Skip to main content
Journal of Scleroderma and Related Disorders logoLink to Journal of Scleroderma and Related Disorders
. 2020 Mar 5;5(2 Suppl):6–16. doi: 10.1177/2397198320903867

Pathogenesis of systemic sclerosis associated interstitial lung disease

Svetlana I Nihtyanova 1, Christopher P Denton 1,
PMCID: PMC8922569  PMID: 35382227

Abstract

Systemic sclerosis is an autoimmune disease leading to vasculopathy and fibrosis of skin and internal organs. Despite likely shared pathogenic mechanisms, the patterns of skin and lung fibrosis differ. Pathogenesis of interstitial lung disease, a major cause of death in systemic sclerosis, reflects the intrinsic disease pathobiology and is associated with distinct clinical phenotypes and laboratory characteristics. The commonest histological pattern of systemic sclerosis–interstitial lung disease is non-specific interstitial pneumonia. Systemic sclerosis–interstitial lung disease pathogenesis involves multiple components, including susceptibility and triggering factors, which could be genetic or environmental. The process is amplified likely through ongoing inflammation and the link between inflammatory activity and fibrosis with IL6 emerging as a key mediator. The disease is driven by epithelial injury, reflected by markers in the serum, such as surfactant proteins and KL-6. In addition, mediators that are produced by epithelial cells and that regulate inflammatory cell trafficking may be important, especially CCL2. Other factors, such as CXCL4 and CCL18, point towards immune-mediated damage or injury response. Monocytes and alternatively activated macrophages appear to be important. Transforming growth factor beta appears central to pathogenesis and regulates epithelial repair and fibroblast activation. Understanding pathogenesis may help to unravel the stages of systemic sclerosis–interstitial lung disease, risks of progression and determinants of outcome. With this article, we set out to review the multiple factors, including genetic, environmental, cellular and molecular, that may be involved in the pathogenesis of systemic sclerosis–interstitial lung disease and the mechanisms leading to sustained fibrosis. We propose a model for the pathogenesis of systemic sclerosis–interstitial lung disease, based on the available literature.

Keywords: Fibroblast, autoantibodies, lung fibrosis, pathogenesis, cytokine

Introduction

Systemic sclerosis (scleroderma; SSc) is an uncommon disease with prevalence of around 1–2 in 10,000 and some clear associations with sex and age. 1 , 2 SSc is a prototypic multisystem fibrotic disease that leads to increased extracellular matrix deposition and structural changes in skin and internal organs. The extent of fibrosis varies between patients and across organs within a patient. However, there are common patterns of involvement that allow SSc to be classified into distinct subsets. Most characteristic is the differing extent of skin fibrosis that underpins the classification into limited or diffuse cutaneous subset. 3

Interstitial lung disease (ILD) is a term that covers a large group of disorders of the lung parenchyma, which involve development of inflammation and/or fibrosis of the lung. 4 Although many of those disorders are idiopathic, some can develop in the context of connective tissue diseases, including SSc. 5 In terms of histopathologic and radiographic features, SSc–ILD most often has features of non-specific interstitial pneumonia (NSIP) in up to 78% of subjects, followed by usual interstitial pneumonia (UIP) in up to 36%, while other patterns, such as organising pneumonia are much rarer.69 In addition, overall survival does not appear to be associated with the histopathologic/radiographic pattern of ILD with no difference in mortality between NSIP and UIP. 6

ILD is a common complication of SSc. Not all SSc patients undergo lung imaging and high-resolution computed tomography (HRCT) is generally performed only when indicated clinically by presence of dyspnoea, cough and late inspiratory crepitations on auscultation or decline in pulmonary function tests (PFT). As a result, estimation of the prevalence of mild ILD is difficult. On the other hand, prevalence of clinically significant lung fibrosis has been studied extensively, and in incident cohorts, approximately half of the patients are estimated to develop significant ILD, requiring immunosuppressive treatment.1012 Over the last three decades, ILD has become a leading disease-related cause of death in SSc patients.1316 A clear association between extent of ILD and mortality among SSc patients exists and a threshold of 20% extent of ILD on HRCT has been shown to distinguish between patents at high risk that require active treatment and those at low risk who would only require PFT monitoring. 17 ILD tends to develop early in the disease course 11 and short disease duration has been identified as one of the strongest predictors of ILD progression. 18

Although more common in diffuse cutaneous (dc)SSc, ILD can occur in either cutaneous subset. 10 , 11 It also has been observed in SSc patients carrying any of the SSc-specific hallmark autoantibodies, including anti-RNA polymerase and anti-centromere (ACA) antibodies, although anti-topoisomerase I (ATA; anti-Scl70) positive subjects are at a much higher risk of ILD development compared with any other antibody group. 11 , 19 , 20 Nevertheless, the risk for ILD development that is related to ATA positivity appears to be independent from cutaneous subset. 11 This suggests that as well as shared mechanisms, there are differences between skin and internal organs in terms of the development of fibrosis.

SSc is an autoimmune rheumatic disease, and the first abnormalities that are observed as the disease develops are typically the presence of autoantibodies and new-onset or worsening Raynaud’s phenomenon. This offers the opportunity for early diagnosis and has underpinned the concept of pre-scleroderma and the development of proposed criteria for the very early diagnosis of systemic sclerosis (VEDOSS). 21 That the immune system is central to the development of SSc is also supported by genetic studies that show replication of immune system genetic associations across multiple cohorts. In SSc, it is well-recognised that the first changes that occur in the skin histologically include microvascular endothelial cell activation and later immune cell infiltration. This includes initially cells of the innate immune system (CD68 positive monocyte/macrophages) and later the adaptive immune system with lymphocyte infiltration, including cells with B and T cell surface markers, followed by cells that show an activated immune phenotype, including T follicular helper cells. 22

Unlike skin, lung fibrosis poses a greater challenge for the researcher. Lung biopsies are very rarely performed in SSc patients, and if done, they are not repeated on multiple occasions and are not possible during the initial stages of disease. For that reason, a substantial part of the information available in the literature on the early stages of SSc–ILD comes from animal models.2326

A plausible model for SSc–ILD pathogenesis is one in which SSc represents a susceptibility phenotype where intrinsic immune mediated or inflammatory injury or extrinsic lung epithelial damage from environmental agents or infection leads to an excessive or exaggerated fibrotic response. The basis for this susceptibility to fibrosis and inability to resolve the fibrotic process once it is established is likely to include additional genetic and local cellular factors. This article explores the basis for these processes. We begin with review of genetic factors that have shown association with SSc–ILD, follow with a discussion of cellular and molecular players, as well as mechanisms leading to sustained fibrosis and finish with a proposed model for the pathogenesis of SSc–ILD, based on the available literature.

Genetics of SSc–ILD susceptibility

Genetic studies have recently highlighted a number of relevant susceptibility factors for SSc–ILD, although many are shared with other autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus.2730 A recent meta-analysis of genome-wide association studies (GWAS) identifies probably most of the relevant common genetic variants associated with disease susceptibility and starts to link variants with subtypes of disease. 31 In idiopathic pulmonary fibrosis (IPF), there are additional genetic mechanisms relating to mucin production, including MUC5B polymorphisms, 32 or possible response to epithelial damage followed by ineffective repair processes, due to changes in telomerase activity and chromosome instability. 33 Nevertheless, studies have failed to demonstrate association of those with SSc–ILD. 32 ,3436

A number of SSc–ILD candidate genes have been identified, although many studies include small numbers or have not been replicated. 37 The majority are genes for immune response molecules, such as interferon regulatory factor 5 (IRF5),3841 signal transducer and activator of transcription 4 (STAT4), 30 , 42 , 43 Interleukin-1 receptor-associated kinase-1 (IRAK1), 44 , 45 CD226 46 and CD247. 47 A number of human leukocyte antigen (HLA) region signals have also been identified, including HLA-DRB1, 3 and 5, DQB1 and DPB1,4855 although those associations are strongest with autoantibody specificities. In addition, genetic factors that are non-immune have been indicated, such as connective tissue growth factor (CTGF) polymorphisms.5658 These have not generally been replicated in all cohorts and may not represent a susceptibility gene in all populations, although the discrepancies may be related to differences in the phenotype definitions between patients from different centres. Modern genetic sequencing approaches, including direct sequencing, may eventually shed light on this by identifying rare functional variants that directly affect pathogenesis of SSc–ILD. 59

Cellular pathogenesis

The cellular pathogenesis of SSc–ILD reflects the complex nature of the disease that involves cells of the innate and adaptive immune system, vasculature and connective tissue as well as specialised lung structures, including alveolar epithelial cells (AECs). Inflammation and immune activation are early events and include infiltration of monocytes and macrophages within the lung parenchyma and accumulation of increased inflammatory cells in the alveolar space. 60 These phagocytic species reflect the need to remove toxic and infective material from the airspaces and highlight the lung as an environment where effective tissue homeostasis and resolution of injury are central to normal function. T and B cell infiltration follows and studies have demonstrated increased numbers of activated CD8+T lymphocytes, which produce profibrotic factors, including IL4, Oncostatin M and may activate latent transforming growth factor beta (TGF-β). 61 These cells can be sampled by bronchoalveolar lavage (BAL), which has provided valuable insight into the pathogenesis of ILD.6265 Granulocytosis has been found in two thirds of the patients with SSc–ILD. 63 Although neutrophilia and eosinophilia can also be observed and earlier studies suggest they predict future progression, 6 , 62 , 64 more recent publications show that the cellular profile of BAL fluid does not associate with SSc–ILD prognosis and is therefore not routinely performed in SSc patients.

AEC injury is another important element of the SSc–ILD pathogenesis and AECs may be damaged by environmental stimuli or from local inflammation. 60 , 66 Instead of repair from proliferation of type II cells, damage to type I cells is followed by migration of fibroblasts that lead to fibrotic tissue development. 67 There are also important populations of specialised epithelial cells that may produce surfactant proteins that are essential for normal physiological lung function and repair of lung injury. 68 The vascular compartment is critical for gas exchange and includes specialised cells within the blood vessel wall. Endothelial cells provide a critical barrier function to facilitate gas exchange as well as the large surface area essential for effective oxygen transfer. Smooth muscle cells, adventitial fibroblasts and specialised pericytes are also involved in response to tissue injury and may contribute to fibrosis. 69

Fibroblasts and myofibroblasts are the driving cells for scar formation and these can arise from multiple lineages including trans-differentiation from endothelial cells, epithelial cells or pericytes. 70 , 71 They may be recruited from a number or circulating precursors including fibrocytes and monocytes. 72 Expansion of local interstitial fibroblasts and resident lung progenitor mesenchymal cells are also important. 73 , 74

Key cytokines and molecular pathways that drive SSc–ILD

Cytokines and growth factors provide the intercellular mediators that co-ordinate and regulate tissue repair and the activation of the cellular players that are described above. It is likely that cytokines act in context and that multiple cell types are regulated by paracrine, autocrine and intracrine processes. 75 TGF-β is the major regulator of connective tissue growth and repair in embryonic development and postnatally. It also is well placed to coordinate post-natal response to tissue injury. It is preformed and sequestered in the extracellular matrix and activated when needed through a number of mechanisms. 76 Some of these such as integrin dependent activation may be especially relevant to lung injury and fibrosis. 77

Other cytokines are produced by lung inflammatory and epithelial cells. These include ubiquitous growth factors such as basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) and chemokines such as CCL2, CCL7 and CCL18.7883 There is considerable interest in these mediators as potential targets for treating fibrosis and this is important because they can be produced by and action multiple relevant cell types. These are not generally disease specific and represent mediators of tissue repair and responses to cellular injury. Chemokines are emerging as major candidate mediators in SSc–ILD based upon the ability of damaged lung tissue, especially epithelial cells, to produce chemokines such as CCL2 that may then modulate inflammation and leucocyte migration. 78 , 79 Other chemokines may derive from platelets, such as CXCL4 (ILD4) 84 , 85 or other infiltrating cells, including immune cells (CCL18). Levels of CCL18 and CCL2 have been associated with outcome in observational cohorts and clinical trials. 78 , 79 , 81 , 82

IL6 has been shown to be important in ILD locally and systemically. Recent studies point towards a particularly important role in early stages of SSc–ILD and it may link inflammation and fibrosis. 86 Intracellular signalling pathway for IL6 and TGF-β converge, including STAT3, and also cellular interaction may link fibroblast derived IL6 and other cell types such as macrophages. 87 There is a growing appreciation of the potential important role of macrophages in fibrosis and that the diverse functional properties and ontogeny of macrophages in the lung may be important in pathogenesis and therapy of ILD. 88 The recent licencing of nintedanib shows that blocking intracellular signalling for multiple ubiquitous growth factors can be very effective. 89 Similarly, the data from the tocilizumab trials suggest that Interleukin 6 receptor (IL6R) blockade can also be effective in those SSc patients most at risk of early progression of ILD.9092

Lung injury is associated with activation of the coagulation pathway and this can result in release of products of the coagulation pathway that are profibrotic. This includes thrombin and later stage factors, such as factor XIII. The latter is a transglutaminase that may link the late stages of coagulation after injury and may promote TGF-β activation via secondary effects on thrombospondin, a major activator of TGF-β in animal models. 93

Lessons from preclinical models

The complex nature of SSc and its hallmark clinical heterogeneity have been a challenge for the development and validation of preclinical models. Not all animal models of SSc develop pulmonary fibrosis. The most widely used models for SSc–ILD are the bleomycin lung injury model and a number of genetic models, including Fra-2, Fli1, uPAR and TbRIIΔk-fib. The bleomycin model is well established in ILD research. 94 Although different protocols exist, which include varying doses and routes of administration (transoral, endotracheal, subcutaneous, intravenous and intraperitoneal), the fibrotic process follows approximately the same course. Acute inflammation develops within the first 7 days, followed by fibrosis 1–2 weeks after bleomycin administration. 94 In wild type mice, the fibrosis generally resolves. Use of genetically modified mice that develop mild spontaneous ILD can show more extreme changes in response to lung injury and have helped to define critical mediators or candidate pathways for attenuation. 95 , 96 A transgenic mouse model with altered TGF-β signalling in fibroblasts (TbRIIΔk-fib) has been shown to develop ILD in approximately 25% of the animals from 6 weeks 97 , 98 and in response to minor epithelial lung injury and may represent a model for milder SSc-associated ILD. 95 , 96 This mouse strain also develops other relevant abnormalities in the skin and vasculature. 23 The Fra-2 transgenic mouse offers another model for SSc and from 12 weeks the animals develop skin and lung fibrosis. 99 , 100

Mechanisms that sustain fibrosis

In SSc, it seems likely that changes in the skin reflect activity of the proinflammatory process and this is reflected by the progression that occurs in the first 12–24 months. This is followed by stabilisation or regression when the normal biology of wound healing prevails and leads to softening of the skin and improvement in the mRSS. It seems likely that to some extent, the progressive phase occurs throughout affected organs, although the timing and extent may differ. In particular, there may be inherent differences between organs in the extent to which regression and de-remodelling of scarred tissue can occur with later regeneration of specialised structures and organs.

In the lung, it is likely that once fibrosis is established, with disorganised lung architecture and structural changes, this may be essentially irreversible. This would fit with observed changes in skin and lung over time that have recently been described in SSc patients. 11 High-risk cases develop severe or extensive ILD relatively early and the hazard of clinically significant ILD development is highest in the first 24–36 months of the disease, rapidly declining thereafter. This is also the time period when mRSS would peak for the majority of SSc patients. While skin then improves for over 80% of the patients, 101 ILD may stabilise or progress, and the rate of progression is determined by the extent of fibrosis, which was used as the basis of the Goh staging system. 17 The trajectories for forced vital capacity (FVC) change over time for a large cohort of SSc cases under long-term regular review are shown in Figure 1. The high-risk cases with ATA experience a significant drop in FVC much earlier than patients with other SSc-specific antibodies. ATA is also associated with the slowest rate of skin improvement. 11 The rates of change in FVC in the later stages of disease appear similar between groups and likely reflects shared pathogenic mechanisms for worsening. These include recurrent lung injury that could be essentially the level of normal environmental challenge that healthy individuals experience, but that in SSc there is greater tendency to fibrosis and less ability to recover without fixed scar. Other factors could include microaspiration with damage from bile salts and acid, despite maximal anti-reflux treatments. 102 , 103 Mechano-sensing due to stiffened lung tissue is also important and this has been shown to drive myofibroblast differentiation via a number of pathways including myocardin-related transcription factor-A (MRTF-A) pathway and yes-associated protein (YAP). 104 All of these mechanisms may represent molecular targets for treatment in the future.

Figure 1.

Figure 1.

Development and progression of SSc–ILD suggests contrasting drivers in disease subsets: (a) Time to clinically significant pulmonary fibrosis and (b) average FVC change over time in subgroups by antibodies and subset. Autoantibody and skin subset appear to determine the development and progression of SSc–ILD. Thus, some antibodies are associated with early development of extensive disease, 17 with ATA having the highest risk. (a) Time to development of clinically significant ILD in 403 SSc patients, positive for the three most common scleroderma-specific autoantibodies, anti-centromere (ACA), anti-topoisomerase I (ATA) and anti-RNA polymerase antibody (ARA). While in their great majority, ACA+ patients develop limited cutaneous and ARA+ diffuse cutaneous subset of SSc (97% and 92% in this cohort), ATA+ subjects can present with either subset (35% limited and 65% diffuse in this cohort), which does affect their overall survival, but not their risk of significant ILD. (b) Modelling FVC changes over time in 297 SSc–ILD patients, who were positive for either ACA, ATA or ARA. Disease progression, based on modelled FVC trajectory, show relevant ANA associated differences. Thus, ARA and ATA overall have similar later-stage rate of progression, but at different levels of impairment, reflecting higher risk of early severe ILD in ATA. This later stage progression likely depends on factors outlined in Figure 1 and may also reflect systemic fibrotic activity, as the slope of decline for ATA+ cases is greater in those with diffuse compared with limited skin involvement.

Phases of pathogenesis for SSc–ILD

The development and progression of ILD in SSc can be envisioned into the main stages that are outlined and summarised in Figure 2.

Figure 2.

Figure 2.

Phases of development and progression of lung fibrosis in systemic sclerosis (SSc–ILD). Schematic illustrating the three independent phases in pathogenesis of SSc–ILD that reflect different susceptibility within the SSc subgroups based upon skin extent and ANA reactivity. In a susceptible patient, triggering events that may reflect lung injury or intrinsic disease-related immune mechanisms lead to interstitial inflammation and fibrosis. This may then extend and become clinically meaningful through similar mechanisms. Outcome of progression, stability or regression will be affected by systemic disease features, including SSc subset, ANA and intrinsic lung fibrotic mechanisms such as tissue stiffness and structural changes to lung architecture.

Early phase – susceptibility and triggering

Like in most complex diseases, aetiopathogenesis of SSc–ILD is likely to be complex and multifactorial and to involve triggering events occurring by chance in a susceptible individual. This susceptibility is likely to be at least genetic or epigenetic, but environmental cofactors may also be relevant, such as the lung microbiome. 105 , 106 Environmental influences are likely to include those that may initiate or trigger the disease and others that modulate progression. The latter may be especially relevant to ILD where there might be susceptibility to tissue damage and a predilection to fibrotic scarring in response to recurrent or persistent lung injury due to environmental chemical exposure, recurrent microaspiration 102 , 107 or episodes of infection. 108 , 109

It seems likely that the early stages of lung involvement in SSc, especially in those patients with the highest risk of ILD, will reflect the same processes observed in other SSc-related organ complications, including endothelial activation and T and B cell infiltration. Later neutrophils predominate in BAL fluid and may reflect the extent of lung damage. 63 High-resolution CT patterns support the importance of early inflammation with amorphous ground glass change although this may represent fine fibrosis rather than pure inflammation even in early stage disease. 9

Cohort studies have highlighted that early SSc is often associated with loss of lung function, especially in the diffuse cutaneous subset, and this is supported by data from several recent clinical trials recruiting very early stage cases. Thus, in RISE-SSc even with average disease duration of less than 9 month, there was evidence of clinically meaningful decline in FVC% predicted over 52 weeks. 110 Likewise, two placebo-controlled trials of tocilizumab that enrolled early stage (less than 2 years average disease duration) dcSSc subjects with evidence of increased acute phase markers, showed very marked overall decline in FVC in the placebo arm, although interestingly this was largely prevented by therapeutic IL-6 receptor blockade in the tocilizumab treatment arms.9092 This highlights the progressivity of mild early ILD in dcSSc and the potentially important role of IL6 as a driver of progression. These interventional trial data are supported by observational data from two cohorts that also identify serum IL6 as a significant predictor of lung function decline and death in early stage or milder ILD. 86 , 111

The importance of early inflammation is supported by preclinical models such as the bleomycin model of ILD that has been used in many laboratory studies. Links between anti-nuclear antibody (ANA) pattern and risk of development of severe ILD also point to the importance of immune mechanisms, but also imply that these may differ between the ANA-defined subsets in SSc. For some high-risk antibodies, this seems to be independent of the extent of skin fibrosis, as it is not impacted by SSc subset once the onset of disease is classified by first non-Raynaud’s manifestation. 11

The consequence of early inflammation is the development of an activated fibroblast population that has a marked pro-fibrotic phenotype. This includes a gene and protein expression profile of TGF-β activation. It is likely that distinct subpopulations of fibroblasts in the lung contribute to the development of fibrosis and these are likely to have distinct origins. 67 They include cells derived from local fibroblast activation and proliferation as well as cells derived from other lineages, notably epithelial cells and endothelial cells as well as progenitor populations including pericytes. Preclinical experiments have defined an essential role for resident lung fibroblasts in regulating the fibrotic process and confirmed that this is dependent upon intact TGF-β signalling. 96

Established phase – progression and failed resolution

Clinically, there is evolution from the early inflammatory lesion towards a more fibrotic phenotype. This equates with the development of a typical NSIP pattern of ILD that may be cellular or fibrotic. 6 The drivers of this process are likely to include ongoing inflammation and the interplay between the innate and adaptive immune system and fibroblasts that leads to increased matrix deposition. It seems likely that this phase of pathogenesis is influenced by the activity of the disease process and determines whether patients remain with mild and more stable ILD or evolve into a more extensive disease. Factors that determine this may include intensity of the inflammatory process, 63 , 64 recruitment of profibrotic cell populations, including circulating fibrocytes, 64 , 72 and failure of apoptosis of myofibroblasts that has been demonstrated to be a key mechanism in experimental mouse models of SSc–ILD. 95 Many of these changes may reflect a background profibrotic susceptibility phenotype in SSc that links to the pattern of diseases, including ANA-defined subgroups. 11 , 112 Factors such as altered microbiome or recurrent aspiration and lung injury may become relevant at this stage and determine whether lung disease progresses.105108 Local activation of fibrotic pathways and recruitment of feed-forward loops involving TGF-β dependent pathways such as mechano-sensors including MRTF-A and YAP–TAZ may be involved. 104 , 113

Late phase – severe fibrosis in a subset of patients

Clinically, the most important stage of ILD in SSc is the established progressive phase. It has been noted that not all cases progress and in fact some remain remarkably stable. However, those that develop more extensive disease have a poor outcome due to ILD and other complications, such as pulmonary hypertension. 114 In this phase, the risk of progression has been demonstrated to reflect the extent of disease and damage. 17 This may be the result of mechanical stiffness and altered lung structure, implying that a pattern of progression more akin to IPF may be relevant, albeit less rapidly progressive overall. 115 This is the phase of disease recruited into most SSc–ILD trials and is probably the most appropriate stage to tackle underlying fibrotic pathways that are shared across different forms of ILD. This would be in keeping with data from emerging clinical trials, such as INBUILD, that show treatment effect is much greater in more severe and progressive cases of ILD than in a mixed cohort of SSc–ILD, although there is almost complete agreement between the proportional impact of anti-fibrotic treatment with nintedanib. 89 , 116 This more extensive and progressive disease is more likely to show major architectural disruption and more UIP-like pattern on CT or lung biopsy. Recurrent infection and aspiration are likely to be major drivers and some cases may develop pleuroparenchymal fibroelastosis that is especially associated with recurrent infection. 117

Concluding remarks

SSc–ILD remains an important challenge, but is now starting to reveal key mechanisms and potential therapeutic avenues. The recent success of targeting tyrosine kinase activity linked to specific receptors for several major growth factors – platelet-derived growth factor (PDGF), VEGF, fibroblast growth factor (FGF) and colony stimulating factor 1 (CSF1), as well as Src family kinases for treatment of ILD. 118 Initial trials were in IPF, but more recently, nintedanib was shown to have a major effect on progressive lung fibrosis of other causes. In SSc, treatment with nintedanib resulted in a comparable reduction in rate of decline of lung function in SSc–ILD patients. 89 The latter is a much less progressive condition overall and so demonstration of treatment effect is a major confirmation of the antifibrotic efficacy of nintedanib. This also suggests that the relevant fibrotic processes, shared with more progressive forms of ILD, are less central in SSc. Conversely, recent results from two studies of anti-IL6R show more marked treatment effect on lung function in a subgroup of early stage active dcSSc at particular risk of ILD.9092 This likely targets the earlier stage pathogenic pathways and mechanisms that underlie the prevalent early progressive phase of ILD in SSc whereas a later less progressive but more IPF like fibrotic mechanisms is targeted by nintedanib. 118 , 119 Thus, reverse translation is likely to shed important light on stages and mechanisms of pathogenesis for SSc–ILD in the future.

Footnotes

Declaration of conflicting interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: C.P.D. has received research grants from GlaxoSmithKline, CSL Behring and Inventiva, and consulting fees or honorarium from Roche, Actelion, GlaxoSmithKline, Sanofi Aventis, Inventiva, CSL Behring, Boehringer Ingelheim, Corbus, Galapagos and Bayer.

Funding: The author(s) received no financial support for the research, authorship and/or publication of this article.

ORCID iD: Christopher P Denton Inline graphic https://orcid.org/0000-0003-3975-8938

References

  • 1. Dougherty DH, Kwakkenbos L, Carrier ME, et al. The scleroderma patient-centered intervention network cohort: baseline clinical features and comparison with other large scleroderma cohorts. Rheumatology 2018; 57(9): 1623–1631. [DOI] [PubMed] [Google Scholar]
  • 2. Barnes J, Mayes MD. Epidemiology of systemic sclerosis: incidence, prevalence, survival, risk factors, malignancy, and environmental triggers. Curr Opin Rheumatol 2012; 24(2): 165–170. [DOI] [PubMed] [Google Scholar]
  • 3. LeRoy EC, Black C, Fleischmajer R, et al. Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol 1988; 15(2): 202–205. [PubMed] [Google Scholar]
  • 4. McLean-Tooke A, Moore I, Lake F. Idiopathic and immune-related pulmonary fibrosis: diagnostic and therapeutic challenges. Clin Transl Immunology 2019; 8(11): e1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wells AU, Denton CP. Interstitial lung disease in connective tissue disease–mechanisms and management. Nat Rev Rheumatol 2014; 10(12): 728–739. [DOI] [PubMed] [Google Scholar]
  • 6. Bouros D, Wells AU, Nicholson AG, et al. Histopathologic subsets of fibrosing alveolitis in patients with systemic sclerosis and their relationship to outcome. Am J Respir Crit Care Med 2002; 165(12): 1581–1586. [DOI] [PubMed] [Google Scholar]
  • 7. Fischer A, Swigris JJ, Groshong SD, et al. Clinically significant interstitial lung disease in limited scleroderma: histopathology, clinical features, and survival. Chest 2008; 134(3): 601–605. [DOI] [PubMed] [Google Scholar]
  • 8. Goldin JG, Lynch DA, Strollo DC, et al. High-resolution CT scan findings in patients with symptomatic scleroderma-related interstitial lung disease. Chest 2008; 134(2): 358–367. [DOI] [PubMed] [Google Scholar]
  • 9. Desai SR, Veeraraghavan S, Hansell DM, et al. CT features of lung disease in patients with systemic sclerosis: comparison with idiopathic pulmonary fibrosis and nonspecific interstitial pneumonia. Radiology 2004; 232(2): 560–567. [DOI] [PubMed] [Google Scholar]
  • 10. Nihtyanova SI, Schreiber BE, Ong VH, et al. Prediction of pulmonary complications and long-term survival in systemic sclerosis. Arthritis Rheumatol 2014; 66(6): 1625–1635. [DOI] [PubMed] [Google Scholar]
  • 11. Nihtyanova SI, Sari A, Harvey JC, et al. Using autoantibodies and cutaneous subset to develop outcome-based disease classification in systemic sclerosis. Arthritis Rheumatol. Epub ahead of print 4 November 2019. DOI: 10.1002/art.41153. [DOI] [PubMed] [Google Scholar]
  • 12. Launay D, Remy-Jardin M, Michon-Pasturel U, et al. High resolution computed tomography in fibrosing alveolitis associated with systemic sclerosis. J Rheumatol 2006; 33(9): 1789–1801. [PubMed] [Google Scholar]
  • 13. Rubio-Rivas M, Royo C, Simeon CP, et al. Mortality and survival in systemic sclerosis: systematic review and meta-analysis. Semin Arthritis Rheum 2014; 44(2): 208–219. [DOI] [PubMed] [Google Scholar]
  • 14. Steen VD, Medsger TA. Changes in causes of death in systemic sclerosis, 1972-2002. Ann Rheum Dis 2007; 66(7): 940–944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Elhai M, Meune C, Avouac J, et al. Trends in mortality in patients with systemic sclerosis over 40 years: a systematic review and meta-analysis of cohort studies. Rheumatology 2012; 51(6): 1017–1026. [DOI] [PubMed] [Google Scholar]
  • 16. Komocsi A, Vorobcsuk A, Faludi R, et al. The impact of cardiopulmonary manifestations on the mortality of SSc: a systematic review and meta-analysis of observational studies. Rheumatology 2012; 51(6): 1027–1036. [DOI] [PubMed] [Google Scholar]
  • 17. Goh NS, Desai SR, Veeraraghavan S, et al. Interstitial lung disease in systemic sclerosis: a simple staging system. Am J Respir Crit Care Med 2008; 177(11): 1248–1254. [DOI] [PubMed] [Google Scholar]
  • 18. Winstone TA, Assayag D, Wilcox PG, et al. Predictors of mortality and progression in scleroderma-associated interstitial lung disease: a systematic review. Chest 2014; 146(2): 422–436. [DOI] [PubMed] [Google Scholar]
  • 19. Walker UA, Tyndall A, Czirjak L, et al. Clinical risk assessment of organ manifestations in systemic sclerosis: a report from the EULAR Scleroderma trials and research group database. Ann Rheum Dis 2007; 66(6): 754–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hesselstrand R, Scheja A, Shen GQ, et al. The association of antinuclear antibodies with organ involvement and survival in systemic sclerosis. Rheumatology 2003; 42(4): 534–540. [DOI] [PubMed] [Google Scholar]
  • 21. Minier T, Guiducci S, Bellando-Randone S, et al. Preliminary analysis of the very early diagnosis of systemic sclerosis (VEDOSS) EUSTAR multicentre study: evidence for puffy fingers as a pivotal sign for suspicion of systemic sclerosis. Ann Rheum Dis 2014; 73(12): 2087–2093. [DOI] [PubMed] [Google Scholar]
  • 22. Taylor DK, Mittereder N, Kuta E, et al. T follicular helper-like cells contribute to skin fibrosis. Sci Transl Med 2018; 10(431): eaaf5307. [DOI] [PubMed] [Google Scholar]
  • 23. Derrett-Smith EC, Denton CP, Sonnylal S. Animal models of scleroderma: lessons from transgenic and knockout mice. Curr Opin Rheumatol 2009; 21(6): 630–635. [DOI] [PubMed] [Google Scholar]
  • 24. Beyer C, Schett G, Distler O, et al. Animal models of systemic sclerosis: prospects and limitations. Arthritis Rheum 2010; 62(10): 2831–2844. [DOI] [PubMed] [Google Scholar]
  • 25. Asano Y, Sato S. Animal models of scleroderma: current state and recent development. Curr Rheumatol Rep 2013; 15(12): 382. [DOI] [PubMed] [Google Scholar]
  • 26. Marangoni RG, Varga J, Tourtellotte WG. Animal models of scleroderma: recent progress. Curr Opin Rheumatol 2016; 28(6): 561–570. [DOI] [PubMed] [Google Scholar]
  • 27. Carmona FD, Martin JE, Beretta L, et al. The systemic lupus erythematosus IRF5 risk haplotype is associated with systemic sclerosis. PLoS ONE 2013; 8(1): e54419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Tang L, Chen B, Ma B, et al. Association between IRF5 polymorphisms and autoimmune diseases: a meta-analysis. Genet Mol Res 2014; 13(2): 4473–4485. [DOI] [PubMed] [Google Scholar]
  • 29. Remmers EF, Plenge RM, Lee AT, et al. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med 2007; 357(10): 977–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Dieude P, Guedj M, Wipff J, et al. STAT4 is a genetic risk factor for systemic sclerosis having additive effects with IRF5 on disease susceptibility and related pulmonary fibrosis. Arthritis Rheum 2009; 60(8): 2472–2479. [DOI] [PubMed] [Google Scholar]
  • 31. Lopez-Isac E, Acosta-Herrera M, Kerick M, et al. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways. Nat Commun 2019; 10(1): 4955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Stock CJ, Sato H, Fonseca C, et al. Mucin 5B promoter polymorphism is associated with idiopathic pulmonary fibrosis but not with development of lung fibrosis in systemic sclerosis or sarcoidosis. Thorax 2013; 68(5): 436–441. [DOI] [PubMed] [Google Scholar]
  • 33. Bilgili H, Bialas AJ, Gorski P, et al. Telomere abnormalities in the pathobiology of idiopathic pulmonary fibrosis. J Clin Med 2019; 8(8): E1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Borie R, Crestani B, Dieude P, et al. The MUC5B variant is associated with idiopathic pulmonary fibrosis but not with systemic sclerosis interstitial lung disease in the European Caucasian population. PLoS ONE 2013; 8(8): e70621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Wu M, Assassi S, Salazar GA, et al. Genetic susceptibility loci of idiopathic interstitial pneumonia do not represent risk for systemic sclerosis: a case control study in Caucasian patients. Arthritis Res Ther 2016; 18: 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Lakota K, Hanumanthu VS, Agrawal R, et al. Short lymphocyte, but not granulocyte, telomere length in a subset of patients with systemic sclerosis. Ann Rheum Dis 2019; 78(8): 1142–1144. [DOI] [PubMed] [Google Scholar]
  • 37. Stock CJW, Renzoni EA. Genetic predictors of systemic sclerosis-associated interstitial lung disease: a review of recent literature. Eur J Hum Genet 2018; 26(6): 765–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Dieude P, Dawidowicz K, Guedj M, et al. Phenotype-haplotype correlation of IRF5 in systemic sclerosis: role of 2 haplotypes in disease severity. J Rheumatol 2010; 37(5): 987–992. [DOI] [PubMed] [Google Scholar]
  • 39. Dieude P, Guedj M, Wipff J, et al. Association between the IRF5 rs2004640 functional polymorphism and systemic sclerosis: a new perspective for pulmonary fibrosis. Arthritis Rheum 2009; 60(1): 225–233. [DOI] [PubMed] [Google Scholar]
  • 40. Wang J, Yi L, Guo X. Association of the IRF5 SNP rs2004640 with systemic sclerosis in Han Chinese. Int J Immunopathol Pharmacol 2014; 27(4): 635–638. [DOI] [PubMed] [Google Scholar]
  • 41. Sharif R, Mayes MD, Tan FK, et al. IRF5 polymorphism predicts prognosis in patients with systemic sclerosis. Ann Rheum Dis 2012; 71(7): 1197–1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Rueda B, Broen J, Simeon C, et al. The STAT4 gene influences the genetic predisposition to systemic sclerosis phenotype. Hum Mol Genet 2009; 18(11): 2071–2077. [DOI] [PubMed] [Google Scholar]
  • 43. Yi L, Wang JC, Guo XJ, et al. STAT4 is a genetic risk factor for systemic sclerosis in a Chinese population. Int J Immunopathol Pharmacol 2013; 26(2): 473–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Carmona FD, Cenit MC, Diaz-Gallo LM. New insight on the Xq28 association with systemic sclerosis. Ann Rheum Dis 2013; 72(12): 2032–2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Dieude P, Bouaziz M, Guedj M. Evidence of the contribution of the X chromosome to systemic sclerosis susceptibility: association with the functional IRAK1 196Phe/532Ser haplotype. Arthritis Rheum 2011; 63(12): 3979–3987. [DOI] [PubMed] [Google Scholar]
  • 46. Bossini-Castillo L, Simeon CP, Beretta L, et al. A multicenter study confirms CD226 gene association with systemic sclerosis-related pulmonary fibrosis. Arthritis Res Ther 2012; 14(2): R85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Dieude P, Boileau C, Guedj M. Independent replication establishes the CD247 gene as a genetic systemic sclerosis susceptibility factor. Ann Rheum Dis 2011; 70(9): 1695–1696. [DOI] [PubMed] [Google Scholar]
  • 48. Zhou X, Lee JE, Arnett FC. HLA-DPB1 and DPB2 are genetic loci for systemic sclerosis: a genome-wide association study in Koreans with replication in North Americans. Arthritis Rheum 2009; 60(12): 3807–3814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Briggs DC, Vaughan RW, Welsh KI, et al. Immunogenetic prediction of pulmonary fibrosis in systemic sclerosis. Lancet 1991; 338: 661–662. [DOI] [PubMed] [Google Scholar]
  • 50. Fanning GC, Welsh KI, Bunn C, et al. HLA associations in three mutually exclusive autoantibody subgroups in UK systemic sclerosis patients. Br J Rheumatol 1998; 37(2): 201–207. [DOI] [PubMed] [Google Scholar]
  • 51. He D, Wang J, Yi L. Association of the HLA-DRB1 with scleroderma in Chinese population. PLoS ONE 2014; 9(9): e0106939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Odani T, Yasuda S, Ota Y, et al. Up-regulated expression of HLA-DRB5 transcripts and high frequency of the HLA-DRB5*01: 05 allele in scleroderma patients with interstitial lung disease. Rheumatology 2012; 51(10): 1765–1774. [DOI] [PubMed] [Google Scholar]
  • 53. Tikly M, Rands A, McHugh N, et al. Human leukocyte antigen class II associations with systemic sclerosis in South Africans. Tissue Antigens 2004; 63(5): 487–490. [DOI] [PubMed] [Google Scholar]
  • 54. Wang J, Guo X, Yi L. Association of HLA-DPB1 with scleroderma and its clinical features in Chinese population. PLoS ONE 2014; 9(1): e87363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Gorlova O, Martin JE, Rueda B. Identification of novel genetic markers associated with clinical phenotypes of systemic sclerosis through a genome-wide association strategy. PLoS Genet 2011; 7(7): e1002178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Zhao W, Yue X, Liu K, et al. The status of pulmonary fibrosis in systemic sclerosis is associated with IRF5, STAT4, IRAK1, and CTGF polymorphisms. Rheumatol Int 2017; 37(8): 1303–1310. [DOI] [PubMed] [Google Scholar]
  • 57. Fonseca C, Lindahl GE, Ponticos M, et al. A polymorphism in the CTGF promoter region associated with systemic sclerosis. N Engl J Med 2007; 357(12): 1210–1220. [DOI] [PubMed] [Google Scholar]
  • 58. Zhang X, Nie S, Si X, et al. Association between the CTGF -945C/G polymorphism and systemic sclerosis: a meta-analysis. Gene 2012; 509(1): 1–6. [DOI] [PubMed] [Google Scholar]
  • 59. Gourh P, Remmers EF, Boyden SE, et al. Brief report: whole-exome sequencing to identify rare variants and gene networks that increase susceptibility to scleroderma in African Americans. Arthritis Rheumatol 2018; 70(10): 1654–1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Schniering J, Guo L, Brunner M, et al. Evaluation of (99m)Tc-rhAnnexin V-128 SPECT/CT as a diagnostic tool for early stages of interstitial lung disease associated with systemic sclerosis. Arthritis Res Ther 2018; 20(1): 183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Luzina IG, Atamas SP, Wise R, et al. Occurrence of an activated, profibrotic pattern of gene expression in lung CD8+ T cells from scleroderma patients. Arthritis Rheum 2003; 48(8): 2262–2274. [DOI] [PubMed] [Google Scholar]
  • 62. Wells AU, Hansell DM, Rubens MB, et al. Fibrosing alveolitis in systemic sclerosis. Bronchoalveolar lavage findings in relation to computed tomographic appearance. Am J Respir Crit Care Med 1994; 150(2): 462–468. [DOI] [PubMed] [Google Scholar]
  • 63. Goh NS, Veeraraghavan S, Desai SR, et al. Bronchoalveolar lavage cellular profiles in patients with systemic sclerosis-associated interstitial lung disease are not predictive of disease progression. Arthritis Rheum 2007; 56(6): 2005–2012. [DOI] [PubMed] [Google Scholar]
  • 64. Kowal-Bielecka O, Kowal K, Highland KB, et al. Bronchoalveolar lavage fluid in scleroderma interstitial lung disease: technical aspects and clinical correlations: review of the literature. Semin Arthritis Rheum 2010; 40(1): 73–88. [DOI] [PubMed] [Google Scholar]
  • 65. De Santis M, Bosello SL, Peluso G, et al. Bronchoalveolar lavage fluid and progression of scleroderma interstitial lung disease. Clin Respir J 2012; 6(1): 9–17. [DOI] [PubMed] [Google Scholar]
  • 66. Asano Y, Takahashi T, Saigusa R. Systemic sclerosis: is the epithelium a missing piece of the pathogenic puzzle? J Dermatol Sci 2019; 94(2): 259–265. [DOI] [PubMed] [Google Scholar]
  • 67. Bagnato G, Harari S. Cellular interactions in the pathogenesis of interstitial lung diseases. Eur Respir Rev 2015; 24(135): 102–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Yamakawa H, Hagiwara E, Kitamura H, et al. Serum KL-6 and surfactant protein-D as monitoring and predictive markers of interstitial lung disease in patients with systemic sclerosis and mixed connective tissue disease. J Thorac Dis 2017; 9(2): 362–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Gilbane AJ, Denton CP, Holmes AM. Scleroderma pathogenesis: a pivotal role for fibroblasts as effector cells. Arthritis Res Ther 2013; 15(3): 215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Rajkumar VS, Howell K, Csiszar K, et al. Shared expression of phenotypic markers in systemic sclerosis indicates a convergence of pericytes and fibroblasts to a myofibroblast lineage in fibrosis. Arthritis Res Ther 2005; 7(5): R1113–R1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Mendoza FA, Piera-Velazquez S, Farber JL, et al. Endothelial cells expressing endothelial and mesenchymal cell gene products in lung tissue from patients with systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol 2016; 68(1): 210–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Maharaj S, Shimbori C, Kolb M. Fibrocytes in pulmonary fibrosis: a brief synopsis. Eur Respir Rev 2013; 22(130): 552–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Abraham DJ, Eckes B, Rajkumar V, et al. New developments in fibroblast and myofibroblast biology: implications for fibrosis and scleroderma. Curr Rheumatol Rep 2007; 9(2): 136–143. [DOI] [PubMed] [Google Scholar]
  • 74. Valenzi E, Bulik M, Tabib T, et al. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann Rheum Dis 2019; 78(10): 1379–1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Jee AS, Sahhar J, Youssef P, et al. Review: serum biomarkers in idiopathic pulmonary fibrosis and systemic sclerosis associated interstitial lung disease–frontiers and horizons. Pharmacol Ther 2019; 202: 40–52. [DOI] [PubMed] [Google Scholar]
  • 76. Lafyatis R. Transforming growth factor beta–at the centre of systemic sclerosis. Nat Rev Rheumatol 2014; 10(12): 706–719. [DOI] [PubMed] [Google Scholar]
  • 77. Henderson NC, Sheppard D. Integrin-mediated regulation of TGFbeta in fibrosis. Biochim Biophys Acta 2013; 1832(7): 891–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Carulli MT, Handler C, Coghlan JG, et al. Can CCL2 serum levels be used in risk stratification or to monitor treatment response in systemic sclerosis? Ann Rheum Dis 2008; 67(1): 105–109. [DOI] [PubMed] [Google Scholar]
  • 79. Wu M, Baron M, Pedroza C, et al. CCL2 in the circulation predicts long-term progression of interstitial lung disease in patients with early systemic sclerosis: data from two independent cohorts. Arthritis Rheumatol 2017; 69(9): 1871–1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Yanaba K, Komura K, Kodera M, et al. Serum levels of monocyte chemotactic protein-3/CCL7 are raised in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis. Ann Rheum Dis 2006; 65(1): 124–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Hoffmann-Vold AM, Tennoe AH, Garen T, et al. High level of chemokine CCL18 is associated with pulmonary function deterioration, lung fibrosis progression, and reduced survival in systemic sclerosis. Chest 2016; 150(2): 299–306. [DOI] [PubMed] [Google Scholar]
  • 82. Schupp J, Becker M, Gunther J, et al. Serum CCL18 is predictive for lung disease progression and mortality in systemic sclerosis. Eur Respir J 2014; 43(5): 1530–1532. [DOI] [PubMed] [Google Scholar]
  • 83. Ong VH, Evans LA, Shiwen X, et al. Monocyte chemoattractant protein 3 as a mediator of fibrosis: overexpression in systemic sclerosis and the type 1 tight-skin mouse. Arthritis Rheum 2003; 48(7): 1979–1991. [DOI] [PubMed] [Google Scholar]
  • 84. van Bon L, Affandi AJ, Broen J, et al. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J Med 2014; 370(5): 433–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Volkmann ER, Tashkin DP, Roth MD, et al. Changes in plasma CXCL4 levels are associated with improvements in lung function in patients receiving immunosuppressive therapy for systemic sclerosis-related interstitial lung disease. Arthritis Res Ther 2016; 18(1): 305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Khan K, Xu S, Nihtyanova S, et al. Clinical and pathological significance of interleukin 6 overexpression in systemic sclerosis. Ann Rheum Dis 2012; 71(7): 1235–1242. [DOI] [PubMed] [Google Scholar]
  • 87. Denton CP, Ong VH, Xu S, et al. Therapeutic interleukin-6 blockade reverses transforming growth factor-beta pathway activation in dermal fibroblasts: insights from the faSScinate clinical trial in systemic sclerosis. Ann Rheum Dis 2018; 77(9): 1362–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp Med 2011; 208(7): 1339–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J Med 2019; 380(26): 2518–2528. [DOI] [PubMed] [Google Scholar]
  • 90. Khanna D, Denton CP, Jahreis A, et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 2016; 387(10038): 2630–2640. [DOI] [PubMed] [Google Scholar]
  • 91. Khanna D, Denton CP, Lin CJF, et al. Safety and efficacy of subcutaneous tocilizumab in systemic sclerosis: results from the open-label period of a phase II randomised controlled trial (faSScinate). Ann Rheum Dis 2018; 77(2): 212–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Khanna D, Lin C, Kuwana M, et al. Efficacy and safety of tocilizumab for the treatment of systemic sclerosis: results from a phase 3 randomized controlled trial [abstract]. Arthritis Rheumatol 2018; 70(Suppl. 10): 898. [Google Scholar]
  • 93. Crawford SE, Stellmach V, Murphy-Ullrich JE, et al. Thrombospondin-1 is a major activator of TGF-beta1 in vivo. Cell 1998; 93(7): 1159–1170. [DOI] [PubMed] [Google Scholar]
  • 94. Liu T, De Los Santos FG, Phan SH. The Bleomycin model of pulmonary fibrosis. Method Mol Biol 2017; 1627: 27–42. [DOI] [PubMed] [Google Scholar]
  • 95. Hoyles RK, Khan K, Shiwen X, et al. Fibroblast-specific perturbation of transforming growth factor beta signaling provides insight into potential pathogenic mechanisms of scleroderma-associated lung fibrosis: exaggerated response to alveolar epithelial injury in a novel mouse model. Arthritis Rheum 2008; 58(4): 1175–1188. [DOI] [PubMed] [Google Scholar]
  • 96. Hoyles RK, Derrett-Smith EC, Khan K, et al. An essential role for resident fibroblasts in experimental lung fibrosis is defined by lineage-specific deletion of high-affinity type II transforming growth factor beta receptor. Am J Respir Crit Care Med 2011; 183(2): 249–261. [DOI] [PubMed] [Google Scholar]
  • 97. Denton CP, Lindahl GE, Khan K, et al. Activation of key profibrotic mechanisms in transgenic fibroblasts expressing kinase-deficient type II transforming growth factor-{beta} receptor (T{beta}RII{delta}k). J Biol Chem 2005; 280(16): 16053–16065. [DOI] [PubMed] [Google Scholar]
  • 98. Denton CP, Zheng B, Evans LA, et al. Fibroblast-specific expression of a kinase-deficient type II transforming growth factor beta (TGFbeta) receptor leads to paradoxical activation of TGFbeta signaling pathways with fibrosis in transgenic mice. J Biol Chem 2003; 278(27): 25109–25119. [DOI] [PubMed] [Google Scholar]
  • 99. Eferl R, Hasselblatt P, Rath M, et al. Development of pulmonary fibrosis through a pathway involving the transcription factor Fra-2/AP-1. Proc Natl Acad Sci USA 2008; 105(30): 10525–10530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Maurer B, Distler JH, Distler O. The Fra-2 transgenic mouse model of systemic sclerosis. Vascul Pharmacol 2013; 58(3): 194–201. [DOI] [PubMed] [Google Scholar]
  • 101. Shand L, Lunt M, Nihtyanova S, et al. Relationship between change in skin score and disease outcome in diffuse cutaneous systemic sclerosis: application of a latent linear trajectory model. Arthritis Rheum 2007; 56(7): 2422–2431. [DOI] [PubMed] [Google Scholar]
  • 102. Chwiesko A, Kowal-Bielecka O, Sierakowski S. Perspectives on the interlinked nature of systemic sclerosis and reflux disease. Expert Rev Gastroenterol Hepatol 2019; 13(3): 213–227. [DOI] [PubMed] [Google Scholar]
  • 103. Christmann RB, Wells AU, Capelozzi VL, et al. Gastroesophageal reflux incites interstitial lung disease in systemic sclerosis: clinical, radiologic, histopathologic, and treatment evidence. Semin Arthritis Rheum 2010; 40(3): 241–249. [DOI] [PubMed] [Google Scholar]
  • 104. Toyama T, Looney AP, Baker BM, et al. Therapeutic targeting of TAZ and YAP by dimethyl fumarate in systemic sclerosis fibrosis. J Invest Dermatol 2018; 138(1): 78–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Denton CP, Murray C. Cause or effect? Interpreting emerging evidence for dysbiosis in systemic sclerosis. Arthritis Res Ther 2019; 21(1): 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Mehta H, Goulet PO, Mashiko S, et al. Early-life antibiotic exposure causes intestinal dysbiosis and exacerbates skin and lung pathology in experimental systemic sclerosis. J Invest Dermatol 2017; 137(11): 2316–2325. [DOI] [PubMed] [Google Scholar]
  • 107. Richardson C, Agrawal R, Lee J, et al. Esophageal dilatation and interstitial lung disease in systemic sclerosis: a cross-sectional study. Semin Arthritis Rheum 2016; 46(1): 109–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Clements PJ, Goldin JG, Kleerup EC, et al. Regional differences in bronchoalveolar lavage and thoracic high-resolution computed tomography results in dyspneic patients with systemic sclerosis. Arthritis Rheum 2004; 50(6): 1909–1917. [DOI] [PubMed] [Google Scholar]
  • 109. De Santis M, Bosello S, La Torre G, et al. Functional, radiological and biological markers of alveolitis and infections of the lower respiratory tract in patients with systemic sclerosis. Respir Res 2005; 6: 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Distler O, Allanore Y, Denton C, et al. Riociguat in patients with early diffuse cutaneous systemic sclerosis: a randomized, double-blind, placebo-controlled phase IIb study (RISE-SSc) [abstract]. Arthritis Rheumatol 2018; 70(Suppl. 10): 903. [DOI] [PubMed] [Google Scholar]
  • 111. De Lauretis A, Sestini P, Pantelidis P, et al. Serum interleukin 6 is predictive of early functional decline and mortality in interstitial lung disease associated with systemic sclerosis. J Rheumatol 2013; 40(4): 435–446. [DOI] [PubMed] [Google Scholar]
  • 112. Nihtyanova SI, Denton CP. Autoantibodies as predictive tools in systemic sclerosis. Nat Rev Rheumatol 2010; 6(2): 112–116. [DOI] [PubMed] [Google Scholar]
  • 113. Shiwen X, Stratton R, Nikitorowicz-Buniak J, et al. A role of myocardin related transcription factor-A (MRTF-A) in scleroderma related fibrosis. PLoS ONE 2015; 10(5): e0126015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Young A, Vummidi D, Visovatti S, et al. Prevalence, treatment, and outcomes of coexistent pulmonary hypertension and interstitial lung disease in systemic sclerosis. Arthritis Rheumatol 2019; 71: 1339–1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Tomasek JJ, Gabbiani G, Hinz B, et al. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol 2002; 3(5): 349–363. [DOI] [PubMed] [Google Scholar]
  • 116. Duarte AC, Santos MJ, Cordeiro A. Anti-fibrotic nintedanib-a new opportunity for systemic sclerosis patients? Clin Rheumatol 2018; 37(4): 1123–1127. [DOI] [PubMed] [Google Scholar]
  • 117. Chua F, Desai SR, Nicholson AG, et al. Pleuroparenchymal fibroelastosis: a review of clinical, radiological and pathological characteristics. Ann Am Thorac Soc 2019; 16: 1351–1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Huang J, Maier C, Zhang Y, et al. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum Dis 2017; 76(11): 1941–1948. [DOI] [PubMed] [Google Scholar]
  • 119. Huang J, Beyer C, Palumbo-Zerr K, et al. Nintedanib inhibits fibroblast activation and ameliorates fibrosis in preclinical models of systemic sclerosis. Ann Rheum Dis 2016; 75(5): 883–890. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Scleroderma and Related Disorders are provided here courtesy of World Scleroderma Foundation, EUSTAR, and SAGE Publications

RESOURCES