Coombs, J.H.; Fletcher, M.P.; Gruben, D.; Krishnaswami, S.; Burgos-Vargas, R.; Wilkinson, B.; Zerbini, C.A.F.; et al. The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase IIa trial of three dosage levels of CP-690,550 versus placebo. Arthritis Rheum. 2009, 60, 1895–1905. [Google Scholar] [CrossRef]
  108. Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2021 update. Pharmacol. Res. 2021, 165, 105463. [Google Scholar] [CrossRef]
  109. Tanaka, Y. A review of upadacitinib in rheumatoid arthritis. Mod. Rheumatol. 2020, 30, 779–787. [Google Scholar] [CrossRef]
  110. Zhang, J.; Wang, D.; Wang, L.; Wang, S.; Roden, A.C.; Zhao, H.; Li, X.; Prakash, Y.S.; Matteson, E.L.; Tschumperlin, D.J.; et al. Profibrotic effect of IL-17A and elevated IL-17RA in idiopathic pulmonary fibrosis and rheumatoid arthritis-associated lung disease support a direct role for IL-17A/IL-17RA in human fibrotic interstitial lung disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L487–L497. [Google Scholar] [CrossRef]
  111. Sendo, S.; Saegusa, J.; Yamada, H.; Nishimura, K.; Morinobu, A. Tofacitinib facilitates the expansion of myeloid-derived suppressor cells and ameliorates interstitial lung disease in SKG mice. Arthritis Res. Ther. 2019, 21, 184. [Google Scholar] [CrossRef] [PubMed]
  112. Citera, G.; Mysler, E.; Madariaga, H.; Cardiel, M.H.; Castañeda, O.; Fischer, A.; Richette, P.; Chartrand, S.; Park, J.K.; Strengholt, S.; et al. Incidence Rates of Interstitial Lung Disease Events in Tofacitinib-Treated Rheumatoid Arthritis Patients: Post Hoc Analysis From 21 Clinical Trials. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2021, 27, e482–e490. [Google Scholar] [CrossRef]
  113. Fleischmann, R.; Mease, P.J.; Schwartzman, S.; Hwang, L.-J.; Soma, K.; Connell, C.A.; Takiya, L.; Bananis, E. Efficacy of tofacitinib in patients with rheumatoid arthritis stratified by background methotrexate dose group. Clin. Rheumatol. 2017, 36, 15–24. [Google Scholar] [CrossRef]
  114. Vacchi, C.; Manfredi, A.; Cassone, G.; Cerri, S.; Della Casa, G.; Andrisani, D.; Salvarani, C.; Sebastiani, M. Tofacitinib for the Treatment of Severe Interstitial Lung Disease Related to Rheumatoid Arthritis. Case Rep. Med. 2021, 2021, 6652845. [Google Scholar] [CrossRef]
  115. Saldarriaga-Rivera, L.M.; López-Villegas, V.J. Janus kinase inhibitors as a therapeutic option in rheumatoid arthritis and associated interstitial lung disease. Report of four cases. Rev. Colomb. Reumatol. Engl. Ed. 2019, 26, 137–139. [Google Scholar] [CrossRef]
  116. Tardella, M.; Di Carlo, M.; Carotti, M.; Ceccarelli, L.; Giovagnoni, A.; Salaffi, F. A retrospective study of the efficacy of JAK inhibitors or abatacept on rheumatoid arthritis-interstitial lung disease. Inflammopharmacology 2022, 30, 705–712. [Google Scholar] [CrossRef] [PubMed]
  117. Lescoat, A.; Lelong, M.; Jeljeli, M.; Piquet-Pellorce, C.; Morzadec, C.; Ballerie, A.; Jouneau, S.; Jego, P.; Vernhet, L.; Batteux, F.; et al. Combined anti-fibrotic and anti-inflammatory properties of JAK-inhibitors on macrophages in vitro and in vivo: Perspectives for scleroderma-associated interstitial lung disease. Biochem. Pharmacol. 2020, 178, 114103. [Google Scholar] [CrossRef] [PubMed]
  118. Silva-Carmona, M.; Vogel, T.P.; Marchal, S.; Guesmi, M.; Dubus, J.-C.; Leroy, S.; Fabre, A.; Barlogis, V.; Forbes, L.R.; Giovannini-Chami, L. Successful Treatment of Interstitial Lung Disease in STAT3 Gain-of-Function Using JAK Inhibitors. Am. J. Respir. Crit. Care Med. 2020, 202, 893–897. [Google Scholar] [CrossRef] [PubMed]
  119. Bader-Meunier, B.; Hadchouel, A.; Berteloot, L.; Polivka, L.; Béziat, V.; Casanova, J.-L.; Lévy, R. Effectiveness and safety of ruxolitinib for the treatment of refractory systemic idiopathic juvenile arthritis like associated with interstitial lung disease: A case report. Ann. Rheum. Dis. 2022, 81, e20. [Google Scholar] [CrossRef]
  120. Smolen, J.S.; Landewé, R.; Bijlsma, J.; Burmester, G.; Chatzidionysiou, K.; Dougados, M.; Nam, J.; Ramiro, S.; Voshaar, M.; van Vollenhoven, R.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann. Rheum. Dis. 2017, 76, 960–977. [Google Scholar] [CrossRef]
  121. Salvarani, C.; Sebastiani, M.; Dieude, P.; Garcia, M.; Deberdt, W.; Rogai, V.; de la Torre, I.; Inciarte-Mundo, J.; Balsa, A. Baricitinib and the Risk of Incident Interstitial Lung Disease: A Descriptive Clinical Case Report from Clinical Trials. Rheumatol. Ther. 2021, 8, 1435–1441. [Google Scholar] [CrossRef]
  122. d’Alessandro, M.; Perillo, F.; Metella Refini, R.; Bergantini, L.; Bellisai, F.; Selvi, E.; Cameli, P.; Manganelli, S.; Conticini, E.; Cantarini, L.; et al. Efficacy of baricitinib in treating rheumatoid arthritis: Modulatory effects on fibrotic and inflammatory biomarkers in a real-life setting. Int. Immunopharmacol. 2020, 86, 106748. [Google Scholar] [CrossRef]
  123. Fraenkel, L.; Bathon, J.M.; England, B.R.; St Clair, E.W.; Arayssi, T.; Carandang, K.; Deane, K.D.; Genovese, M.; Huston, K.K.; Kerr, G.; et al. 2021 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Care Res. 2021, 73, 924–939. [Google Scholar] [CrossRef]
  124. Canadian Agency for Drugs and Technologies in Health. Pharmacoeconomic Review Report: Upadacitinib (Rinvoq): (AbbVie): Indication: For the Treatment of Adults with Moderately to Severely Active Rheumatoid Arthritis who have had an Inadequate Response or Intolerance to Methotrexate; CADTH Common Drug Reviews; Canadian Agency for Drugs and Technologies in Health: Ottawa, ON, Canada, 2020. [Google Scholar]
  125. Venerito, V.; Manfredi, A.; Carletto, A.; Gentileschi, S.; Atzeni, F.; Guiducci, S.; Lavista, M.; La Corte, L.; Pedrollo, E.; Scardapane, A.; et al. Evolution of Rheumatoid-Arthritis-Associated Interstitial Lung Disease in Patients Treated with JAK Inhibitors: A Retrospective Exploratory Study. J. Clin. Med. 2023, 12, 957. [Google Scholar] [CrossRef]
  126. Jourdan, J.-P.; Bureau, R.; Rochais, C.; Dallemagne, P. Drug repositioning: A brief overview. J. Pharm. Pharmacol. 2020, 72, 1145–1151. [Google Scholar] [CrossRef]
  127. Hirota, N.; Ito, T.; Miyazaki, S.; Ebina, M.; Homma, S. Gene expression profiling of lung myofibroblasts reveals the anti-fibrotic effects of cyclosporine. Tohoku J. Exp. Med. 2014, 233, 283–293. [Google Scholar] [CrossRef] [PubMed]
  128. Wójcik-Pszczoła, K.; Chłoń-Rzepa, G.; Jankowska, A.; Ślusarczyk, M.; Ferdek, P.E.; Kusiak, A.A.; Świerczek, A.; Pociecha, K.; Koczurkiewicz-Adamczyk, P.; Wyska, E.; et al. A Novel, Pan-PDE Inhibitor Exerts Anti-Fibrotic Effects in Human Lung Fibroblasts via Inhibition of TGF-β Signaling and Activation of cAMP/PKA Signaling. Int. J. Mol. Sci. 2020, 21, 4008. [Google Scholar] [CrossRef]
  129. Xie, X.; Wu, X.; Zhao, D.; Liu, Y.; Du, Q.; Li, Y.; Xu, Y.; Li, Y.; Qiu, Y.; Yang, Y. Fluvoxamine alleviates bleomycin-induced lung fibrosis via regulating the cGAS-STING pathway. Pharmacol. Res. 2023, 187, 106577. [Google Scholar] [CrossRef] [PubMed]
  130. Noble, P.W. Epithelial fibroblast triggering and interactions in pulmonary fibrosis. Eur. Respir. Rev. 2008, 17, 123–129. [Google Scholar] [CrossRef]
  131. Li, X.; Zhu, L.; Wang, B.; Yuan, M.; Zhu, R. Drugs and Targets in Fibrosis. Front. Pharmacol. 2017, 8, 855. [Google Scholar] [CrossRef] [PubMed]
  132. Chambers, R.C.; Mercer, P.F. Mechanisms of alveolar epithelial injury, repair, and fibrosis. Ann. Am. Thorac. Soc. 2015, 12 (Suppl. S1), S16–S20. [Google Scholar] [CrossRef]
  133. Bagnato, G.; Harari, S. Cellular interactions in the pathogenesis of interstitial lung diseases. Eur. Respir. Rev. Off. J. Eur. Respir. Soc. 2015, 24, 102–114. [Google Scholar] [CrossRef]
  134. Mahalanobish, S.; Saha, S.; Dutta, S.; Sil, P.C. Matrix metalloproteinase: An upcoming therapeutic approach for idiopathic pulmonary fibrosis. Pharmacol. Res. 2020, 152, 104591. [Google Scholar] [CrossRef]
  135. Makarev, E.; Izumchenko, E.; Aihara, F.; Wysocki, P.T.; Zhu, Q.; Buzdin, A.; Sidransky, D.; Zhavoronkov, A.; Atala, A. Common pathway signature in lung and liver fibrosis. Cell Cycle Georget. Tex 2016, 15, 1667–1673. [Google Scholar] [CrossRef] [PubMed]
  136. McEntee, C.P.; Gunaltay, S.; Travis, M.A. Regulation of barrier immunity and homeostasis by integrin-mediated transforming growth factor β activation. Immunology 2020, 160, 139–148. [Google Scholar] [CrossRef] [PubMed]
  137. Kelly, A.; Gunaltay, S.; McEntee, C.P.; Shuttleworth, E.E.; Smedley, C.; Houston, S.A.; Fenton, T.M.; Levison, S.; Mann, E.R.; Travis, M.A. Human monocytes and macrophages regulate immune tolerance via integrin αvβ8-mediated TGFβ activation. J. Exp. Med. 2018, 215, 2725–2736. [Google Scholar] [CrossRef] [PubMed]
  138. Herrera, I.; Cisneros, J.; Maldonado, M.; Ramírez, R.; Ortiz-Quintero, B.; Anso, E.; Chandel, N.S.; Selman, M.; Pardo, A. Matrix metalloproteinase (MMP)-1 induces lung alveolar epithelial cell migration and proliferation, protects from apoptosis, and represses mitochondrial oxygen consumption. J. Biol. Chem. 2013, 288, 25964–25975. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, B.; Crampton, S.P.; Hughes, C.C.W. Wnt signaling induces matrix metalloproteinase expression and regulates T cell transmigration. Immunity 2007, 26, 227–239. [Google Scholar] [CrossRef] [PubMed]
  140. Pilewski, J.M.; Liu, L.; Henry, A.C.; Knauer, A.V.; Feghali-Bostwick, C.A. Insulin-like growth factor binding proteins 3 and 5 are overexpressed in idiopathic pulmonary fibrosis and contribute to extracellular matrix deposition. Am. J. Pathol. 2005, 166, 399–407. [Google Scholar] [CrossRef] [PubMed]
  141. Nishimura, K.; Sugiyama, D.; Kogata, Y.; Tsuji, G.; Nakazawa, T.; Kawano, S.; Saigo, K.; Morinobu, A.; Koshiba, M.; Kuntz, K.M.; et al. Meta-analysis: Diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis. Ann. Intern. Med. 2007, 146, 797–808. [Google Scholar] [CrossRef] [PubMed]
  142. Kurowska, W.; Kuca-Warnawin, E.H.; Radzikowska, A.; Maśliński, W. The role of anti-citrullinated protein antibodies (ACPA) in the pathogenesis of rheumatoid arthritis. Cent.-Eur. J. Immunol. 2017, 42, 390–398. [Google Scholar] [CrossRef]
  143. Correia, C.S.; Briones, M.R.; Guo, R.; Ostrowski, R.A. Elevated anti-cyclic citrullinated peptide antibody titer is associated with increased risk for interstitial lung disease. Clin. Rheumatol. 2019, 38, 1201–1206. [Google Scholar] [CrossRef]
  144. Shao, T.; Shi, X.; Yang, S.; Zhang, W.; Li, X.; Shu, J.; Alqalyoobi, S.; Zeki, A.A.; Leung, P.S.; Shuai, Z. Interstitial Lung Disease in Connective Tissue Disease: A Common Lesion With Heterogeneous Mechanisms and Treatment Considerations. Front. Immunol. 2021, 12, 684699. [Google Scholar] [CrossRef]
  145. Mouawad, J.E.; Feghali-Bostwick, C. The Molecular Mechanisms of Systemic Sclerosis-Associated Lung Fibrosis. Int. J. Mol. Sci. 2023, 24, 2963. [Google Scholar] [CrossRef]
  146. Wu, D.; Gao, W.; Li, X.; Tian, C.; Jiao, N.; Fang, S.; Xiao, J.; Xu, Z.; Zhu, L.; Zhang, G.; et al. Dr AFC: Drug repositioning through anti-fibrosis characteristic. Brief. Bioinform. 2021, 22, bbaa115. [Google Scholar] [CrossRef] [PubMed]
  147. Mozzicafreddo, M.; Benfaremo, D.; Paolini, C.; Agarbati, S.; Svegliati Baroni, S.; Moroncini, G. Screening and Analysis of Possible Drugs Binding to PDGFRα: A Molecular Modeling Study. Int. J. Mol. Sci. 2023, 24, 9623. [Google Scholar] [CrossRef] [PubMed]
  148. Gimeno, A.; Cuffaro, D.; Nuti, E.; Ojeda-Montes, M.J.; Beltrán-Debón, R.; Mulero, M.; Rossello, A.; Pujadas, G.; Garcia-Vallvé, S. Identification of Broad-Spectrum MMP Inhibitors by Virtual Screening. Molecules 2021, 26, 4553. [Google Scholar] [CrossRef] [PubMed]
  149. Wang, R.; Xu, J.; Yan, R.; Liu, H.; Zhao, J.; Xie, Y.; Deng, W.; Liao, W.; Nie, Y. Virtual screening and activity evaluation of multitargeting inhibitors for idiopathic pulmonary fibrosis. Front. Pharmacol. 2022, 13, 998245. [Google Scholar] [CrossRef]
  150. Lounkine, E.; Keiser, M.J.; Whitebread, S.; Mikhailov, D.; Hamon, J.; Jenkins, J.L.; Lavan, P.; Weber, E.; Doak, A.K.; Côté, S.; et al. Large-scale prediction and testing of drug activity on side-effect targets. Nature 2012, 486, 361–367. [Google Scholar] [CrossRef]
  151. Zhu, W.; Li, Y.; Zhao, J.; Wang, Y.; Li, Y.; Wang, Y. The mechanism of triptolide in the treatment of connective tissue disease-related interstitial lung disease based on network pharmacology and molecular docking. Ann. Med. 2022, 54, 541–552. [Google Scholar] [CrossRef]
  152. Lu, Y.; Li, A.; Lai, X.; Jiang, J.; Zhang, L.; Zhong, Z.; Zhao, W.; Tang, P.; Zhao, H.; Ren, X. Identification of differentially expressed genes and signaling pathways using bioinformatics in interstitial lung disease due to tyrosine kinase inhibitors targeting the epidermal growth factor receptor. Investig. New Drugs 2019, 37, 384–400. [Google Scholar] [CrossRef]
  153. Zhu, H. Big Data and Artificial Intelligence Modeling for Drug Discovery. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 573–589. [Google Scholar] [CrossRef]
  154. Wen, M.; Zhang, Z.; Niu, S.; Sha, H.; Yang, R.; Yun, Y.; Lu, H. Deep-Learning-Based Drug-Target Interaction Prediction. J. Proteome Res. 2017, 16, 1401–1409. [Google Scholar] [CrossRef]
  155. Li, X.; Xu, Y.; Lai, L.; Pei, J. Prediction of Human Cytochrome P450 Inhibition Using a Multitask Deep Autoencoder Neural Network. Mol. Pharm. 2018, 15, 4336–4345. [Google Scholar] [CrossRef] [PubMed]
  156. Olson, A.L.; Swigris, J.J.; Sprunger, D.B.; Fischer, A.; Fernandez-Perez, E.R.; Solomon, J.; Murphy, J.; Cohen, M.; Raghu, G.; Brown, K.K. Rheumatoid Arthritis–Interstitial Lung Disease–associated Mortality. Am. J. Respir. Crit. Care Med. 2011, 183, 372–378. [Google Scholar] [CrossRef] [PubMed]
  157. Spagnolo, P.; Lee, J.S.; Sverzellati, N.; Rossi, G.; Cottin, V. The Lung in Rheumatoid Arthritis: Focus on Interstitial Lung Disease. Arthritis Rheumatol. 2018, 70, 1544–1554. [Google Scholar] [CrossRef]
  158. Rangel-Moreno, J.; Hartson, L.; Navarro, C.; Gaxiola, M.; Selman, M.; Randall, T.D. Inducible bronchus-associated lymphoid tissue (iBALT) in patients with pulmonary complications of rheumatoid arthritis. J. Clin. Investig. 2006, 116, 3183–3194. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic diagram in which immunosuppressants target rheumatoid arthritis-interstitial lung disease (RA-ILD). Mechanisms of several immunosuppressants against RA-ILD are presented. Therapeutic antibodies, such as rituximab and abatacept, immobilize immune cells, and anti-metabolites cause DNA damage. Multiple clinical trials have been conducted to evaluate their efficacy against RA-ILD. Created with BioRender.com (accessed on 22 Feburary 2024).
Figure 1. Schematic diagram in which immunosuppressants target rheumatoid arthritis-interstitial lung disease (RA-ILD). Mechanisms of several immunosuppressants against RA-ILD are presented. Therapeutic antibodies, such as rituximab and abatacept, immobilize immune cells, and anti-metabolites cause DNA damage. Multiple clinical trials have been conducted to evaluate their efficacy against RA-ILD. Created with BioRender.com (accessed on 22 Feburary 2024).
Ijms 25 02682 g001
Figure 2. Schematic diagram of the currently used drugs against RA-ILD. Immunosuppressants and anti-fibrotic agents control joint inflammation and pulmonary fibrosis to impede the fibrotic process of RA-ILD. Pulmonary fibrosis can be controlled via four mechanisms: diminishing fibrogenic cellular activity, restoring ECM homeostasis, decreasing oxidative stress, and controlling inflammation. Clinical trial results demonstrated that drugs for IPF treatment, nintedanib, and pirfenidone have anti-fibrotic effects against RA-ILD. Four JAK inhibitors, tofacitinib, ruxolitinib, baricitinib, and upadacitinib are targeted synthetic DMRARDs and not pure anti-fibrotics, but studies suggest their anti-fibrotic effects to a certain point. Created with BioRender.com (accessed on 22 Feburary 2024).
Figure 2. Schematic diagram of the currently used drugs against RA-ILD. Immunosuppressants and anti-fibrotic agents control joint inflammation and pulmonary fibrosis to impede the fibrotic process of RA-ILD. Pulmonary fibrosis can be controlled via four mechanisms: diminishing fibrogenic cellular activity, restoring ECM homeostasis, decreasing oxidative stress, and controlling inflammation. Clinical trial results demonstrated that drugs for IPF treatment, nintedanib, and pirfenidone have anti-fibrotic effects against RA-ILD. Four JAK inhibitors, tofacitinib, ruxolitinib, baricitinib, and upadacitinib are targeted synthetic DMRARDs and not pure anti-fibrotics, but studies suggest their anti-fibrotic effects to a certain point. Created with BioRender.com (accessed on 22 Feburary 2024).
Ijms 25 02682 g002
Figure 3. Schematic diagram of using features of the disease to identify potential targets and applying computational screening processes to screen potent drugs for further validation processes. Drug repositioning via computational screening can be an efficient process for further RA-ILD drug development. Pathologic features of RA-ILD can be acquired through its symptoms, transcriptional profile, pathogenic pathways, or other phenotypic information. Through this process, potential targets for drugs can be identified, and three potential targets were suggested: TGF-β, CTGF, and MMP. Computational platforms can embody the 2D and 3D structures of a specific target and calculate its interaction with multiple candidate drugs and screen potent drugs. Moreover, possible adverse effects of the drugs can also be identified, which will be helpful in further in vivo and in vitro or clinical trials. Created with BioRender.com (accessed on 22 Feburary 2024).
Figure 3. Schematic diagram of using features of the disease to identify potential targets and applying computational screening processes to screen potent drugs for further validation processes. Drug repositioning via computational screening can be an efficient process for further RA-ILD drug development. Pathologic features of RA-ILD can be acquired through its symptoms, transcriptional profile, pathogenic pathways, or other phenotypic information. Through this process, potential targets for drugs can be identified, and three potential targets were suggested: TGF-β, CTGF, and MMP. Computational platforms can embody the 2D and 3D structures of a specific target and calculate its interaction with multiple candidate drugs and screen potent drugs. Moreover, possible adverse effects of the drugs can also be identified, which will be helpful in further in vivo and in vitro or clinical trials. Created with BioRender.com (accessed on 22 Feburary 2024).
Ijms 25 02682 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jeong, E.; Hong, H.; Lee, Y.-A.; Kim, K.-S. Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development. Int. J. Mol. Sci. 2024, 25, 2682. https://doi.org/10.3390/ijms25052682

AMA Style

Jeong E, Hong H, Lee Y-A, Kim K-S. Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development. International Journal of Molecular Sciences. 2024; 25(5):2682. https://doi.org/10.3390/ijms25052682

Chicago/Turabian Style

Jeong, Eunji, Hyunseok Hong, Yeon-Ah Lee, and Kyoung-Soo Kim. 2024. "Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development" International Journal of Molecular Sciences 25, no. 5: 2682. https://doi.org/10.3390/ijms25052682

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