Back to Journals » Journal of Inflammation Research » Volume 16

Biomarkers in the Pathogenesis, Diagnosis, and Treatment of Systemic Sclerosis

Authors Muruganandam M, Ariza-Hutchinson A, Patel RA, Sibbitt WL Jr 

Received 15 April 2023

Accepted for publication 27 September 2023

Published 17 October 2023 Volume 2023:16 Pages 4633—4660

DOI https://doi.org/10.2147/JIR.S379815

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Ning Quan



Maheswari Muruganandam, Angie Ariza-Hutchinson, Rosemina A Patel, Wilmer L Sibbitt Jr

Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA

Correspondence: Maheswari Muruganandam, Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, MSC 10 5550, 5th FL ACC, Albuquerque, NM, 87131, USA, Tel +1 (505) 272-4761, Fax +1 (505) 272-3624, Email [email protected]

Abstract: Systemic sclerosis (SSc) is a complex autoimmune disease characterized by vascular damage, vasoinstability, and decreased perfusion with ischemia, inflammation, and exuberant fibrosis of the skin and internal organs. Biomarkers are analytic indicators of the biological and disease processes within an individual that can be accurately and reproducibly measured. The field of biomarkers in SSc is complex as recent studies have implicated at least 240 pathways and dysregulated proteins in SSc pathogenesis. Anti-nuclear antibodies (ANA) are classical biomarkers with well-described clinical classifications and are present in more than 90% of SSc patients and include anti-centromere, anti-Th/To, anti-RNA polymerase III, and anti-topoisomerase I antibodies. Transforming growth factor-β (TGF-β) is central to the fibrotic process of SSc and is intimately intertwined with other biomarkers. Tyrosine kinases, interferon-1 signaling, IL-6 signaling, endogenous thrombin, peroxisome proliferator-activated receptors (PPARs), lysophosphatidic acid receptors, and amino acid metabolites are new biomarkers with the potential for developing new therapeutic agents. Other biomarkers implicated in SSc-ILD include signal transducer and activator of transcription 4 (STAT4), CD226 (DNAX accessory molecule 1), interferon regulatory factor 5 (IRF5), interleukin-1 receptor–associated kinase-1 (IRAK1), connective tissue growth factor (CTGF), pyrin domain containing 1 (NLRP1), T-cell surface glycoprotein zeta chain (CD3ζ) or CD247, the NLR family, SP-D (surfactant protein), KL-6, leucine-rich α 2-glycoprotein-1 (LRG1), CCL19, genetic factors including DRB1 alleles, the interleukins (IL-1, IL-4, IL-6, IL-8, IL-10 IL-13, IL-16, IL-17, IL-18, IL-22, IL-32, and IL-35), the chemokines CCL (2,3,5,13,20,21,23), CXC (8,9,10,11,16), CX3CL1 (fractalkine), and GDF15. Adiponectin (an indicator of PPAR activation) and maresin 1 are reduced in SSc patients. A new trend has been the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy. The present review is an update of the various biomarker molecules, pathways, and receptors involved in the pathology of SSc.

Keywords: biomarker, systemic sclerosis, scleroderma, cytokine, autoantibody

Introduction

Biomarkers are analytic indicators of the presence of a biological or disease process and/or activity of that process within an individual that can be accurately and reproducibly measured.1–11 Thus, a biomarker could be a physical measure such as blood pressure, a cytokine such as IL-1, a cell or cellular manifestation such as a membrane receptor, a metabolite such lactate, or other reproducible indicator of a biologic or medical process. Biomarkers have become an increasingly important aspect of systemic sclerosis (SSc) research and clinical care.12 The field of biomarkers in SSc is complex and evolving as recent studies have implicated at least 240 pathways and numerous dysregulated proteins in the pathogenesis of SSc.5–14

SSc is a complex autoimmune disease characterized by prominent fibrosis of the skin, macro- and micro-vascular damage, vasoinstability, and decreased peripheral blood perfusion with ischemia, inflammation, and exuberant fibrosis of internal organs including the lungs, kidneys, bowel, heart, and esophagus.15–17 The presence of skin thickening of the fingers extending proximal to the metacarpophalangeal joint is adequate to be classified as SSc. However, if proximal skin thickening is not present, the weighted addition of the following findings: skin thickening of the fingers, finger tip lesions (ischemic ulcerations or pitting scars), telangiectasia, abnormal nail fold capillaries, interstitial lung disease, pulmonary arterial hypertension, Raynaud’s phenomenon, and/or SSc-related autoantibodies (Table 1) may still permit the diagnosis of SSc.18 SSc is presently formally classified into two main forms: 1) limited cutaneous SSc characterized by mostly distal skin thickening and the presence of anti-centromere antibodies (Figure 1), and 2) diffuse cutaneous SSc with widespread distal and proximal cutaneous changes usually with the presence of anti-topoisomerase antibodies, anti-RNA-III polymerase antibodies or other anti-nucleolar pattern antinuclear antibodies.18–21 Nail fold capillaroscopy, which is now formally used to classify SSc, is a noninvasive and reliable method to detect diagnostic microvascular involvement in SSc.17,18,22 The presence of an ANA, recurrent Raynaud’s phenomenon, and nail fold capillaroscopic abnormalities predict progression to definite SSc.21 Anti-topoisomerase I antibodies also predict the development of diffuse cutaneous involvement with SSc and digital ulcers in the first 3 years of disease, as well as severe interstitial lung disease (ILD) (Figure 2).21,23

Table 1 Biomarkers in Systemic Sclerosis (SSc)

Figure 1 Sclerodactyly in SSC. The tightened skin extends beyond the metacarpophalangeal joints confirming the diagnosis of SSc. Also note contractures at the proximal interphalangeal joints and calcinosis cutis at the interphalangeal joint of the 5th digit (arrow).

Figure 2 Computed tomographic (CT) Image of Interstitial Lung Disease (ILD) in SSc. (A) Interstitial lung disease (ILD) with bilateral anterior upper lobe reticular opacities and dependent ground glass opacities and bronchiectasis. Central airways are clear. (B) Similar scattered ground glass opacities most predominant in the lower lobes with extensive bronchiectasis and cystic changes.

Mortality in SSc is significantly increased and usually related to life-threatening manifestations of SSc including interstitial lung disease, scleroderma renal crisis, pulmonary arterial hypertension, cardiac involvement, secondary malignancy, and infections from tissue necrosis, aspiration pneumonia, and immunosuppression.1,24

Autoantibodies as Biomarkers in SSc

Autoantibodies are the most commonly used biomarkers in SSc and are most specifically useful for the diagnosis, classification, and prognosis of SSc (Table 1); further, biomarkers can be potential therapeutic targets.2–4,21,25 Anti-nuclear antibodies (ANA) are present in more than 90% of SSc patients with anti-centromere, anti-Th/To, and anti-topoisomerase I antibodies considered as classical biomarkers occurring 60% of SSc and defining patients with well-described clinical classifications.18,21 Other autoantibodies are present in the majority of patients with SSc and besides ANA include autoantibodies directed against endothelial or fibroblast antibodies, angiotensin II type 1 receptor, endothelin-1 type A receptor, platelet-derived growth factor receptor (anti-PDGFR) and extracellular matrix (ECM) proteins.26–30 More complex autoantibody systems in SSc against G-protein-coupled receptors, growth factors and respective receptors have also been described.4 Ten percent of SSc patients are ANA-negative, but novel antibodies including anti-elF2B, anti-RuvBL1/2 complex, anti-U11/U12 RNP, Anti-U3RNP, anti-BICD2, Anti-Ku, and Anti-PM/Scl can be seen in both ANA positive and negative patients.7,31 The simultaneous presence of two different SSc-specific autoantibodies such as anti-centromere antibodies and anti-RNA polymerase III antibodies is so unusual that they are for practical purposes regarded as mutually exclusive, although there are always cases that may break the rule.32 For instance, a recent study by Clark et al demonstrated that only 5% of SSc patients had ≥2 any autoantibody positivity and only 2.3% had ≥2 SSc-specific antibody positivity, with the most common combination being anti-U1RNP and anti-topoisomerase I antibodies.33

ANA may not be just biomarkers of disease in SSc but may have a pathogenic role through immune-complex-mediated mechanisms and molecular mimicry. ANA (particularly anti-topoisomerase-I and anti-RNA polymerase III antibodies) appear to be transported into the cell with direct interaction with intercellular components and receptors, targeting intracellular topoisomerase and RNA polymerase by the corresponding antibodies.34,35 Anti-RNA polymerase III is a biomarker predicting rapid skin thickness progression, gastric antral vascular ectasia, SSc-associated cancers, scleroderma renal crisis, and possibly autoimmune syndromes associated with silicone breast implants.21,36 Further supporting this concept of pathogenic autoantibodies in SSc is the response of certain SSc patients to select anti-B cell therapies and the role of activated B cells in the successful allogeneic bone marrow transplant to treat SSc.37,38

Immunogenetic and HLA Antigens as Biomarkers in SSc

SSc has been reported previously to be weakly associated with a restricted number of class II antigens in the major histocompatibility complex (MHC), especially HLA-DR1, DR2, DR3, DR5, and DR52 (Table 1).39 However, these HLA-DR specificities that are weakly associated with SSc are in linkage disequilibrium with HLA-DQ alleles that are strongly associated with the specific autoantibodies peculiar to SSc. Thus, anticentromere antibodies occur most frequently in the presence of HLA-DQB1*0501 (DQ5), DQB1*0301 (DQ7) and similar DQB1 alleles characterized by a tyrosine or glycine residue in the outermost domain, specifically in position 26. Anti-topoisomerase I antibodies have been associated with the presence of HLA-DQB1*0301 (DQ7), DQB1*0302 (DQ8), DQB1*0601 and other DQB1 alleles evincing in position 30 a tyrosine residue. Anti-RNA polymerase III antibodies have been associated with HLA- DQB1*02:01, DRB1*04:05, DRB4*01, and DQB1*04:01.40,41 Recently, Class 1 HLA antigens have also been associated with SSc, in particular, the haplotype HLA-B*44:03-HLA-C*16:01 that interacts with the KIR locus suggesting that genetic modulation of lymphocyte activation also contributes to SSc onset.42,43 Specific KIR2 phenotypes appear to also promote human herpesvirus-6 (HHV-6) infection and reactivation and HHV-6 reactivation has been associated with fibrosis and the development and severity of SSc.44 Similarly, the immunodominant peptides of topoisomerase 1, fibrillarin, and centromere protein A are homologous to viral protein sequences from the Mimiviridae and Phycodnaviridae families, suggesting a virus-immune receptor interaction that may trigger specific autoantibody production and the subsequent development of SSc.45

Similarly, higher copy number polymorphisms of the complement C4 gene appear to provide less risk to developing SSc, and lower copy numbers of C4A and C4B provide augmented risk of developing SSc, with the serum levels of C4 protein paralleling the gene copy numbers and decreased or increased risk for developing SSc.46 Thus, C4 genetics are another immunogenetic factor that independently decreases and increases the risk of SSc along with amino acid variants of HLA-DRB1 and HLA-DQB1.

Other Biomarkers in SSc

Since the immune system and healing mechanisms of the human body are generally activated in SSc, multiple molecules are increased or decreased in SSc (Table 2) depending on the presence, activity, and therapy of the disease.1–10 Toll-like receptors (TLRs) recognize pathogens and internal activation signals resulting in activation of multiple pathways that finally result in inflammation and alternations in innate immunity that occur with SSc.47 Internal activation signals include damage-associated molecular patterns (DAMPs) that are intracellular molecules released under significant tissue injury or cellular stress and bind as endogenous ligands on TLRs.48 DAMPs-TLR interaction on fibroblasts directly activates these collagen-producing cells to generate large amounts of collagen contributing to ECM expansion typical of SSc and the complications of SSc.47 Further, ligand activation of TLR on dendritic cells results in increased production of Th17-related cytokines including IL-1β, IL-17F, IL-21 IL-22, and IL-33 resulting in aberrant T cell polarization and profibrotic inflammation.49

Table 2 Non-Antibody Protein Biomarkers in SSc

Transforming growth factor-b (TGF-β) is central to the process of fibrosis as well as dysregulation of the immune system toward inflammation.50–52 Injured or stressed cells produce TGF-β that recruits and stimulates macrophages that secrete more TGF-β that then upregulates genes responsible for ECM production and progressive fibrosis.53,54 Serum cytokeratin 17 (CK17), marginal zone B1 protein (MZB1) and leucine-rich α2-glycoprotein-1 (LRG1) are potential biomarkers for SSc, with CK17 negatively associated with SSc disease severity, with higher CK17 values being protective.10 Endostatin has been associated with vascular manifestations in SSc and is specifically elevated in progressive SSc and has been considered as a marker of SSc severity and potentially as a therapeutic target.55 Periostin is secreted by fibroblasts and epithelial cells and is associated with cell adhesion, fibrosis, angiogenesis, survival, and matrix remodeling.56,57 Circulating levels of periostin are elevated in SSc and are associated with disease duration, skin fibrosis, and cardiomyopathy.58 CC chemokine 2 (CCL2) has been implicated in the development of fibrosis in SSc.59

MicroRNAs (miRNAs) are short nucleotide sequences involved in cellular regulation. The microRNAs miR-138 and miR-27a suppress major pathways involved in epithelial to mesenchymal cell transition and subsequent fibrosis, and the relative expression of miR-138 and miR-27a are significantly lower in patients with SSc compared to controls, while only miR-138 is further depressed in diffuse cutaneous SSc, thus potentially both could be used as a diagnostic biomarker with miR-138 specifically to predict severity.60–62 The suppression of tumorigenicity 2 (ST2) receptor binds IL-33 and the serum soluble ST2 (sST2) suppresses IL-33 signaling.63 Elevated serum levels of sST2 serum levels are associated with higher articular disease activity and greater hand dysfunction in SSc, indicating that sST2 might be a biomarker to predict SSc articular involvement.64

Angiopoietins (Ang-1 and Ang-2) interact with the specific tyrosine kinase receptor Tie2 to modulate endothelial cell activation, vascular modeling, and angiogenesis.65 Ang-1 is decreased and Ang-2 increased in SSc patients relative to controls, and this imbalance may contribute to both vascular ablation and the formation of abnormal new blood vessels.

An increased activation and expression of type 1 interferons are typical in SSc and appear similar to interferon abnormalities observed in systemic lupus erythematosus and may similarly provide a potential therapeutic target.66–68 Increased IFN-I is associated with anti-U1-RNP antibodies, anti-topoisomerase antibodies, and more aggressive lung, skin, and skeletal muscle involvement.69.70 Dendritic cells in the tissues of patients with SSc are typically activated and produce IFN-α and CXCL4 with the unexpected presence of TLR8, an RNA-sensing TLR.50,69 CXCL4 modulates IFN-I responses by metabolic reprogramming of dendritic cells and an increased fibrotic phenotype.70 Similarly, an increased expression of IFN-I–associated genetic loci correlates with a higher ILD progression rate.54,68 The IFN-regulated protein sialic acid–binding Ig-like lectin 1 (SIGLEC-1) is upregulated in SSc compared to controls but does not associate with specific complications.71

Drugs that interfere with INF-1 signaling and thus the downstream activation of tyrosine kinases, inflammation, and fibrotic processes have considerable potential for the therapy of SSc.72 Anifrolumab is a monoclonal antibody directed against IFN-I receptor subunit 1 and has been shown to be effective in systemic lupus erythematosus.68 In early clinical trials, anifrolumab has also been shown to clinically decrease fibrosis in SSc and is associated with upregulation of type III collagen degradation markers and downregulation of T cell–associated proteins both suggesting a reduced fibrotic state.73

Tyrosine kinases that are activated downstream by INF-1 signaling include receptor-activated tyrosine kinases (for example, receptor kinases activated by growth factors - platelet-derived growth factor, fibroblast growth factor, or vascular endothelial growth factor) and nonreceptor tyrosine kinases (c-Abl, Src, Janus, and STATs).74 Receptor-activated tyrosine kinases and their ligands are implicated in the migration, proliferation, ECM secretion, differentiation, and contraction of fibroblast precursors, tissue fibroblasts and myofibroblasts in SSc, and thus are intimately involved in the fibrotic process.75–77 Nintedanib, an inhibitor of receptor activated tyrosine kinases, has been shown to be effective in reducing pulmonary fibrosis in SSc and down-regulating fibrotic processes, confirming the role of receptor activated tyrosine kinases in the pathologic processes of SSc.74–77

Similarly, downstream from INF-1 signaling in SSc is increased levels of IL-6 in SSc that ultimately activate nonreceptor tyrosine kinases especially Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) proteins resulting in fibroblast differentiation, proliferation, and ECM and collagen production, providing promising therapeutic targets.51,78,79 In line with these observations, tocilizumab, an IL-6 receptor blocker shown to be effective in ILD of SSc, reduces biomarkers of inflammation, ECM turnover, and macrophage activation, including collagen degradation and formation neoepitopes.80,81 Further, there is early evidence that the JAK inhibitors tofacitinib and baricitinib reduce fibrosis in SSc in both lungs and skin, consistent with the known role of JAK/STAT activation in fibrosis in SSc.82–86

SSc manifests extensive endothelial injury, upregulation of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), E-selectin, a von Willebrand factor (vWF), tissue factor (TF), and tissue thrombin resulting in local hypercoagulability.85,86 Enhanced expression of thrombin stimulates endothelial cells and fibroblasts, resulting in enhanced collagen production, reduced matrix metalloproteinases, and expansion of the ECM contributing to the fibrosis of SSc. SSc is characterized by increased endogenous thrombin potential, thrombin expression, and endothelial damage biomarkers as well as lower thrombomodulin, fibrinolysis, and platelet levels associated with vasculopathy and digital ulcers.84,85 These findings suggest that thrombin inhibitors might be useful for SSc, and early human studies using the thrombin inhibitor dabigatran preliminarily demonstrated reasonable safety and reduced skin thickness, suggesting that thrombin inhibitors are a viable therapeutic possibility.86

Peroxisome proliferator-activated receptors (PPARs) are ligand-responsive transcription factors of the nuclear hormone receptor family.87 PPAR- γ is in particular an anti-inflammatory receptor that down-regulates activation of macrophages, lymphocytes, and dendritic cells. PPAR-γ reduces fibrogenesis by interfering with the TGFβ pathway by inhibiting the TGFβ driven differentiation of fibroblasts into active myofibroblasts, thus limiting collagen and ECM formation and decreasing tissue contraction.88,89 Thus, impaired PPAR-γ expression or function may contribute to the excessive fibroblast activation and fibrosis of SSc. Expression of PPAR-γ is markedly diminished in skin tissues, lung biopsies, and fibroblasts from patients with SSc, and PPAR-γ appears to be downregulated by excessive expression of TGF-ß.90 Adiponectin, a marker for PPAR-γ activation is also decreased in SSc, and these reduced levels are associated with the degree of skin fibrosis, suggesting that both adiponectin and membrane PPAR-γ are potential biomarkers for SSc.91 However, circulating PPAR-γ is increased in SSc, suggesting a ligand scavenger effect that prevents membrane PPAR-γ activation thus promoting fibrogenesis.92 PPAR-γ agonists reduce lung fibrosis and skin thickness in animal models of SSc, suggesting that the PPAR-γ pathway and PPAR-γ agonists are important potential therapeutic approaches.93

Lysophosphatidic acid (LPA) is a lipid mediator that is generated by the enzyme lysophospholipase (autoxain) wherever there is cell injury or inflammation, and LPA binds to G protein-coupled LPA receptors (LPAR1–6).94 LPAR1 after activation mediates the physiological effects of LPA including migration, survival, and infiltration of macrophages, activation of inflammasomes, release of proinflammatory mediators, including IL-1 and IL-18, promoting differentiation of mesenchymal cell to myofibroblasts, and collagen excretion.52 Circulating LPA is increased in SSc patients compared to controls and thus is a natural biomarker and target for treatment of SSc.94,95 Treatment of SSc patients with an LPAR1 inhibitor resulted in decreases in skin scores and a reduction in LPA-related activation genes, indicating a potentially useful intervention, although more clinical trials are required.96

SSc is generally characterized by the abnormal accumulation of ECM.1,97 ECM catabolism is regulated by matrix metalloproteinases (MMP-1 to MMP-28) whose activity is in turn inhibited by tissue inhibitors of MMPs (TIMP-1 to TIMP-4).98,99 In this line, the inhibitor, TIMP-4, is increased in SSc patients with respect to healthy subjects.100 Similarly, semaphorins (Sema3A-F) have anti-angiogenic effects and are increased in SSc patients.101,102

The cytokines, interleukins and chemokines are often elevated in SSc. IL-1, IL-4, IL-6, IL-10 IL-13, IL-16, IL-18, IL-22, IL-32, and IL-35 have all been reported to be elevated in SSc.103–105 IL-17A is not specifically increased, while IL-17B, IL-17E, and IL-17F are higher SSc.106 The chemokines CCL2,3,5,13,20,21,23, CXC8,9,10,11,16, CX3CL1 (fractalkine), and growth differentiation factor-15 (GDF-15) are all increased in SSc.11,104,107–111 Adiponectin and maresin 1 are reduced, while galectin 3 is higher in SSc patients.112–114 Thrombomodulin (TM) and cluster of differentiation 163 (CD163) are elevated in SSc.115,116 Salusin-alpha, a regulator of secretion of pro-inflammatory cytokines and vascular smooth muscle proliferation, is increased in SSc.117 The nuclear receptor subfamily 4A (including NR4A1, NR4A2, and NR4A3) are important down regulators of inflammation and fibrosis in SSc, and enhancing the expression of these receptors may be an approach to treating SSc.118

Recently, metabolic profiling has also be applied to SSc, demonstrating alterations in homocysteine, proline, alpha-N-phenylacetyl-L-glutamine, glutamine, asymmetric dimethylarginine, citrulline and ornithine, kynurenine, tryptophan, acylcarnitines associated from long-chain fatty acids, and tricarboxylic acids such as citrate and succinate with differences between the different SSc subtypes.119

In the following sections the role of biomarkers relevant to specific organ system disease in SSc will be discussed.

Interstitial Lung Disease (ILD) in SSc (SSc-ILD)

SSc is frequently complicated with interstitial lung disease (ILD) as a major contributing cause of both mortality and disability.15,16 Approximately 65–85% of patients with SSc develop ILD (SSc-ILD) of variable severity and 25–30% develop aggressive disease that is associated with the significant mortality of 40% over a 10-year period.1,19,120,121 Lung involvement results in an estimated 35% of all SSc-related deaths.122 Risk factors of SSc-ILD include male sex, diffuse cutaneous SSc, African American heredity, and the presence of anti–Scl-70 (anti-topoisomerase I) antibodies.123,124

Lung endothelial damage is central to the pathogenesis of ILD. The typical histologic pattern in SSc-ILD is nonspecific interstitial pneumonia unlike idiopathic pulmonary fibrosis that is usually a usual interstitial pneumonia pattern.1 SSc-ILD is characterized by inflammation early in the disease, extensive endothelial dysfunction, and increased deposition of ECM, especially collagen produced by activated myofibroblasts in resident tissues.1,12,20 The increased ECM increases the stiffness of lung tissues resulting in restrictive lung disease with reduction of lung compliance, decreased lung volumes and decreased diffusion capacity resulting in impaired exercise tolerance, dyspnea, fatigue, hypoxia, increased pulmonary artery pressures, work disability, and reduced life expectancy. The pathological process is believed to be initiated by repetitive epithelial and endothelial cell injury with activation of the immune system, recruitment of fibroblasts, and phenotypic transformation of the fibroblast to a myofibroblast that then secretes excessive ECM resulting in fibrosis.125 The initial endothelial and epithelial injuries are likely to be autoimmune and inflammatory in nature but could be induced also by pathogens and environmental factors.126 Apoptosis occurs in certain epithelial cells denuding the alveoli, and simultaneously other epithelial cell transition into myofibroblasts with reduced apoptosis, loss of polarity, increased migration, and increased production of ECM, including collagen.53 The reduced apoptosis of myofibroblasts may cause abnormal persistence of these active cells, contributing the progressive fibrosis.53

SSc-ILD is detected after diagnosis by high-resolution CT (HR-CT) and progression by both HR-CT and pulmonary function tests (PFT) (Figure 2).127–129 Quantitative CT has also been used to detect early SSc-ILD and differentiate it from interstitial pneumonia and to more precisely follow the progression of SSc-ILD.130 Recently, intercostal ultrasound has also been used to screen for SSc-ILD to detect characteristic B-lines and subpleural disease.131 Significant or progressive SSc-ILD usually prompts therapy. The PFT-based OMERACT (outcome measures in rheumatic diseases) detects progression of SSc-ILD defined as ≥10% decline in forced vital capacity (FVC) or ≥5% to <10% decline in FVC with ≥15% relative decline in DLCO.129,132 It should be realized, however, that with effective therapy for SSc-ILD, the lung fibrosis is stabilized or the rate of decline of lung function is reduced, rather than complete restoration of lung function.20 Thus, the emphasis for earlier diagnosis and prompt therapy of SSc-ILD before significant irreversible lung damage has accumulated.

The general treatment guidelines for SSc-ILD have recently been reviewed and revised.133–136 Drugs used to treat SSc-ILD include non-specific immunosuppressives (cyclophosphamide, mycophenolate), specific immunosuppressive drugs including anti-IL-6 agents (tocilizumab), anti-B-cell drugs (rituximab), and antifibrotic agents (nintedanib - a tyrosine kinase inhibitor).20,137,138 The non-specific immunosuppressives, mycophenolate, an inhibitor of the synthesis of guanosine nucleotides, and cyclophosphamide, an alkylating agent, decrease proliferation of fibroblast, T-helper cells, and B-cells, and thus, have significant anti-fibrotic effects.139,140 Indeed, mycophenolate is currently considered the standard and basic underlying therapy for SSc-ILD.20 Tocilizumab is increasingly used for SSc-ILD. Tocilizumab inhibition of the IL-6 receptor decreases myofibroblast activation and reduces M2 macrophage polarization, both important to the antifibrotic effects of tocilizumab.141,142 B cell depletion suppresses pro-fibrotic macrophage differentiation and thus inhibits fibrosis, providing an additional rationale for unapproved but often used anti-B cell agents such as rituximab in SSC-ILD.143 The anti-fibrotic drug nintedanib, a tyrosine kinase inhibitor, inhibits the receptors of PDGF, FGF, and vascular endothelial growth factor (VEGF), reducing fibrosis.75 However, the anti-fibrotic pirfenidone appears to have less beneficial effects in SSc and is presently not approved for treatment of this entity.144 JAK inhibitors have been used and seem to be effective for refractory cases of SSc-ILD, but remain off-label.51 For rapidly progressive disease non-responsive to these agents or for very early SSc-ILD, an alternative is autologous haematopoietic stem cell transplant (AHSCT) and for end-stage lung disease lung transplantation.14,145,146

SSc-ILD has been especially associated with anti-topoisomerase I antibody (anti-Scl-70 antibody), anti-U11/U12 antibodies, nucleolar pattern antinuclear antibodies (including anti-RNA-polymerase III, anti-NOR-90, anti-PM/Scl-75, anti-U3-RNP/Fibrillarin anti- antibodies) (Table 3).1,147,148 Anti-PM/Scl defines SSc patients with high frequency of ILD, calcinosis, dermatomyositis skin changes, and severe myositis.21 Further autoantibodies against anti-phosphatidylinositol-5-phosphate 4-kinase type 2 beta (PIP4K2B) and AKT serine/threonine kinase 3 (AKT3) have been tied to increased lung fibrosis in SSc.149

Table 3 Biomarkers in SSc-Interstitial Lung Disease (SSc-ILD)

Transforming growth factor-b (TGF-β) is produced by stressed or injured lung cells that stimulate and recruit tissue macrophages that amplify TGF-β production that then up upregulates genes responsible for ECM production and progressive fibrosis.53,54,113,150 Other biomarkers typically implicated in SSc-ILD include STAT4, CD226 (DNAX accessory molecule 1), interferon regulatory factor 5 (IRF5), interleukin-1 receptor–associated kinase-1 (IRAK1), connective tissue growth factor (CTGF), pyrin domain containing 1 (NLRP1), T-cell surface glycoprotein zeta chain (CD3ζ) or CD247, the NLR family, SP-D (surfactant protein), KL-6 (Krebs von den Lungen-6), IL-8, LRG1, and CCL19, as well as genetic factors including DRB1 alleles.1,7,55,127,151–156 The biomarkers most associated with active lung disease and progression specifically in SSc-ILD are KL-6, SP-D (surfactant protein), C-reactive protein, and CCL19 although other non-specific biomarkers can also be elevated.1,152 Th2-lymphocytes produce IL-13 and IL-4 that stimulate fibroblasts and activate the pro-fibrotic M2 macrophages that induce TGF-β, platelet-derived growth factor (PDGF), and fibroblast growth factors (FGF) inducing myofibroblast activation.104 The biomarker chemokines including CCL18, CX3CL1 and CXCL4 with and without RNA complexes have recently been associated with SSc-ILD.59,108,152,155,157

IL-6 is important in the progression of SSc-ILD and is secreted by myofibroblasts, M1 macrophages, and B-cells.15,143 IL-6 enhances the expression of IL-4 and IL-13-receptors increasing polarization of M2 macrophage and enhancing fibrosis.158,159 This central role is further supported by tocilizumab inhibition of the IL-6 receptor decreasing myofibroblast activation and reduced M2 macrophage polarization, resulting in a portion of the demonstrated antifibrotic effects of tocilizumab in the setting of SSc-ILD.141 B-cell activation is also common in SSc, and B cells increase a number of angiogenic factors.160 B cell depletion suppresses pro-fibrotic macrophage differentiation and thus inhibits fibrosis, providing rationale for anti-B cell agents, such as rituximab in SSC-ILD.143

Of the above biomarkers, autoantibodies and C-reactive protein are the only biomarkers typically used in contemporary routine clinical practice. In that line, if anti-topoisomerase-I antibodies are present and the anticentromere antibodies are not present, there is an increased incidence of progressive SSc ILD.1,124,161 However, various other biomarkers are being explored for clinical use including KL-6, CCL18 (chemokine [C-C motif] ligand 18), MMP7 (matrix metalloproteinase-7), MMP12 (matrix metalloproteinase-12), IL-6, CXCL4 (chemokine [C-X-C motif] ligand 4), CXCL3 (chemokine [C-X-C motif] ligand 4), and chitinase-3–like protein 1(YKL-40) as recently reviewed.1,132 MMP-12 are raised in SSc-ILD compared with SSc without ILD and correlated with the degree of pulmonary fibrosis.162 Sirtuins are NAD-dependent protein deacetylases that regular angiogenesis; SIRT1 and SIRT3 correlate with the degree of lung fibrosis in SSC.162 The chemokine CCL2 is increased in SSc and predicts long-term progression of SSc-ILD.163 The cold-inducible RNA-binding protein (CIRP) was also associated with SSc-ILD and may be a marker of disease activity and response to therapy.164 Jee et al have recently described a composite biomarker index consisting of SP-D, Ca15-3 and ICAM-1 that identifies SSc-ILD.165 The use of biomarker panels, composite biomarker measures, machine learning, and artificial intelligence is a growing trend in the field.165–167

In conclusion, multiple arms of the immune system are activated in SSc-ILD, providing many candidate biomarkers and potential therapeutic targets with the trend being the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy.

Vascular Injury in SSc Including Pulmonary Arterial Hypertension (PAH)

The most obvious clinical manifestation of vascular disease in SSc is Raynaud’s phenomenon (RP) and digital ischemia.168, RP is followed by telangiectases, ischemic digital ulcers, pitting scars, periungual microvascular abnormalities, pulmonary arterial hypertension (PAH) (Figure 3), and cardiac disease affecting function and exercise tolerance.15,169–171 All are considered outcomes of vascular injury in SSc as recently reviewed by Pattanaik et al.172

PAH occurs in 7% to 19% of SSc patients depending on the population and duration of the disease.19 Risk factors for PAH include severe Raynaud’s phenomenon, severe digital ischemia, cutaneous telangiectasia, chronic disease, late onset of disease, older age, postmenopausal status, reduced diffusion capacity (DLCO < 50%), DLCO/alveolar volume less than 70%, forced vital capacity/DLCO less than 1.6 and an elevation in right ventricular systolic pressure greater than 2 mmHg/year.173 Screening should include specific autoantibodies (anti-topoisomerase I (SCL-70), anti-centromere and anti-RNA polymerase III and antiphospholipid antibodies), pulmonary function tests, echocardiography, serum N-terminal pro-brain natriuretic peptide (NT-proBNP), nail fold capillaroscopy, and initial high-resolution CT to exclude ILD, and if there is a question of PAH, a right heart catheterization to determine PA pressure.174–178 Treatment of PAH in SSc has recently reviewed with guidelines and includes a stepwise approach using single agents or combined therapy with phosphodiesterase type 5 (PDE-5) inhibitors (including sildenafil and tadalafil), soluble guanylate cyclase (sGC) stimulators (including riociguat), endothelin receptor antagonists (including bosentan, ambrisentan, and macitentan), prostacyclin analogs (epoprostenol, treprostinil and iloprost), or selective prostacyclin IP receptor agonists (selexipag) supported by anticoagulants, diuretics, digoxin, and calcium channel blockers where appropriate.133,174,178,179

Figure 3 Computed tomographic (CT) Image of the Mediastinum. CT angiographic image of thorax in a SSc patient demonstrating a markedly dilated pulmonary artery (arrow) without thrombus consistent with pulmonary artery hypertension (PAH).

Raynaud’s phenomenon (RP) can occur without a systemic disease (primary RP), but RP is an almost universal SSc-associated phenomenon with a characteristic 3-phase color change associated with symptoms of pain, burning, ischemia, and in some cases, ischemic ulcers and/or necrosis of the digits.163,180,181 Bernero et al have demonstrated that a large proportion of initially primary RP progress to secondary RP – that is they eventually develop a definite autoimmune disease such as SSc or other autoimmune disease.181 The pathophysiology of RP is similar to the other mechanisms of vascular injury in SSc discussed in detail in this section. New methods to quantify RP beyond nailfold capillaroscopy include laser Doppler imaging, laser speckle contrast analysis, thermal imaging, and color and spectral Doppler imaging.182,183 Pharmacologic therapy for RD has recently been reviewed and is generally initiated first with calcium channel blockers, and if ineffective followed by PDE-5 inhibitors or fluoxetine with endothelin-1 receptor blockers and intravenous prostanoids reserved for the most resistant cases.133–136,182,184

Vascular injury and subsequent fibrosis may occur from the activation and apoptosis of endothelial cells, specific autoantibodies, infectious agents, endogenous hypercoagulability, reactive oxygen species, as well as other causes providing many potential biomarkers (Table 4).84,185 Once activated endothelial cells secrete endothelin-1 (ET-1), von Willebrand factor (vWF), nitric oxide, and endothelial nitric oxide synthase, resulting in instability of the vascular tone with less vasodilation and more vasoconstriction causing tissue ischemia and hypoxia.186–188 Endothelin-1 stimulates fibroblasts to convert to activated myofibroblasts with increased ECM secretion, intimal hyperplasia, luminal narrowing, reduced capillary blood flow vessel obliteration, and ischemia.189 Local secretion of von Willebrand factor causes platelet aggregation, hypercoagulability, and fibrin deposition leading to terminal vascular damage.186–188 Myofibroblasts are also created by the endothelial-to-mesenchymal transition.128 Activated endothelium also expresses increased adhesion molecules and specific chemokines, recruiting immune cells and perivascular infiltrates leading to further inflammation and fibrosis.186–190 Notably, angiogenesis is also reduced due to imbalanced cytokines including endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-9, endoglin, ET-1) and angiostatic (pentraxin 3 (PTX3), MMP-12, endostatin, angiostatin, semaphorin3E (Sema3E), and Slit2) factors and the dysfunction and impaired recruitment of endothelial progenitor cells (EPCs).169,191–193

Table 4 Biomarkers in SSc-Pulmonary Arterial Hypertension (PAH)

Enhanced endogenous thrombin generation potential and higher thrombin peak are present in SSc, accompanied by increased inflammatory markers, increased factor VIII activity, blood eosinophilia, thrombocytopenia, reduced VCAM-1, and lower thrombomodulin, indicating an important role for the thrombin system in vascular injury of SSc.84 Dabigatran, a direct thrombin inhibitor, is well tolerated in SSc patients with ILD and appears to show some improvements, but long-term clinical trials still need to be performed.86

Peroxisome proliferator-activated receptor-gamma (PPAR) is an important regulator of fibroblast growth, ECM formation, and connective tissue remodeling.87–92 PPAR interferes with the TGFβ pathway by inhibiting the ability of TGFβ to induce fibroblasts from differentiating into myofibroblasts and inhibiting collagen production via the transcriptional coactivator system.89 PPAP activity and expression of adiponectin, a sensitive and specific index of PPAR activity, are both reduced in SSc and associated with more progressive skin fibrosis.91 Circulating levels of PPAR are increased in SSc, particularly in diffuse cutaneous SSc with increased skin fibrosis, suggesting a defect in PPAR expression in SSc that interferes with activation of membrane bound PPAR enhancing fibrosis.92 PPAR agonists are effective in reducing fibrosis in animal models of SSc; thus, activation of PPAR and levels of adiponectin have potential as biomarkers for fibrotic activity in SSc.93,194

Selectins are molecules that permit cell trafficking and cell homing. L-selectin (CD62L) is expressed on leukocytes, P-selectin is expressed on platelets, and E-selectin is present on endothelial cells and megakaryocytes, but there have been inconsistent results regarding selectins in SSc104 immunoglobulin-like cell adhesion molecules including intercellular adhesion molecules (ICAMs), vascular cell adhesion molecules (VCAMs), and junctional adhesion molecules (JAMs). Circulating levels of ICAM-1 and VCAM-1 are higher in SSc patients with digital ulcers than without but may not be predictive of the occurrence of new DUs in SSc.195 Similarly, VCAM-1 can be elevated in SSc both with and without PAH.190 JAMs are adhesion molecules on endothelial cells, fibroblasts, epithelial cells, and blood cells, and circulating levels have been associated with microvascular disease and digital ulcers.102,190 Soluble VCAM-1, ICAM-1, and P-selectin are elevated in SSc-PAH compared to controls and with effective therapy with bosentan return to normal values.196

The vascular endothelial growth factor (VEGF) family includes the VEGF-A, -B, -C, and –D, and placental growth factor and are elevated in both the blood and skin in SSc.8,66,166,195,197, VEGF levels also correlate with PAH but are lower in patients with digital ulcers.129,198,199 TGF-β remains important to all the manifestations of SSc including PAH.200 Endoglin (CD105) is an accessory receptor for TGF-β and higher circulating endoglin correlated with digital ulcers, suggesting that this was a biomarker for vascular injury in SSc.201

Endothelin-1 (ET-1) is secreted by endothelial cells and activated smooth muscle cells, fibroblasts, epithelial cells, and inflammatory cells.202 Increased ET-1 is present in SSc-PAH when compared to SSc patients without PAH and healthy controls.203 Treatment with bosentan decreased ET-1 in SSc patients with PAH to levels present in SSc without PAH, indicating that this biomarker could detect the severity of vascular injury and the response to bosentan therapy.203

Pentraxin 3 is a receptor produced by activated endothelial cells, macrophages, smooth muscle cells, dendritic cells, and fibroblasts.204 However, levels of pentraxin 3 have only variable associations with vasculopathy in SSc204 Endostatin, is an angiostatic peptide that blocks VEGF activity and has been associated with PAH, scleroderma renal crisis, and cardiac involvement11 Angiostatin antagonizes a number of growth factors, including VEGF and is elevated in patients with more advance vascular disease in SSc11,205 Matrix metalloproteinases break down ECM and levels of MMP-9 are decreased in SSc-PAH and upregulated with bosentan therapy, while MMP-12 was increased patients with digital ulcers and nailfold capillary abnormalities.206,207

SSc-PAH is associated with elevated tissue inhibitors of metalloproteinases including TIMP-4 levels, indicating a cardiopulmonary vasculature-specific role of TIMP-4 activation in SSc.100 Neuropilins (NRP1-2) are non-tyrosine kinase glycoprotein receptors expressed on endothelial cells and are potential biomarkers predicting PAH, nailfold capillary abnormalities, and digital ulcers.102,166 The slit glycoproteins (Slit1-3) are implicated in angiogenesis and are increased in SSc and in patients with microvascular disease.208 Sirtuins (SIRT1-7) NAD-dependent protein deacetylases that regulate angiogenesis; SIRT1 and SIRT3 are decreased in SSc and microvascular disease, and SIRT3 specifically is related to the presence of digital ulcers.162 Carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-positive monocytes are associated with inflammation and ILD in SSc patients.209

Circulating IL-18 binding protein levels are higher SSc positively correlated with PAH.103 Similarly, IL-33 and soluble suppression of tumorigenicity 2 (ST2) are increased in SSc especially with digital ulcers and PAH.210 IL-32 and macrophage migration inhibitory factor (MIF) are elevated in SSc patients with PAH.211 Chemokines CCL20, CCL21, and CCL23 are also elevated in SSc-PAH.107–109 Elevated levels of CXCL4 and reduced levels of CXCL5 have recently also been associated with SSc-digital ulcers.212,213 Similarly, the chemokines CXCL16 and GDF15 are elevated in SSc-PAH.110,197 CX3CL1 (fractalkine) is elevated in SSc with digital ulcers.111 Resistin is increased in SSc with digital ulcers and in SSc-PAH.214 Galectin 3 was also found to be higher in SSc patients with digital ulcers.68 Adipsin, visfatin, interferon-gamma, and type 1 interferons are elevated in SSc-PAH.66,215–217 Aptamer proteomics of serum exosomes define patterns that could distinguish primary Raynaud’s disease from early SSc and with RNA networks are potential biomarkers for vascular disease in SSc.180,218,219

Thrombomodulin (TM), CD163, and NT-proBNP are elevated in SSc-PAH.115,116,220 Elevated levels of maresin 1 are associated with the development of digital ulcers in SSc.50 Elevated asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide (NO) inhibitor that affects endothelial function and is elevated in microvascular disease in SSc.221,222 Hypochromic erythrocytes have been closely associated with the prognosis of SSC-PAH.223 Hemoglobin and ferritin are significantly lower in patients with pulmonary hypertension (PH) in SSc compared to those with pulmonary hypertension, while uric acid and NT-proBNP are significantly higher.224 Circulating CCL21 in SSc is a biomarker associated with PAH and the development of PAH.108

Many molecules have been associated with the vascular complications of SSc, thus there are many potential biomarkers for PAH and vascular disease in SSc.

The Skin in SSc

Skin thickening is universal in SSc and generally necessary for a definite diagnosis with certain exceptions18 There are many candidate biomarkers for skin disease in SSc (Table 5). Generally, anti-topoisomerase-I and anti-RNA polymerase III antibodies are associated with more extensive and severe skin involvement, while anti-centromere antibodies are associated with limited skin involvement.1,2,4,225 Further, autoantibodies against PIP4K2B and AKT3 have been tied to more extensive skin fibrosis in SSc.149 In skin biopsies, TGF-β1, TGF-βR1, and TGF-βR2 expression levels are higher in SSc patients than controls.226 IL-6 and JAK associated pathways are also implicated in the skin thickening both directly and by response to specific inhibitors that block these pathways.227,228 Skin gene-expression of macrophage-associated biomarkers (CD14, IL13RA1) and TGF-β- associated biomarkers (OSMR SERPINE1, CTGF) are associated with progression of skin disease in SSc.229 Marginal zone B1 protein (MZB1) appears to be a good biomarker for skin fibrosis.10 Circulating levels of periostin are elevated in SSc with extensive skin fibrosis.58 Sirtuins are NAD-dependent protein deacetylases that regular angiogenesis; SIRT1 and SIRT3 correlated with the degree of skin fibrosis in SSC.162 Adiponectin is reduced in SSc involved skin.114 Recently, the fibrillar collagen molecule COL4A1, the matricellular protein COMP, the gene coding for spondin SPON1, TNC, and another ECM protein were upregulated in SSc skin and completely distinguished SSc from normal skin.230

Table 5 Biomarkers in SSc Skin Disease

Skin thickness in SSc often improves spontaneously over time, confounding many interventional trials as recently reported with belimumab and nintedanib.231–233 However, contemporary treatment guidelines note that mycophenolate, cyclophosphamide, and methotrexate have been demonstrated to improve the modified Rodnan skin score and reduce skin thickness in SSc.133,178,179,234,235 Methotrexate can be problematic in SSc since it can occasionally cause lung inflammation that can be confused with SSc-ILD.236 Recently, the JAK inhibitor tofacitinib, although not approved for SSc, was shown to be more effective than methotrexate in decreasing the modified Rodnan skin score, skin thickness by ultrasound, and musculoskeletal symptoms and reduced interferon regulated biomarker genes in SSc.142,237 Ziritaxestat is a small-molecule selective autotaxin inhibitor and reduced the modified Rodnan skin score in SSc and thus is a promising new agent currently under clinical trials.238 There is some evidence that rituximab and tocilizumab also improve skin scores in SSc.81,227,228,239,240

Skin ulceration in SSc is classified as calcinosis-related, traumatic, or ischemic.241 Treatment of skin ulceration in SSc includes avoiding vasoconstrictors (eg, caffeine, amphetamines, cocaine, nicotine), cold temperature, and trauma, the cessation of tobacco products, and appropriate wound care. Ulcerations from calcinosis cutis in SSc develop from hydroxyapatite crystal deposition within the subcutaneous tissues and may be treated with surgical excision or debridement, topical antibiotics, low-dose tetracyclines (minocycline or doxycycline) or topical or intravenous sodium thiosulfate.242,243 Vasodilators to treat and prevent skin ulceration are similar as used for Raynaud’s phenomenon and include calcium channel blockers (especially amlodipine and nifedipine), topical nitroglycerin ointment, and phosphodiesterase inhibitors (cilostazol, sildenafil).244,245

The Gastrointestinal System in SSc

The gastrointestinal system involvement in SSc is profound and includes bowel and esophageal dysmotility and fibrosis, bowel ischemia, primary sclerosing cholangitis, primary biliary cirrhosis, bacterial overgrowth, increased bowel malignancies, and bowel inflammation amongst other complications.246 Outcomes are usually anatomically specific; however, a gastrointestinal patient reported outcome, the Scleroderma Clinical Trial Consortium GIT 2.0 has been used recently.247 Treatment of gastrointestinal complications is typically focused on individual problems of gastroesophageal reflux (proton pump inhibitors, H2-blockers, sucralfate), stricture (dilation), dysmotility and bacterial overgrowth (erythromycin, azithromycin, metoclopramide, domperidone, cisapride).248 In the line of biomarkers, anti-U11/12 antibodies have been associated with severe gastrointestinal dysmotility.31 Further, there are elevated fecal levels of the inflammatory biomarker calprotectin in SSc, suggesting that fecal calprotectin could be an effective biomarker for bowel disease as it is in other inflammatory bowel diseases.249,250

Biomarkers of Renal Disease in SSc

Renal manifestations of SSc are dominated by scleroderma renal crisis (SRC), while true immune-mediated glomerulonephritis and interstitial nephritis that occur in SSc are usually associated with overlap disease of SSc with vasculitis and/or systemic lupus erythematosus or drug reactions rather than pure SSc.251–253 SRC is characterized by malignant hypertension, microangiopathic hemolysis, microthrombosis, thrombocytopenia, vasospasm, and progressive renal failure that can be provoked by corticosteroids, cocaine, cyclosporine, and tacrolimus.251,254,255 Pathologically, SRC is characterized by rather bland or subtle findings, but may demonstrate the typical “onion bulb” histopathology, hyperplasia of the juxtaglomerular apparatus, membranoproliferation, renovascular endothelial injury, intimal proliferation, thrombotic angiopathy, fibrin microthrombi, hemolysis, mesangiolysis, narrowing of renal arterioles, vasospasm, vascular occlusion, ischemia, necrosis, vascular remodeling, and eventual fibrosis with associated with hyperreninemia and accelerated hypertension.251,256

Diagnostic criteria for SRC are as follows: 1) systolic blood pressure (SBP) >140mm Hg, 2) diastolic blood pressure (DBP) >90 mm Hg, 3) increase in baseline SBP ≥ 30 mm Hg, 4) rise in DBP ≥20 mm Hg, 5) increase in serum creatinine by ≥50% over baseline or serum creatinine >120% of upper limit of normal for the local laboratory, 6) proteinuria ≥2+ by dipstick and confirmed by spot urine protein:creatinine ratio ≥ upper limit of normal, 7) hematuria ≥2+ on dip stick or ≥10 red blood cells/high power field, 8) platelet count <100,000/mm3, 9) hemolysis (evidenced by schistocytes, RBC fragments on peripheral blood smear and elevated reticulocyte count), and in certain studies, 10) hypertensive encephalopathy.255,257–259 Prevention of SRC is based on avoiding the use of high-dose corticosteroids and vasoconstrictants in SSc. Therapy of SRC has recently been reviewed and is focused on the early diagnosis of SRC, prompt use of angiotensin-converting enzyme inhibitors, dialysis, plasma exchange, and other supportive measures including eculizumab or endothelin-1 blockage in completely resistant cases in anticipation that the renal function will recover.133,251,260,261

Risk factors for SRC (Table 6) include diffuse cutaneous involvement, rapidly progressive skin thickening, disease duration <4 years, anti-RNA polymerase III antibodies, antiphospholipid antibodies, autoantibodies to methionine sulfoxide reductase A, absence of anti-centromere antibodies, new cardiac events (pericardial effusion, congestive heart failure, arrhythmias), anemia due to intravascular hemolysis, the HLA-DRB types 1*0407 and 1*1304, and prior drug use, particularly corticosteroids and vasoconstrictants.258,259,261

Table 6 Biomarkers for Predisposition to Renal Disease in SSc (RD-SSc)

Biomarkers of SRC (Table 7) include hypertension, elevated uric acid, decreased renal function, thrombocytopenia, hemolytic anemia, and elevated serum-soluble CD147 and CD163, renin, mannose-binding lectin (MBL), endothelin-1, soluble vascular adhesion molecules, E-selectin, lipocalin-2, angiogenin, apelin, chemerin, complement components, and NT-proBNP levels.251,254,261–264 Serum soluble CD147 (sCD147), an ECM metalloproteinase inhibitor and CD163 (sCD163), cysteine-rich scavenger receptor, have been shown to be elevated in patients with SRC.202,264 Similarly, increased endothelin-1 levels and endothelin receptor carriage have been associated with SRC.265–267 Similarly, soluble vascular adhesion molecules (VCAM-1) and soluble E-selectin have been associated with SRC.258,268 NT-proBNP is a useful biomarker for SRC and predicts the need for dialysis and renal outcome.266,269 CXCL10 is an IFN-inducible chemokine and potent chemoattractant for Th1 cells and is found to be elevated in patients with SRC.8 IL17B is increased specifically in SSc with renal abnormalities compared to those without.106

Table 7 Biomarkers for Active Renal Disease and SRC in SSc

Anti-fibrillarin antibodies, anti-RNA polymerase III antibodies, and speckle pattern ANA have been most closely associated with the development of SRD; however, in certain populations anti-topoisomerase antibodies have also been associated with high incidences of SRC.251,258,267,270–272 Antiphospholipid antibodies, in particular, IgG antiphospholipid antibodies, are a significant risk factor for renal disease in SSc versus antibody negative patients.273 Autoantibodies to methionine sulfoxide reductase A, an important enzyme in the antioxidant pathways, have been associated with the development of renal and cardiac disease in SSc.274

Renin, although increased in SRC and central to the pathology and treatment of SRC, is not predictive of SRC, as it is often elevated in SSc patients without SRC.251,275 Elevated serum uric acid, a purine metabolite, can be associated with inflammation, endothelial dysfunction, and renal dysfunction.276 Gigante et al have demonstrated in SSc that uric acid is significantly associated with serum creatinine, renal artery resistivity and decreases in glomerulofiltration rate (GFR), as well as multivascular damage in SSc.276 Similarly, disordered levels of pro-angiogenic molecules and angiogenesis inhibitors have been associated with the progression of renal microvascular damage, defective vascular repair and fibrosis in SSC. Specifically, levels of endostatin, an inhibitor of angiogenesis, have been found to be elevated in renal disease in SSc, and are associated with decreased GFR, increased renal artery resistivity, and progression of peripheral microvascular disease.204,277 High levels of both angiogenin and lipocalin-2 are associated with decreased GFR and may be involved in pathogenesis of SRC.265,278,279 Apelin and chemerin are adipokines that bind to receptors on endothelial cells and are elevated in SRC.280–283 The complement system also appears to be involved in SRC, with higher levels of C4d and lower levels of C3bBbP and soluble terminal complement complex (sTCC) in SSc patients with SRC versus without SRC.198 Mannose-binding lectin (MBL) is also involved in the complement system, and MBL levels are substantially increased in SRC compared to SSc without SRC.263 In the urine, monocyte chemoattractant protein 1 (MCP1) and soluble adhesion molecule vascular cell adhesion molecule 1 (VCAM-1) are elevated in SSc patients with renal disease compared to those without renal disease.284

As summarized in Table 6 and Table 7, there are multiple biomarkers that associated with the risk for SRC in SSc or delineate the activity of SRC, and many of these biomarkers are intimately involved in both the pathogenesis and characteristics of the disorder.

Conclusion

The field of biomarkers in SSc continues to expand in scope and complexity. The sheer number of molecules, pathways, and receptors involved in the pathology of SSc reflects the many complexities and nuances of the disease. Tyrosine kinases, interferon-1 signaling, IL-6 signaling, endogenous thrombin, peroxisome proliferator-activated receptors (PPARs), lysophosphatidic acid and receptors, and amino acid metabolites have all provided new biomarkers and the potential for new therapeutic agents. Because multiple arms of the immune system and healing mechanisms are activated in SSc-ILD, and there are many candidate biomarkers and potential therapeutic targets, with the trend being the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy. Biomarkers are likely to be of increasing importance for research as well as for the diagnosis and therapeutic approaches to SSc and associated disease manifestations.

Data Accessibility

Data can be obtained from the corresponding author on a reasonable request.

Acknowledgments

The authors would like to thank the Reviewers for their insight and advice on how to improve the manuscript, to Dr. Matthew K. McElwee, MD for assistance with literature searches and Ms. Jackie Cremar for assistance in the preparation of this manuscript.

Funding

There was no internal or external support for this study.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Khanna D, Tashkin DP, Denton CP, Renzoni EA, Desai SR, Varga J. Etiology, Risk Factors, and Biomarkers in Systemic Sclerosis with Interstitial Lung Disease. Am J Respir Crit Care Med. 2020;201(6):650–660. doi:10.1164/rccm.201903-0563CI

2. Kang EH, Ha YJ, Lee YJ. Autoantibody Biomarkers in Rheumatic Diseases. Int J Mol Sci. 2020;21(4):1382. doi:10.3390/ijms21041382

3. Ligon C, Hummers LK. Biomarkers in Scleroderma: progressing from Association to Clinical Utility. Curr Rheumatol Rep. 2016;18(3):17. doi:10.1007/s11926-016-0565-0

4. Graßhoff H, Fourlakis K, Comdühr S, Riemekasten G. Autoantibodies as Biomarker and Therapeutic Target in Systemic Sclerosis. Biomedicines. 2022;10(9):2150. doi:10.3390/biomedicines10092150

5. Chairta PP, Nicolaou P, Christodoulou K. Enriched in silico analysis of MS-based extracted candidate proteomic biomarkers highlights pathogenic pathways in systemic sclerosis. Sci Rep. 2023;13(1):1934. doi:10.1038/s41598-023-29054-5

6. Abignano G, Del Galdo F. Biomarkers as an opportunity to stratify for outcome in systemic sclerosis. Eur J Rheumatol. 2020;7(Suppl 3):S193–S202.

7. Bălănescu P, Bălănescu A, Bălănescu E, Băicuş C. Candidate proteomic biomarkers in systemic sclerosis discovered using mass-spectrometry: an update of a systematic review (2014-2020). Rom J Intern Med. 2021;59(2):101–111. doi:10.2478/rjim-2020-0037

8. Utsunomiya A, Oyama N, Hasegawa M. Potential Biomarkers in Systemic Sclerosis: a Literature Review and Update. J Clin Med. 2020;9(11):3388. doi:10.3390/jcm9113388

9. Wermuth PJ, Piera-Velazquez S, Jimenez SA. Identification of novel systemic sclerosis biomarkers employing aptamer proteomic analysis. Rheumatology. 2018;57(10):1698–1706. doi:10.1093/rheumatology/kex404

10. Bălănescu P, Bălănescu E, Băicuș C, Bălănescu A. Circulatory cytokeratin 17, marginal zone B1 protein and leucine-rich α2-glycoprotein-1 as biomarkers for disease severity and fibrosis in systemic sclerosis patients. Biochem Med (Zagreb). 2022;32(3):030707. doi:10.11613/BM.2022.030707

11. Gigante A, Navarini L, Margiotta D, et al. Angiogenic and angiostatic factors in renal scleroderma-associated vasculopathy. Microvasc Res. 2017;114:41–45. doi:10.1016/j.mvr.2017.06.003

12. Mostmans Y, Cutolo M, Giddelo C, et al. The role of endothelial cells in the vasculopathy of systemic sclerosis: a systematic review. Autoimmun Rev. 2017;16(8):774–786. doi:10.1016/j.autrev.2017.05.024

13. Zhang Y, Zhu M, Xie L, Zhang H, Deng T. Identification and validation of key immune-related genes with promising diagnostic and predictive value in systemic sclerosis. Life Sci. 2023;312:121238. doi:10.1016/j.lfs.2022.121238

14. Kania G, Rudnik M, Distler O. Involvement of the myeloid cell compartment in fibrogenesis and systemic sclerosis. Nat Rev Rheumatol. 2019;15(5):288–302. doi:10.1038/s41584-019-0212-z

15. Denton CP, Khanna D. Systemic sclerosis. Lancet. 2017;390(10103):1685–1699. doi:10.1016/S0140-6736(17)30933-9

16. Frantz C, Avouac J, Distler O, et al. Impaired quality of life in systemic sclerosis and patient perception of the disease: a large international survey. Semin Arthritis Rheum. 2016;46(1):115–123. doi:10.1016/j.semarthrit.2016.02.005

17. D’Oria M, Gandin I, Riccardo P, et al. Correlation between Microvascular Damage and Internal Organ Involvement in Scleroderma: focus on Lung Damage and Endothelial Dysfunction. Diagnostics. 2022;13(1):55. doi:10.3390/diagnostics13010055

18. van den Hoogen F, Khanna D, Fransen J, et al. 2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against Rheumatism collaborative initiative. Arthritis Rheum. 2013;65(11):2737–2747. doi:10.1002/art.38098

19. 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:10.1002/art.38390

20. Khanna D, Lescoat A, Roofeh D, et al. Systemic Sclerosis-Associated Interstitial Lung Disease: how to Incorporate Two Food and Drug Administration-Approved Therapies in Clinical Practice. Arthritis Rheumatol. 2022;74(1):13–27. doi:10.1002/art.41933

21. Cavazzana I, Vojinovic T, Airo’ P, et al. Systemic Sclerosis-Specific Antibodies: novel and Classical Biomarkers. Clin Rev Allergy Immunol. 2022;64(3):412–430. doi:10.1007/s12016-022-08946-w

22. Soulaidopoulos S, Triantafyllidou E, Garyfallos A, et al. The role of nailfold capillaroscopy in the assessment of internal organ involvement in systemic sclerosis: a critical review. Autoimmun Rev. 2017;16(8):787–795. doi:10.1016/j.autrev.2017.05.019

23. Vondenberg JA, Muruganandam M, Nunez SE, Emil NS, Sibbitt WL. Increased malignancies in systemic sclerosis. Int J Rheum Dis. 2022;25(1):90–92. doi:10.1111/1756-185X.14244

24. Varjú C, Pauling JD, Saketkoo LA. Multi-Organ System Screening, Care, and Patient Support in Systemic Sclerosis. Rheum Dis Clin North Am. 2023;49(2):211–248. doi:10.1016/j.rdc.2023.01.002

25. Affandi AJ, Radstake TR, Marut W. Update on biomarkers in systemic sclerosis: tools for diagnosis and treatment. Semin Immunopathol. 2015;37(5):475–487. doi:10.1007/s00281-015-0506-4

26. Mihai C, Tervaert JW. Anti-endothelial cell antibodies in systemic sclerosis. Ann Rheum Dis. 2010;69(2):319–324. doi:10.1136/ard.2008.102400

27. Fineschi S, Goffin L, Rezzonico R, et al. Antifibroblast antibodies in systemic sclerosis induce fibroblasts to produce profibrotic chemokines, with partial exploitation of toll-like receptor 4. Arthritis Rheum. 2008;58(12):3913–3923. doi:10.1002/art.24049

28. Moroncini G, Svegliati Baroni S, Gabrielli A. Agonistic antibodies in systemic sclerosis. Immunol Lett. 2018;195:83–87. doi:10.1016/j.imlet.2017.10.007

29. Sato S, Fujimoto M, Hasegawa M, et al. Serum soluble CTLA-4 levels are increased in diffuse cutaneous systemic sclerosis. Rheumatology. 2004;43(10):1261–1266. doi:10.1093/rheumatology/keh303

30. Tan FK, Arnett FC, Antohi S, et al. Autoantibodies to the extracellular matrix microfibrillar protein, fibrillin-1, in patients with scleroderma and other connective tissue diseases. J Immunol. 1999;163(2):1066–1072. doi:10.4049/jimmunol.163.2.1066

31. Fritzler MJ, Bentow C, Beretta L, et al. Anti-U11/U12 Antibodies as a Rare but Important Biomarker in Patients with Systemic Sclerosis: a Narrative Review. Diagnostics. 2023;13(7):1257. doi:10.3390/diagnostics13071257

32. Nihtyanova SI, Denton CP. Autoantibodies as predictive tools in systemic sclerosis. Nat Rev Rheumatol. 2010;6(2):112–116. doi:10.1038/nrrheum.2009.238

33. Clark KEN, Campochiaro C, Host LV, et al. Combinations of scleroderma hallmark autoantibodies associate with distinct clinical phenotypes. Sci Rep. 2022;12(1):11212. doi:10.1038/s41598-022-15062-4

34. Chepy A, Bourel L, Koether V, Launay D, Dubucquoi S, Sobanski V. Can Antinuclear Antibodies Have a Pathogenic Role in Systemic Sclerosis? Front Immunol. 2022;13:930970. doi:10.3389/fimmu.2022.930970

35. Tsai CY, Hsieh SC, Wu TH, et al. Pathogenic Roles of Autoantibodies and Aberrant Epigenetic Regulation of Immune and Connective Tissue Cells in the Tissue Fibrosis of Patients with Systemic Sclerosis. Int J Mol Sci. 2020;21(9):3069. doi:10.3390/ijms21093069

36. Hoa S, Lazizi S, Baron M, Wang M, Fritzler MJ, Hudson M; Canadian Scleroderma Research Group. Association between autoantibodies in systemic sclerosis and cancer in a national registry. Rheumatology. 2022;61(7):2905–2914. doi:10.1093/rheumatology/keab735

37. Liem SIE, Neppelenbroek S, Fehres CM, et al. Autoreactive B cell responses targeting nuclear antigens in systemic sclerosis: implications for disease pathogenesis. Semin Arthritis Rheum. 2023;58:152136. doi:10.1016/j.semarthrit.2022.152136

38. Loisel S, Lansiaux P, Rossille D, et al. Regulatory B Cells Contribute to the Clinical Response After Bone Marrow-Derived Mesenchymal Stromal Cell Infusion in Patients With Systemic Sclerosis. Stem Cells Transl Med. 2023:szad010. doi:10.1093/stcltm/szad010

39. Arnett FC. HLA and autoimmunity in scleroderma (systemic sclerosis). Int Rev Immunol. 1995;12(2–4):107–128. doi:10.3109/08830189509056707

40. Kuwana M, Okano Y, Kaburaki J, Medsger TA, Wright TM. Autoantibodies to RNA polymerases recognize multiple subunits and demonstrate cross-reactivity with RNA polymerase complexes. Arthritis Rheum. 1999;42(2):275–284. doi:10.1002/1529-0131(199902)42:2<275::AID-ANR9>3.0.CO;2-P

41. Kuwana M, Pandey JP, Silver RM, et al. HLA class II alleles in systemic sclerosis patients with anti-RNA polymerase I/III antibody: associations with subunit reactivities. J Rheumatol. 2003;30(11):2392–2397.

42. Hanson AL, Sahhar J, Ngian GS, et al. Contribution of HLA and KIR Alleles to Systemic Sclerosis Susceptibility and Immunological and Clinical Disease Subtypes. Front Genet. 2022;13:913196. doi:10.3389/fgene.2022.913196

43. Acosta-Herrera M, Kerick M, Lopéz-Isac E, et al. Comprehensive analysis of the major histocompatibility complex in systemic sclerosis identifies differential HLA associations by clinical and serological subtypes. Ann Rheum Dis. 2021;80(8):1040–1047. doi:10.1136/annrheumdis-2021-219884

44. Caselli E, Soffritti I, D’Accolti M, et al. HHV-6A Infection and Systemic Sclerosis: clues of a Possible Association. Microorganisms. 2019;8(1):39. doi:10.3390/microorganisms8010039

45. Gourh P, Safran SA, Alexander T, et al. HLA and autoantibodies define scleroderma subtypes and risk in African and European Americans and suggest a role for molecular mimicry. Proc Natl Acad Sci U S A. 2020;117(1):552–562. doi:10.1073/pnas.1906593116

46. Kerick M, Acosta-Herrera M, Simeón-Aznar CP, et al. Complement component C4 structural variation and quantitative traits contribute to sex-biased vulnerability in systemic sclerosis. NPJ Genom Med. 2022;7(1):57. doi:10.1038/s41525-022-00327-8

47. O’Reilly S. Toll-like receptor triggering in systemic sclerosis: time to target. Rheumatology. 2023;62(SI):SI12–SI19. doi:10.1093/rheumatology/keac421

48. Bale S, Verma P, Varga J, Bhattacharyya S. Extracellular Matrix-Derived Damage-Associated Molecular Patterns: implications in Systemic Sclerosis and Fibrosis. J Invest Dermatol. 2023;143(10):1877–1885. doi:10.1016/j.jid.2023.04.030

49. Choreño-Parra JA, Cervantes-Rosete D, Jiménez-álvarez LA, et al. Dendritic cells drive profibrotic inflammation and aberrant T cell polarization in systemic sclerosis. Rheumatology. 2023;62(4):1687–1698. doi:10.1093/rheumatology/keac489

50. Liu X, Mayes MD, Tan FK, et al. Correlation of interferon-inducible chemokine plasma levels with disease severity in systemic sclerosis. Arthritis Rheum. 2013;65(1):226–235. doi:10.1002/art.37742

51. Wang W, Bhattacharyya S, Marangoni RG, et al. The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib. J Scleroderma Relat Disord. 2020;5(1):40–50. doi:10.1177/2397198319865367

52. Jiang S, Yang H, Li M. Emerging Roles of Lysophosphatidic Acid in Macrophages and Inflammatory Diseases. Int J Mol Sci. 2023;24(15):12524. doi:10.3390/ijms241512524

53. Akter T, Silver RM, Bogatkevich GS. Recent advances in understanding the pathogenesis of scleroderma-interstitial lung disease. Curr Rheumatol Rep. 2014;16(4):411. doi:10.1007/s11926-014-0411-1

54. Christmann RB, Sampaio-Barros P, Stifano G, et al. Association of Interferon- and transforming growth factor β-regulated genes and macrophage activation with systemic sclerosis-related progressive lung fibrosis. Arthritis Rheumatol. 2014;66(3):714–725. doi:10.1002/art.38288

55. Bellocchi C, Assassi S, Lyons M, et al. Proteomic aptamer analysis reveals serum markers that characterize preclinical systemic sclerosis (SSc) patients at risk for progression toward definite SSc. Arthritis Res Ther. 2023;25(1):15. doi:10.1186/s13075-023-02989-w

56. Kudo A, Kii I. Periostin function in communication with extracellular matrices. J Cell Commun Signal. 2018;12(1):301–308. doi:10.1007/s12079-017-0422-6

57. Kii I. Periostin Functions as a Scaffold for Assembly of Extracellular Proteins. Adv Exp Med Biol. 2019;1132:23–32.

58. El-Adili F, Lui JK, Najem M, et al. Periostin overexpression in scleroderma cardiac tissue and its utility as a marker for disease complications. Arthritis Res Ther. 2022;24(1):251. doi:10.1186/s13075-022-02943-2

59. Distler O, Distler J, Kowal-Bielecka O, Gay RE, Müller-Ladner U, Gay S. Chemokines and chemokine receptors in the pathogenesis of systemic sclerosis. Mod Rheumatol. 2002;12(2):107–112. doi:10.3109/s101650200019

60. Bayati P, Poormoghim H, Mojtabavi N. Aberrant expression of miR-138 as a novel diagnostic biomarker in systemic sclerosis. Biomark Insights. 2022;17:11772719221135442. doi:10.1177/11772719221135442

61. Bayati P, Kalantari M, Assarehzadegan MA, Poormoghim H, Mojtabavi N. MiR-27a as a diagnostic biomarker and potential therapeutic target in systemic sclerosis. Sci Rep. 2022;12(1):18932. doi:10.1038/s41598-022-23723-7

62. Wajda A, Walczyk M, Dudek E, et al. Serum microRNAs in Systemic Sclerosis, Associations with Digital Vasculopathy and Lung Involvement. Int J Mol Sci. 2022;23(18):10731. doi:10.3390/ijms231810731

63. Iannazzo F, Pellicano C, Colalillo A, et al. Interleukin-33 and soluble suppression of tumorigenicity 2 in scleroderma cardiac involvement. Clin Exp Med. 2022;23(3):897–903. doi:10.1007/s10238-022-00864-7

64. Colalillo A, Pellicano C, Rosato E. Serum-soluble ST2 and systemic sclerosis arthropathy. Clin Rheumatol. 2023;42(3):871–877. doi:10.1007/s10067-022-06367-w

65. Michalska-Jakubus M, Cutolo M, Smith V, Krasowska D. Imbalanced serum levels of Ang1, Ang2 and VEGF in systemic sclerosis: integrated effects on microvascular reactivity. Microvasc Res. 2019;125:103881. doi:10.1016/j.mvr.2019.103881

66. Kakkar V, Assassi S, Allanore Y, et al. Type 1 interferon activation in systemic sclerosis: a biomarker, a target or the culprit. Curr Opin Rheumatol. 2022;34(6):357–364. doi:10.1097/BOR.0000000000000907

67. Ciechomska M, Skalska U. Targeting interferons as a strategy for systemic sclerosis treatment. Immunol Lett. 2018;195:45–54. doi:10.1016/j.imlet.2017.10.011

68. Londe AC, Fernandez-Ruiz R, Rogério Julio P, Appenzeller S, Niewold TB. Type I Interferons in Autoimmunity: implications in Clinical Phenotypes and Treatment Response. J Rheumatol. 2023;jrheum.2022–0827.

69. Assassi S, Mayes MD, Arnett FC, et al. Systemic sclerosis and lupus: points in an interferon-mediated continuum. Arthritis Rheum. 2010;62(2):589–598. doi:10.1002/art.27224

70. Chaudhary V, Ah Kioon MD, Hwang SM, et al. Chronic activation of pDCs in autoimmunity is linked to dysregulated ER stress and metabolic responses. J Exp Med. 2022;219(11):e20221085. doi:10.1084/jem.20221085

71. Höppner J, Casteleyn V, Biesen R, et al. SIGLEC-1 in Systemic Sclerosis: a Useful Biomarker for Differential Diagnosis. Pharmaceuticals. 2022;15(10):1198. doi:10.3390/ph15101198

72. Wu M, Assassi S. Dysregulation of Type 1 Interferon Signaling in Systemic Sclerosis: a Promising Therapeutic Target? Curr Treatm Opt Rheumatol. 2021;7(4):349–360. doi:10.1007/s40674-021-00188-9

73. Guo X, Higgs BW, Bay-Jensen AC, et al. Suppression of T cell activation and collagen accumulation by an anti-IFNAR1 mAb, anifrolumab, in adult patients with systemic sclerosis. J Invest Dermatol. 2015;135(10):2402–2409. doi:10.1038/jid.2015.188

74. Mendoza FA, Piera-Velazquez S, Jimenez SA. Tyrosine kinases in the pathogenesis of tissue fibrosis in systemic sclerosis and potential therapeutic role of their inhibition. Transl Res. 2021;231:139–158. doi:10.1016/j.trsl.2021.01.001

75. Wollin L, Maillet I, Quesniaux V, et al. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther. 2014;349(2):209–220. doi:10.1124/jpet.113.208223

76. Wollin L, Wex E, Pautsch A, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45(5):1434–1445. doi:10.1183/09031936.00174914

77. Sato S, Shinohara S, Hayashi S, et al. Anti-fibrotic efficacy of nintedanib in pulmonary fibrosis via the inhibition of fibrocyte activity. Respir Res. 2017;18(1):172. doi:10.1186/s12931-017-0654-2

78. Avouac J, Fürnrohr BG, Tomcik M, et al. Inactivation of the transcription factor STAT-4 prevents inflammation-driven fibrosis in animal models of systemic sclerosis. Arthritis Rheum. 2011;63(3):800–809. doi:10.1002/art.30171

79. Krishna Swaroop A, Krishnan Namboori PK, Esakkimuthukumar M, et al. Leveraging decagonal in-silico strategies for uncovering IL-6 inhibitors with precision. Comput Biol Med. 2023;163:107231. doi:10.1016/j.compbiomed.2023.107231

80. Sheng XR, Gao X, Schiffman C, et al. Biomarkers of fibrosis, inflammation, and extracellular matrix in the Phase 3 trial of tocilizumab in systemic sclerosis. Clin Immunol. 2023;254:109695. doi:10.1016/j.clim.2023.109695

81. Khanna D, Lin CJF, Furst DE, et al. Long-Term Safety and Efficacy of Tocilizumab in Early Systemic Sclerosis-Interstitial Lung Disease: open-Label Extension of a Phase 3 Randomized Controlled Trial. Am J Respir Crit Care Med. 2022;205(6):674–684. doi:10.1164/rccm.202103-0714OC

82. Junfei Z, Meihua G, Shuai Z, et al. Retrospective comparative study of the efficacy of JAK inhibitor (tofacitinib) in the treatment of systemic sclerosis-associated interstitial lung disease. Clin Rheumatol. 2023;42(10):2823–2832. doi:10.1007/s10067-023-06660-2

83. Moriana C, Moulinet T, Jaussaud R, Decker P. JAK inhibitors and systemic sclerosis: a systematic review of the literature. Autoimmun Rev. 2022;21(10):103168. doi:10.1016/j.autrev.2022.103168

84. Kuszmiersz P, Pacholczak-Madej R, Siwiec A, et al. Thrombin generation potential is enhanced in systemic sclerosis: impact of selected endothelial biomarkers. Clin Exp Rheumatol. 2021;39 Suppl 131(4):13–19. doi:10.55563/clinexprheumatol/d03dnc

85. Colic J, Pruner I, Damjanov N, et al. Impaired Fibrinolysis Is Linked With Digital Vasculopathy and Onset of New Digital Ulcers in Systemic Sclerosis. J Rheumatol. 2022;49(6):598–606. doi:10.3899/jrheum.210931

86. Silver RM, Wilson DA, Akter T, et al. Safety and Tolerability of Thrombin Inhibition in Scleroderma-Associated Interstitial Lung Disease. ACR Open Rheumatol. 2019;1(7):403–411. doi:10.1002/acr2.11049

87. Liu Y, Wang J, Luo S, Zhan Y, Lu Q. The roles of PPARγ and its agonists in autoimmune diseases: a comprehensive review. J Autoimmun. 2020;113:102510. doi:10.1016/j.jaut.2020.102510

88. Leask A. Possible strategies for anti-fibrotic drug intervention in scleroderma. J Cell Commun Signal. 2011;5(2):125–129. doi:10.1007/s12079-011-0122-6

89. Ghosh AK, Bhattacharyya S, Wei J, et al. Peroxisome proliferator-activated receptor-gamma abrogates Smad-dependent collagen stimulation by targeting the p300 transcriptional coactivator. FASEB J. 2009;23(9):2968–2977. doi:10.1096/fj.08-128736

90. Wei J, Ghosh AK, Sargent JL, et al. PPARγ downregulation by TGFß in fibroblast and impaired expression and function in systemic sclerosis: a novel mechanism for progressive fibrogenesis. PLoS One. 2010;5(11):e13778. doi:10.1371/journal.pone.0013778

91. Lakota K, Wei J, Carns M, et al. Levels of adiponectin, a marker for PPAR-gamma activity, correlate with skin fibrosis in systemic sclerosis: potential utility as biomarker? Arthritis Res Ther. 2012;14(3):R102. doi:10.1186/ar3827

92. Żółkiewicz J, Stochmal A, Zaremba M, et al. Circulating peroxisome proliferator-activated receptor γ is elevated in systemic sclerosis. Postepy Dermatol Alergol. 2020;37(6):921–926. doi:10.5114/ada.2019.84746

93. Derrett-Smith E, Clark KEN, Shiwen X, et al. The pan-PPAR agonist lanifibranor reduces development of lung fibrosis and attenuates cardiorespiratory manifestations in a transgenic mouse model of systemic sclerosis. Arthritis Res Ther. 2021;23(1):234. doi:10.1186/s13075-021-02592-x

94. Tigyi G. Aiming drug discovery at lysophosphatidic acid targets. Br J Pharmacol. 2010;161(2):241–270. doi:10.1111/j.1476-5381.2010.00815.x

95. Tokumura A, Carbone LD, Yoshioka Y, et al. Elevated serum levels of arachidonoyl-lysophosphatidic acid and sphingosine 1-phosphate in systemic sclerosis. Int J Med Sci. 2009;6(4):168–176. doi:10.7150/ijms.6.168

96. Allanore Y, Distler O, Jagerschmidt A, et al. Lysophosphatidic Acid Receptor 1 Antagonist SAR100842 for Patients With Diffuse Cutaneous Systemic Sclerosis: a Double-Blind, Randomized, Eight-Week Placebo-Controlled Study Followed by a Sixteen-Week Open-Label Extension Study. Arthritis Rheumatol. 2018;70(10):1634–1643. doi:10.1002/art.40547

97. Feng D, Gerarduzzi C. Emerging Roles of Matricellular Proteins in Systemic Sclerosis. Int J Mol Sci. 2020;21(13):4776. doi:10.3390/ijms21134776

98. Leong E, Bezuhly M, Marshall JS. Distinct Metalloproteinase Expression and Functions in Systemic Sclerosis and Fibrosis: what We Know and the Potential for Intervention. Front Physiol. 2021;12:727451. doi:10.3389/fphys.2021.727451

99. Peng WJ, Yan JW, Wan YN, et al. Matrix metalloproteinases: a review of their structure and role in systemic sclerosis. J Clin Immunol. 2012;32(6):1409–1414. doi:10.1007/s10875-012-9735-7

100. Elias GJ, Ioannis M, Theodora P, et al. Circulating tissue inhibitor of matrix metalloproteinase-4 (TIMP-4) in systemic sclerosis patients with elevated pulmonary arterial pressure. Mediators Inflamm. 2008;2008:164134. doi:10.1155/2008/164134

101. Mazzotta C, Romano E, Bruni C, et al. Plexin-D1/Semaphorin 3E pathway may contribute to dysregulation of vascular tone control and defective angiogenesis in systemic sclerosis. Arthritis Res Ther. 2015;17(1):221. doi:10.1186/s13075-015-0749-4

102. Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. Circulating Neurovascular Guidance Molecules and Their Relationship with Peripheral Microvascular Impairment in Systemic Sclerosis. Life. 2022;12(7):1056. doi:10.3390/life12071056

103. Nakamura K, Asano Y, Taniguchi T, et al. Serum levels of interleukin-18-binding protein isoform a: clinical association with inflammation and pulmonary hypertension in systemic sclerosis. J Dermatol. 2016;43(8):912–918. doi:10.1111/1346-8138.13252

104. Fioretto BS, Rosa I, Matucci-Cerinic M, Romano E, Manetti M. Current Trends in Vascular Biomarkers for Systemic Sclerosis: a Narrative Review. Int J Mol Sci. 2023;24(4):4097. doi:10.3390/ijms24044097

105. Kardum Ž, Milas-Ahić J, Šahinović I, Masle AM, Uršić D, Kos M. Serum levels of interleukin 17 and 22 in patients with systemic sclerosis: a single-center cross-sectional study. Rheumatol Int. 2023;43(2):345–354. doi:10.1007/s00296-022-05250-w

106. Robak E, Gerlicz-Kowalczuk Z, Dziankowska-Bartkowiak B, Wozniacka A, Bogaczewicz J. Serum concentrations of IL-17A, IL-17B, IL-17E and IL-17F in patients with systemic sclerosis. Arch Med Sci. 2019;15(3):706–712. doi:10.5114/aoms.2019.84738

107. Ikawa T, Miyagawa T, Fukui Y, et al. Association of serum CCL20 levels with pulmonary vascular involvement and primary biliary cholangitis in patients with systemic sclerosis. Int J Rheum Dis. 2021;24(5):711–718. doi:10.1111/1756-185X.14103

108. Didriksen H, Molberg Ø, Mehta A, et al. Target organ expression and biomarker characterization of chemokine CCL21 in systemic sclerosis associated pulmonary arterial hypertension. Front Immunol. 2022;13:991743. doi:10.3389/fimmu.2022.991743

109. Yanaba K, Yoshizaki A, Muroi E, et al. Serum CCL23 levels are increased in patients with systemic sclerosis. Arch Dermatol Res. 2011;303(1):29–34. doi:10.1007/s00403-010-1078-8

110. Cossu M, van Bon L, Preti C, Rossato M, Beretta L, Radstake TRDJ. Earliest Phase of Systemic Sclerosis Typified by Increased Levels of Inflammatory Proteins in the Serum. Arthritis Rheumatol. 2017;69(12):2359–2369. doi:10.1002/art.40243

111. Hasegawa M, Sato S, Echigo T, Hamaguchi Y, Yasui M, Takehara K. Up regulated expression of fractalkine/CX3CL1 and CX3CR1 in patients with systemic sclerosis. Ann Rheum Dis. 2005;64(1):21–28. doi:10.1136/ard.2003.018705

112. Taniguchi T, Asano Y, Akamata K, et al. Serum levels of galectin-3: possible association with fibrosis, aberrant angiogenesis, and immune activation in patients with systemic sclerosis. J Rheumatol. 2012;39(3):539–544. doi:10.3899/jrheum.110755

113. Pellicano C, Romaggioli L, Miglionico M, et al. Maresin1 is a predictive marker of new digital ulcers in systemic sclerosis patients. Microvasc Res. 2022;142:104366. doi:10.1016/j.mvr.2022.104366

114. Arakawa H, Jinnin M, Muchemwa FC, et al. Adiponectin expression is decreased in the involved skin and sera of diffuse cutaneous scleroderma patients. Exp Dermatol. 2011;20(9):764–766. doi:10.1111/j.1600-0625.2011.01310.x

115. Stratton RJ, Pompon L, Coghlan JG, et al. Soluble thrombomodulin concentration is raised in scleroderma associated pulmonary hypertension. Ann Rheum Dis. 2000;59(2):132–134. doi:10.1136/ard.59.2.132

116. Hassan WA, Baraka EA, Elnady BM, et al. Serum Soluble CD163 and its association with various disease parameters in patients with systemic sclerosis. Eur J Rheumatol. 2016;3(3):95–100. doi:10.5152/eurjrheum.2016.15088

117. Nowaczyk J, Blicharz L, Zawistowski M, et al. The Clinical Significance of Salusins in Systemic Sclerosis-A Cross-Sectional Study. Diagnostics. 2023;13(5):848. doi:10.3390/diagnostics13050848

118. Servaas NH, Hiddingh S, Chouri E, et al. Nuclear Receptor Subfamily 4A Signaling as a Key Disease Pathway of CD1c+ Dendritic Cell Dysregulation in Systemic Sclerosis. Arthritis Rheumatol. 2023;75(2):279–292. doi:10.1002/art.42319

119. Morales-González V, Galeano-Sánchez D, Covaleda-Vargas JE, et al. Metabolic fingerprinting of systemic sclerosis: a systematic review. Front Mol Biosci. 2023;10:1215039. doi:10.3389/fmolb.2023.1215039

120. Perelas A, Silver RM, Arrossi AV, Highland KB. Systemic sclerosis-associated interstitial lung disease. Lancet Respir Med. 2020;8(3):304–320. doi:10.1016/S2213-2600(19)30480-1

121. Hoffmann-Vold AM, Allanore Y, Alves M, et al. Progressive interstitial lung disease in patients with systemic sclerosis-associated interstitial lung disease in the EUSTAR database. Ann Rheum Dis. 2021;80(2):219–227. doi:10.1136/annrheumdis-2020-217455

122. Tyndall AJ, Bannert B, Vonk M, et al. Causes and risk factors for death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis. 2010;69(10):1809–1815. doi:10.1136/ard.2009.114264

123. Steen V. Predictors of end stage lung disease in systemic sclerosis. Ann Rheum Dis. 2003;62(2):97–99. doi:10.1136/ard.62.2.97

124. Wangkaew S, Euathrongchit J, Wattanawittawas P, Kasitanon N, Louthrenoo W. Incidence and predictors of interstitial lung disease (ILD) in Thai patients with early systemic sclerosis: inception cohort study. Mod Rheumatol. 2016;26(4):588–593. doi:10.3109/14397595.2015.1115455

125. Giacomelli R, Liakouli V, Berardicurti O, et al. Interstitial lung disease in systemic sclerosis: current and future treatment. Rheumatol Int. 2017;37(6):853–863. doi:10.1007/s00296-016-3636-7

126. Wells AU, Denton CP. Interstitial lung disease in connective tissue disease--mechanisms and management. Nat Rev Rheumatol. 2014;10(12):728–739. doi:10.1038/nrrheum.2014.149

127. Ahmed S, Handa R. Management of Connective Tissue Disease-related Interstitial Lung Disease. Curr Pulmonol Rep. 2022;11(3):86–98. doi:10.1007/s13665-022-00290-w

128. DeMizio DJ, Bernstein EJ. Detection and classification of systemic sclerosis-related interstitial lung disease: a review. Curr Opin Rheumatol. 2019;31(6):553–560. doi:10.1097/BOR.0000000000000660

129. Distler O, Assassi S, Cottin V, et al. Predictors of progression in systemic sclerosis patients with interstitial lung disease. Eur Respir J. 2020;55(5):1902026. doi:10.1183/13993003.02026-2019

130. Temiz Karadag D, Cakir O, San S, Yazici A, Ciftci E, Cefle A. Association of quantitative computed tomography indices with lung function and extent of pulmonary fibrosis in patients with systemic sclerosis. Clin Rheumatol. 2022;41(2):513–521. doi:10.1007/s10067-021-05918-x

131. Gasperini ML, Gigante A, Iacolare A, Pellicano C, Lucci S, Rosato E. The predictive role of lung ultrasound in progression of scleroderma interstitial lung disease. Clin Rheumatol. 2020;39(1):119–123. doi:10.1007/s10067-019-04686-z

132. Khanna D, Mittoo S, Aggarwal R, et al. Connective Tissue Disease-associated Interstitial Lung Diseases (CTD-ILD) - Report from OMERACT CTD-ILD Working Group. J Rheumatol. 2015;42(11):2168–2171. doi:10.3899/jrheum.141182

133. Kowal-Bielecka O, Fransen J, Avouac J, et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76(8):1327–1339. doi:10.1136/annrheumdis-2016-209909

134. Denton CP, De Lorenzis E, Roblin E, et al. Management of systemic sclerosis: British Society for Rheumatology guideline scope. Rheumatol Adv Pract. 2023;7(1):rkad022. doi:10.1093/rap/rkad022

135. Raghu G, MontesiSB, Silver RM, et al. Treatment of Systemic Sclerosis-associated Interstitial Lung Disease: Evidence-based Recommendations. An Official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med. In press 2023. doi:10.1164/rccm.202306-1113ST

136. de Vries-Bouwstra JK, Allanore Y, Matucci-Cerinic M, Balbir-Gurman A. Worldwide Expert Agreement on Updated Recommendations for the Treatment of Systemic Sclerosis. J Rheumatol. 2020;47(2):249–254. doi:10.3899/jrheum.181173

137. Hoffmann-Vold AM, Maher TM, Philpot EE, Ashrafzadeh A, Distler O. Assessment of recent evidence for the management of patients with systemic sclerosis-associated interstitial lung disease: a systematic review. ERJ Open Res. 2021;7(1):00235–2020. doi:10.1183/23120541.00235-2020

138. Furue M, Mitoma C, Mitoma H, et al. Pathogenesis of systemic sclerosis-current concept and emerging treatments. Immunol Res. 2017;65(4):790–797. doi:10.1007/s12026-017-8926-y

139. Petrova DT, Brandhorst G, Koch C, et al. Mycophenolic acid reverses TGF beta-induced cell motility, collagen matrix contraction and cell morphology in vitro. Cell Biochem Funct. 2015;33(7):503–508. doi:10.1002/cbf.3149

140. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 2000;47(2–3):85–118. doi:10.1016/S0162-3109(00)00188-0

141. 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:10.1136/annrheumdis-2018-213031

142. Khanna D, Padilla C, Tsoi LC, et al. Tofacitinib blocks IFN-regulated biomarker genes in skin fibroblasts and keratinocytes in a systemic sclerosis trial. JCI Insight. 2022;7(17):e159566. doi:10.1172/jci.insight.159566

143. Numajiri H, Kuzumi A, Fukasawa T, et al. B Cell Depletion Inhibits Fibrosis via Suppression of Profibrotic Macrophage Differentiation in a Mouse Model of Systemic Sclerosis. Arthritis Rheumatol. 2021;73(11):2086–2095. doi:10.1002/art.41798

144. Acharya N, Sharma SK, Mishra D, Dhooria S, Dhir V, Jain S. Efficacy and safety of pirfenidone in systemic sclerosis-related interstitial lung disease-a randomised controlled trial. Rheumatol Int. 2020;40(5):703–710. doi:10.1007/s00296-020-04565-w

145. Sullivan KM, Goldmuntz EA, Keyes-Elstein L, et al. Myeloablative Autologous Stem-Cell Transplantation for Severe Scleroderma. N Engl J Med. 2018;378(1):35–47. doi:10.1056/NEJMoa1703327

146. Bernstein EJ, Peterson ER, Sell JL, et al. Survival of adults with systemic sclerosis following lung transplantation: a nationwide cohort study. Arthritis Rheumatol. 2015;67(5):1314–1322. doi:10.1002/art.39021

147. Nihtyanova SI, Sari A, Harvey JC, et al. Using Autoantibodies and Cutaneous Subset to Develop Outcome-Based Disease Classification in Systemic Sclerosis. Arthritis Rheumatol. 2020;72(3):465–476. doi:10.1002/art.41153

148. Kuwana M, Gil-Vila A, Selva-O’Callaghan A. Role of autoantibodies in the diagnosis and prognosis of interstitial lung disease in autoimmune rheumatic disorders. Ther Adv Musculoskelet Dis. 2021;13:1759720X211032457. doi:10.1177/1759720X211032457

149. Geroldinger-Simić M, Bayati S, Pohjanen E, Sepp N, Nilsson P, Pin E. Autoantibodies against PIP4K2B and AKT3 Are Associated with Skin and Lung Fibrosis in Patients with Systemic Sclerosis. Int J Mol Sci. 2023;24(6):5629. doi:10.3390/ijms24065629

150. Jog NR, James JA. Biomarkers in connective tissue diseases. J Allergy Clin Immunol. 2017;140(6):1473–1483. doi:10.1016/j.jaci.2017.10.003

151. 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:10.1038/s41431-018-0104-8

152. Elhai M, Avouac J, Allanore Y. Circulating lung biomarkers in idiopathic lung fibrosis and interstitial lung diseases associated with connective tissue diseases: where do we stand? Semin Arthritis Rheum. 2020;50(3):480–491. doi:10.1016/j.semarthrit.2020.01.006

153. Fields A, Potel KN, Cabuhal R, Aziri B, Stewart ID, Schock BC. Mediators of systemic sclerosis-associated interstitial lung disease (SSc-ILD): systematic review and meta-analyses. Thorax. 2022:thoraxjnl-2022–219226. doi:10.1136/thorax-2022-219226

154. Bonhomme O, André B, Gester F, et al. Biomarkers in systemic sclerosis-associated interstitial lung disease: review of the literature. Rheumatology. 2019;58(9):1534–1546. doi:10.1093/rheumatology/kez230

155. Elhai M, Hoffmann-Vold AM, Avouac J, et al. Performance of Candidate Serum Biomarkers for Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol. 2019;71(6):972–982. doi:10.1002/art.40815

156. 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:10.1016/j.pharmthera.2019.05.014

157. Pietraforte I, Butera A, Gaddini L, et al. CXCL4-RNA Complexes Circulate in Systemic Sclerosis and Amplify Inflammatory/Pro-Fibrotic Responses by Myeloid Dendritic Cells. Int J Mol Sci. 2022;24(1):653. doi:10.3390/ijms24010653

158. Hoffmann-Vold AM, Fretheim H, Meier C, Maurer B. Circulating biomarkers of systemic sclerosis - interstitial lung disease. J Scleroderma Relat Disord. 2020;5(2 Suppl):41–47. doi:10.1177/2397198319894851

159. Mauer J, Chaurasia B, Goldau J, et al. Signaling by IL-6 promotes alternative activation of macrophages to limit endotoxemia and obesity-associated resistance to insulin. Nat Immunol. 2014;15(5):423–430. doi:10.1038/ni.2865

160. Sanges S, Guerrier T, Duhamel A, et al. Soluble markers of B cell activation suggest a role of B cells in the pathogenesis of systemic sclerosis-associated pulmonary arterial hypertension. Front Immunol. 2022;13:954007. doi:10.3389/fimmu.2022.954007

161. Ho KT, Reveille JD. The clinical relevance of autoantibodies in scleroderma. Arthritis Res Ther. 2003;5(2):80–93. doi:10.1186/ar628

162. Manetti M, Rosa I, Fioretto BS, Matucci-Cerinic M, Romano E. Decreased Serum Levels of SIRT1 and SIRT3 Correlate with Severity of Skin and Lung Fibrosis and Peripheral Microvasculopathy in Systemic Sclerosis. J Clin Med. 2022;11(5):1362. doi:10.3390/jcm11051362

163. 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:10.1002/art.40171

164. Omori I, Sumida H, Sugimori A, et al. Serum cold-inducible RNA-binding protein levels as a potential biomarker for systemic sclerosis-associated interstitial lung disease. Sci Rep. 2023;13(1):5017. doi:10.1038/s41598-023-32231-1

165. Jee AS, Stewart I, Youssef P, et al. A composite serum biomarker index for the diagnosis of systemic sclerosis interstitial lung disease: a multicentre, observational, cohort study. Arthritis Rheumatol. 2023;75(8):1424–1433. doi:10.1002/art.42491

166. Bauer Y, de Bernard S, Hickey P, et al. Identifying early pulmonary arterial hypertension biomarkers in systemic sclerosis: machine learning on proteomics from the DETECT cohort. Eur Respir J. 2021;57(6):2002591. doi:10.1183/13993003.02591-2020

167. Huang H, Fava A, Guhr T, et al. A methodology for exploring biomarker--phenotype associations: application to flow cytometry data and systemic sclerosis clinical manifestations. BMC Bioinform. 2015;16:293. doi:10.1186/s12859-015-0722-x

168. Yu L, Domsic RT, Saketkoo LA, et al. Assessment of the Systemic Sclerosis-Associated Raynaud’s Phenomenon Questionnaire: item Bank and Short-Form Development. Arthritis Care Res. 2022. doi:10.1002/acr.25038

169. Asano Y. The Pathogenesis of Systemic Sclerosis: an Understanding Based on a Common Pathologic Cascade across Multiple Organs and Additional Organ-Specific Pathologies. J Clin Med. 2020;9(9):2687. doi:10.3390/jcm9092687

170. Sundaram SM, Chung L. An Update on Systemic Sclerosis-Associated Pulmonary Arterial Hypertension: a Review of the Current Literature. Curr Rheumatol Rep. 2018;20(2):10. doi:10.1007/s11926-018-0709-5

171. Theodorakopoulou MP, Minopoulou I, Sarafidis P, et al. Vascular endothelial injury assessed with functional techniques in systemic sclerosis patients with pulmonary arterial hypertension versus systemic sclerosis patients without pulmonary arterial hypertension: a systematic review and meta-analysis. Rheumatol Int. 2021;41(6):1045–1053. doi:10.1007/s00296-021-04850-2

172. Pattanaik D, Brown M, Postlethwaite AE. Vascular involvement in systemic sclerosis (scleroderma). J Inflamm Res. 2011;4:105–125. doi:10.2147/JIR.S18145

173. Yaqub A, Chung L. Epidemiology and risk factors for pulmonary hypertension in systemic sclerosis. Curr Rheumatol Rep. 2013;15(1):302. doi:10.1007/s11926-012-0302-2

174. Huang WC, Hsieh SC, Wu YW, et al. 2023 Taiwan Society of Cardiology (TSOC) and Taiwan College of Rheumatology (TCR) Joint Consensus on Connective Tissue Disease-Associated Pulmonary Arterial Hypertension. Acta Cardiol Sin. 2023;39(2):213–241. doi:10.6515/ACS.202303_39(2).20230117A

175. Bruni C, De Luca G, Lazzaroni MG, et al. Screening for pulmonary arterial hypertension in systemic sclerosis: a systematic literature review. Eur J Intern Med. 2020;78:17–25. doi:10.1016/j.ejim.2020.05.042

176. Jiang Y, Turk MA, Pope JE. Factors associated with pulmonary arterial hypertension (PAH) in systemic sclerosis (SSc). Autoimmun Rev. 2020;19(9):102602. doi:10.1016/j.autrev.2020.102602

177. Lewis RA, Durrington C, Condliffe R, Kiely DG. BNP/NT-proBNP in pulmonary arterial hypertension: time for point-of-care testing? Eur Respir Rev. 2020;29(156):200009. doi:10.1183/16000617.0009-2020

178. Semalulu T, Rudski L, Huynh T, et al. An evidence-based strategy to screen for pulmonary arterial hypertension in systemic sclerosis. Semin Arthritis Rheum. 2020;50(6):1421–1427. doi:10.1016/j.semarthrit.2020.02.013

179. Giuggioli D, Bruni C, Cacciapaglia F, et al. Pulmonary arterial hypertension: guidelines and unmet clinical needs. Reumatismo. 2021;72(4):228–246. doi:10.4081/reumatismo.2020.1310

180. Piera-Velazquez S, Dillon ST, Gu X, Libermann TA, Jimenez SA, Nita-Lazar A. Aptamer proteomics of serum exosomes from patients with Primary Raynaud’s and patients with Raynaud’s at risk of evolving into Systemic Sclerosis. PLoS One. 2022;17(12):e0279461. doi:10.1371/journal.pone.0279461

181. Bernero E, Sulli A, Ferrari G, et al. Prospective capillaroscopy-based study on transition from primary to secondary Raynaud’s phenomenon: preliminary results. Reumatismo. 2013;65(4):186–191. doi:10.4081/reumatismo.2013.186

182. Ruaro B, Smith V, Sulli A, et al. Innovations in the Assessment of Primary and Secondary Raynaud’s Phenomenon. Front Pharmacol. 2019;10:360. doi:10.3389/fphar.2019.00360

183. D’Alessandro R, Garcia Gonzalez E, et al. Peripheral Macrovascular Involvement in Systemic Sclerosis: a Cohort Study by Color and Spectral Doppler Ultrasonography. Life. 2023;13(2):487. doi:10.3390/life13020487

184. Belch J, Carlizza A, Carpentier PH, et al. ESVM guidelines - The diagnosis and management of Raynaud’s phenomenon. Vasa. 2017;46(6):413–423. doi:10.1024/0301-1526/a000661

185. Hickey PM, Lawrie A, Condliffe R. Circulating Protein Biomarkers in Systemic Sclerosis Related Pulmonary Arterial Hypertension: a Review of Published Data. Front Med. 2018;5:175. doi:10.3389/fmed.2018.00175

186. Moccaldi B, De Michieli L, Binda M, et al. Serum Biomarkers in Connective Tissue Disease-Associated Pulmonary Arterial Hypertension. Int J Mol Sci. 2023;24(4):4178. doi:10.3390/ijms24044178

187. Matucci-Cerinic M, Kahaleh B, Wigley FM. Review: evidence that systemic sclerosis is a vascular disease. Arthritis Rheum. 2013;65(8):1953–1962. doi:10.1002/art.37988

188. Zanin-Silva DC, Santana-Gonçalves M, Kawashima-Vasconcelos MY, Oliveira MC. Management of Endothelial Dysfunction in Systemic Sclerosis: current and Developing Strategies. Front Med. 2021;8:788250. doi:10.3389/fmed.2021.788250

189. Pulito-Cueto V, Genre F, López-Mejías R, et al. Endothelin-1 as a Biomarker of Idiopathic Pulmonary Fibrosis and Interstitial Lung Disease Associated with Autoimmune Diseases. Int J Mol Sci. 2023;24(2):1275. doi:10.3390/ijms24021275

190. Manetti M, Romano E, Rosa I, et al. Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann Rheum Dis. 2017;76(5):924–934. doi:10.1136/annrheumdis-2016-210229

191. Manetti M, Guiducci S, Romano E, et al. Differential expression of junctional adhesion molecules in different stages of systemic sclerosis. Arthritis Rheum. 2013;65(1):247–257. doi:10.1002/art.37712

192. Moschetti L, Piantoni S, Vizzardi E, et al. Endothelial Dysfunction in Systemic Lupus Erythematosus and Systemic Sclerosis: a Common Trigger for Different Microvascular Diseases. Front Med. 2022;9:849086. doi:10.3389/fmed.2022.849086

193. Chora I, Guiducci S, Manetti M, et al. Vascular biomarkers and correlation with peripheral vasculopathy in systemic sclerosis. Autoimmun Rev. 2015;14(4):314–322. doi:10.1016/j.autrev.2014.12.001

194. Sha W, Thompson K, South J, et al. Loss of PPARγ expression by fibroblasts enhances dermal wound closure. Fibrogenesis Tissue Repair. 2012;5(1):5. doi:10.1186/1755-1536-5-5

195. Avouac J, Meune C, Ruiz B, et al. Angiogenic biomarkers predict the occurrence of digital ulcers in systemic sclerosis. Ann Rheum Dis. 2012;71(3):394–399. doi:10.1136/annrheumdis-2011-200143

196. Iannone F, Riccardi MT, Guiducci S, et al. Bosentan regulates the expression of adhesion molecules on circulating T cells and serum soluble adhesion molecules in systemic sclerosis-associated pulmonary arterial hypertension. Ann Rheum Dis. 2008;67(8):1121–1126. doi:10.1136/ard.2007.080424

197. Oller-Rodríguez JE, Vicens Bernabeu E, Gonzalez-Mazarío R, Grau García E, Ortiz Sanjuan FM, Román Ivorra JA. Utility of cytokines CXCL4, CXCL8 and GDF15 as biomarkers in systemic sclerosis. Med Clin (Barc). 2022;159(8):359–365. doi:10.1016/j.medcli.2021.12.006

198. Papaioannou AI, Zakynthinos E, Kostikas K, et al. Serum VEGF levels are related to the presence of pulmonary arterial hypertension in systemic sclerosis. BMC Pulm Med. 2009;9(1):18. doi:10.1186/1471-2466-9-18

199. Silva I, Almeida C, Teixeira A, Oliveira J, Vasconcelos C. Impaired angiogenesis as a feature of digital ulcers in systemic sclerosis. Clin Rheumatol. 2016;35(7):1743–1751. doi:10.1007/s10067-016-3219-8

200. Xu B, Xu G, Yu Y, Lin J. The role of TGF-β or BMPR2 signaling pathway-related miRNA in pulmonary arterial hypertension and systemic sclerosis. Arthritis Res Ther. 2021;23(1):288. doi:10.1186/s13075-021-02678-6

201. Grignaschi S, Sbalchiero A, Spinozzi G, et al. Endoglin and Systemic Sclerosis: a PRISMA-driven systematic review. Front Med. 2022;9:964526. doi:10.3389/fmed.2022.964526

202. Odler B, Foris V, Gungl A, et al. Biomarkers for Pulmonary Vascular Remodeling in Systemic Sclerosis: a Pathophysiological Approach. Front Physiol. 2018;9:587. doi:10.3389/fphys.2018.00587

203. Kawashiri SY, Ueki Y, Terada K, Yamasaki S, Aoyagi K, Kawakami A. Improvement of plasma endothelin-1 and nitric oxide in patients with systemic sclerosis by bosentan therapy. Rheumatol Int. 2014;34(2):221–225. doi:10.1007/s00296-013-2861-6

204. Wu Q, Cao F, Tao J, Li X, Zheng SG, Pan HF. Pentraxin 3: a promising therapeutic target for autoimmune diseases. Autoimmun Rev. 2020;19(12):102584. doi:10.1016/j.autrev.2020.102584

205. Kanno Y. The Role of Fibrinolytic Regulators in Vascular Dysfunction of Systemic Sclerosis. Int J Mol Sci. 2019;20(3):619. doi:10.3390/ijms20030619

206. Giannelli G, Iannone F, Marinosci F, Lapadula G, Antonaci S. The effect of bosentan on matrix metalloproteinase-9 levels in patients with systemic sclerosis-induced pulmonary hypertension. Curr Med Res Opin. 2005;21(3):327–332. doi:10.1185/030079905X30680

207. Manetti M, Guiducci S, Romano E, et al. Increased serum levels and tissue expression of matrix metalloproteinase-12 in patients with systemic sclerosis: correlation with severity of skin and pulmonary fibrosis and vascular damage. Ann Rheum Dis. 2012;71(6):1064–1072. doi:10.1136/annrheumdis-2011-200837

208. Romano E, Manetti M, Rosa I, et al. Slit2/Robo4 axis may contribute to endothelial cell dysfunction and angiogenesis disturbance in systemic sclerosis. Ann Rheum Dis. 2018;77(11):1665–1674. doi:10.1136/annrheumdis-2018-213239

209. Yokoyama K, Mitoma H, Kawano S, et al. CEACAM 1, 3, 5 and 6 -positive classical monocytes correlate with interstitial lung disease in early systemic sclerosis. Front Immunol. 2022;13:1016914. doi:10.3389/fimmu.2022.1016914

210. Pellicano C, Iannazzo F, Romaggioli L, Rosato E. IL33 and sST2 serum level in systemic sclerosis microvascular involvement. Microvasc Res. 2022;142:104344. doi:10.1016/j.mvr.2022.104344

211. Kolstad KD, Khatri A, Donato M, et al. Cytokine signatures differentiate systemic sclerosis patients at high versus low risk for pulmonary arterial hypertension. Arthritis Res Ther. 2022;24(1):39. doi:10.1186/s13075-022-02734-9

212. Jiang Z, Chen C, Yang S, He H, Zhu X, Liang M. Contribution to the peripheral vasculopathy and endothelial cell dysfunction by CXCL4 in Systemic Sclerosis. J Dermatol Sci. 2021;104(1):63–73. doi:10.1016/j.jdermsci.2021.07.006

213. Ichimura Y, Asano Y, Akamata K, et al. Fli1 deficiency contributes to the suppression of endothelial CXCL5 expression in systemic sclerosis. Arch Dermatol Res. 2014;306(4):331–338. doi:10.1007/s00403-013-1431-9

214. Iannone F, Praino E, Rotondo C, et al. Body mass index and adipokines/cytokines dysregulation in systemic sclerosis. Clin Exp Immunol. 2021;206(2):153–160. doi:10.1111/cei.13651

215. Korman BD, Marangoni RG, Hinchcliff M, et al. Brief Report: association of Elevated Adipsin Levels With Pulmonary Arterial Hypertension in Systemic Sclerosis. Arthritis Rheumatol. 2017;69(10):2062–2068. doi:10.1002/art.40193

216. Sawicka K, Michalska-Jakubus M, Potembska E, Kowal M, Pietrzak A, Krasowska D. Visfatin and chemerin levels correspond with inflammation and might reflect the bridge between metabolism, inflammation and fibrosis in patients with systemic sclerosis. Postepy Dermatol Alergol. 2019;36(5):551–565. doi:10.5114/ada.2018.79104

217. Bălănescu P, Lădaru A, Bălănescu E, Nicolau A, Băicuş C, Dan GA. IL-17, IL-6 and IFN-γ in Systemic Sclerosis Patients. Rom J Intern Med. 2015;53(1):44–49. doi:10.1515/rjim-2015-0006

218. Sun C, Zhu H, Wang Y, et al. Serum metabolite differences detected by HILIC UHPLC-Q-TOF MS in systemic sclerosis. Clin Rheumatol. 2023;42(1):125–134. doi:10.1007/s10067-022-06372-z

219. Long H, Wang X, Chen Y, Wang L, Zhao M, Lu Q. Dysregulation of microRNAs in autoimmune diseases: pathogenesis, biomarkers and potential therapeutic targets. Cancer Lett. 2018;428:90–103. doi:10.1016/j.canlet.2018.04.016

220. Zhang Y, Qin D, Qin L, Yang X, Luo Q, Wang H. Diagnostic value of cardiac natriuretic peptide on pulmonary hypertension in systemic sclerosis: a systematic review and meta-analysis. Joint Bone Spine. 2022;89(2):105287. doi:10.1016/j.jbspin.2021.105287

221. Pagkopoulou E, Soulaidopoulos S, Katsiki N, et al. The role of asymmetric dimethylarginine in endothelial dysfunction and abnormal nitric oxide metabolism in systemic sclerosis: results from a pilot study. Clin Rheumatol. 2023;42(4):1077–1085. doi:10.1007/s10067-022-06472-w

222. Pagkopoulou E, Soulaidopoulos S, Triantafyllidou E, et al. Asymmetric dimethylarginine correlates with worsening peripheral microangiopathy in systemic sclerosis. Microvasc Res. 2023;145:104448. doi:10.1016/j.mvr.2022.104448

223. Xanthouli P, Gordjani O, Benjamin N, et al. Hypochromic red cells as a prognostic indicator of survival among patients with systemic sclerosis screened for pulmonary hypertension. Arthritis Res Ther. 2023;25(1):38. doi:10.1186/s13075-023-03020-y

224. Chikhoune L, Brousseau T, Morell-Dubois S, et al. Association between Routine Laboratory Parameters and the Severity and Progression of Systemic Sclerosis. J Clin Med. 2022;11(17):5087. doi:10.3390/jcm11175087

225. Kayser C, Fritzler MJ. Autoantibodies in systemic sclerosis: unanswered questions. Front Immunol. 2015;6:167. doi:10.3389/fimmu.2015.00167

226. Lomelí-Nieto JA, Muñoz-Valle JF, Baños-Hernández CJ, et al. Transforming growth factor beta isoforms and TGF-βR1 and TGF-βR2 expression in systemic sclerosis patients. Clin Exp Med. 2022;23(2):471–481. doi:10.1007/s10238-022-00841-0

227. Isomura Y, Shirai Y, Kuwana M. Clinical worsening following discontinuation of tocilizumab in diffuse cutaneous systemic sclerosis: a single-centre experience in Japan. Rheumatology. 2022;61(11):4491–4496. doi:10.1093/rheumatology/keac136

228. Lescoat A, Roofeh D, Kuwana M, et al. Therapeutic Approaches to Systemic Sclerosis: recent Approvals and Future Candidate Therapies. Clin Rev Allergy Immunol. 2023;64(3):239–261. doi:10.1007/s12016-021-08891-0

229. Stifano G, Sornasse T, Rice LM, et al. Skin Gene Expression Is Prognostic for the Trajectory of Skin Disease in Patients With Diffuse Cutaneous Systemic Sclerosis. Arthritis Rheumatol. 2018;70(6):912–919. doi:10.1002/art.40455

230. Clark KEN, Csomor E, Campochiaro C, et al. Integrated analysis of dermal blister fluid proteomics and genome-wide skin gene expression in systemic sclerosis: an observational study. Lancet Rheumatol. 2022;4(7):e507–e516. doi:10.1016/S2665-9913(22)00094-7

231. Gordon JK, Martyanov V, Franks JM, et al. Belimumab for the Treatment of Early Diffuse Systemic Sclerosis: results of a Randomized, Double-Blind, Placebo-Controlled, Pilot Trial. Arthritis Rheumatol. 2018;70(2):308–316. doi:10.1002/art.40358

232. Wirz EG, Jaeger VK, Allanore Y, et al. Incidence and predictors of cutaneous manifestations during the early course of systemic sclerosis: a 10-year longitudinal study from the EUSTAR database. Ann Rheum Dis. 2016;75(7):1285–1292. doi:10.1136/annrheumdis-2015-207271

233. Kuwana M, Allanore Y, Denton CP, et al. Nintedanib in Patients With Systemic Sclerosis-Associated Interstitial Lung Disease: subgroup Analyses by Autoantibody Status and Modified Rodnan Skin Thickness Score. Arthritis Rheumatol. 2022;74(3):518–526. doi:10.1002/art.41965

234. Namas R, Tashkin DP, Furst DE, et al. Efficacy of Mycophenolate Mofetil and Oral Cyclophosphamide on Skin Thickness: post Hoc Analyses From Two Randomized Placebo-Controlled Trials. Arthritis Care Res. 2018;70(3):439–444. doi:10.1002/acr.23282

235. Johnson SR, Feldman BM, Pope JE, Tomlinson GA. Shifting our thinking about uncommon disease trials: the case of methotrexate in scleroderma. J Rheumatol. 2009;36(2):323–329. doi:10.3899/jrheum.071169

236. Roden AC, Camus P. Iatrogenic pulmonary lesions. Semin Diagn Pathol. 2018;35(4):260–271. doi:10.1053/j.semdp.2018.03.002

237. Karalilova RV, Batalov ZA, Sapundzhieva TL, Matucci-Cerinic M, Batalov AZ. Tofacitinib in the treatment of skin and musculoskeletal involvement in patients with systemic sclerosis, evaluated by ultrasound. Rheumatol Int. 2021;41(10):1743–1753. doi:10.1007/s00296-021-04956-7

238. Khanna D, Denton CP, Furst DE, et al. A 24-Week, Phase IIa, Randomized, Double-blind, Placebo-controlled Study of Ziritaxestat in Early Diffuse Cutaneous Systemic Sclerosis (NOVESA). Arthritis Rheumatol. 2023. doi:10.1002/art.42477

239. Kuzumi A, Ebata S, Fukasawa T, et al. Long-term Outcomes After Rituximab Treatment for Patients With Systemic Sclerosis: follow-up of the DESIRES Trial With a Focus on Serum Immunoglobulin Levels. JAMA Dermatol. 2023;159(4):374–383. doi:10.1001/jamadermatol.2022.6340

240. Ebata S, Oba K, Kashiwabara K, et al. Predictors of rituximab effect on modified Rodnan skin score in systemic sclerosis: a machine-learning analysis of the DesiReS trial. Rheumatology. 2022;61(11):4364–4373. doi:10.1093/rheumatology/keac023

241. Frech TM, Frech M, Saknite I, et al. Novel therapies and innovation for systemic sclerosis skin ulceration. Best Pract Res Clin Rheumatol. 2023;4:101813. doi:10.1016/j.berh.2022.101813

242. Davuluri S, Lood C, Chung L. Calcinosis in systemic sclerosis. Curr Opin Rheumatol. 2022;34(6):319–327. doi:10.1097/BOR.0000000000000896

243. Touart DM, Sau P. Cutaneous deposition diseases. Part I. J Am Acad Dermatol. 1998;39(2 Pt 1):149–171. doi:10.1016/S0190-9622(98)70069-6

244. Herrick AL. Raynaud’s phenomenon and digital ulcers: advances in evaluation and management. Curr Opin Rheumatol. 2021;33(6):453–462. doi:10.1097/BOR.0000000000000826

245. Flavahan NA. New mechanism-based approaches to treating and evaluating the vasculopathy of scleroderma. Curr Opin Rheumatol. 2021;33(6):471–479. doi:10.1097/BOR.0000000000000830

246. Volkmann ER, McMahan Z. Gastrointestinal involvement in systemic sclerosis: pathogenesis, assessment and treatment. Curr Opin Rheumatol. 2022;34(6):328–336. doi:10.1097/BOR.0000000000000899

247. Khanna D, Hays RD, Maranian P, et al. Reliability and validity of the University of California, Los Angeles Scleroderma Clinical Trial Consortium Gastrointestinal Tract Instrument. Arthritis Rheum. 2009;61(9):1257–1263. doi:10.1002/art.24730

248. Lafyatis R, Valenzi E. Assessment of disease outcome measures in systemic sclerosis. Nat Rev Rheumatol. 2022;18(9):527–541. doi:10.1038/s41584-022-00803-6

249. Hamberg V, Wallman JK, Mogard E, Lindqvist E, Olofsson T, Andréasson K. Elevated fecal levels of the inflammatory biomarker calprotectin in early systemic sclerosis. Rheumatol Int. 2023;43(5):961–967. doi:10.1007/s00296-022-05264-4

250. Ebrahimi B, Nazarinia M, Molayem M. Calprotectin, an available prognostic biomarker in systemic sclerosis: a systematic review. Clin Rheumatol. 2021;40(5):1709–1715. doi:10.1007/s10067-020-05446-0

251. Cole A, Ong VH, Denton CP. Renal Disease and Systemic Sclerosis: an Update on Scleroderma Renal Crisis. Clin Rev Allergy Immunol. 2023;64(3):378–391. doi:10.1007/s12016-022-08945-x

252. Clark KE. Review of systemic sclerosis and antineutrophil cytoplasmic antibody vasculitis overlap: using autoantibodies for a personalised medicine approach. J Scleroderma Relat Disord. 2023;8(1):7–13. doi:10.1177/23971983221126850

253. Chew E, Barnado A, Ikizler TA, Zent R, Frech T. Evaluation of hypertension in systemic sclerosis and systemic lupus erythematosus overlap. J Scleroderma Relat Disord. 2023;8(1):14–19. doi:10.1177/23971983221122673

254. Bose N, Chiesa-Vottero A, Chatterjee S. Scleroderma renal crisis. Semin Arthritis Rheum. 2015;44(6):687–694. doi:10.1016/j.semarthrit.2014.12.001

255. Denton CP, Sweny P, Abdulla A, Black CM. Acute renal failure occurring in scleroderma treated with cyclosporin A: a report of three cases. Br J Rheumatol. 1994;33(1):90–92. doi:10.1093/rheumatology/33.1.90

256. Tonsawan P, Talabthong K, Puapairoj A, Foocharoen C. Renal pathology and clinical associations in systemic sclerosis: a historical cohort study. Int J Gen Med. 2019;12:323–331. doi:10.2147/IJGM.S221471

257. Hudson M, Baron M, Tatibouet S, Furst DE, Khanna D. International Scleroderma Renal Crisis Study Investigators. Exposure to ACE inhibitors prior to the onset of scleroderma renal crisis-results from the International Scleroderma Renal Crisis Survey. Semin Arthritis Rheum. 2014;43(5):666–672. doi:10.1016/j.semarthrit.2013.09.008

258. Steen VD, Mayes MD, Merkel PA. Assessment of kidney involvement. Clin Exp Rheumatol. 2003;21(3 Suppl 29):S29–31.

259. Berger M, Steen VD. Role of anti-receptor autoantibodies in pathophysiology of scleroderma. Autoimmun Rev. 2017;16(10):1029–1035. doi:10.1016/j.autrev.2017.07.019

260. Nagaraja V. Management of scleroderma renal crisis. Curr Opin Rheumatol. 2019;31(3):223–230. doi:10.1097/BOR.0000000000000604

261. Hoa S, Stern EP, Denton CP, Hudson M; Scleroderma Clinical Trials Consortium Scleroderma Renal Crisis Working Group Investigators of the Scleroderma Clinical Trials Consortium Scleroderma Renal Crisis Working Group. Towards developing criteria for scleroderma renal crisis: a scoping review. Autoimmun Rev. 2017;16(4):407–415. doi:10.1016/j.autrev.2017.02.012

262. Okrój M, Johansson M, Saxne T, Blom AM, Hesselstrand R. Analysis of complement biomarkers in systemic sclerosis indicates a distinct pattern in scleroderma renal crisis. Arthritis Res Ther. 2016;18(1):267. doi:10.1186/s13075-016-1168-x

263. Osthoff M, Jaeger VK, Heijnen IAFM, et al. Role of lectin pathway complement proteins and genetic variants in organ damage and disease severity of systemic sclerosis: a cross-sectional study. Arthritis Res Ther. 2019;21(1):76. doi:10.1186/s13075-019-1859-1

264. Shimizu K, Ogawa F, Yoshizaki A, et al. Increased serum levels of soluble CD163 in patients with scleroderma. Clin Rheumatol. 2012;31(7):1059–1064. doi:10.1007/s10067-012-1972-x

265. Yanaba K, Asano Y, Tada Y, et al. Increased serum soluble CD147 levels in patients with systemic sclerosis: association with scleroderma renal crisis. Clin Rheumatol. 2012;31(5):835–839. doi:10.1007/s10067-012-1949-9

266. Fonseca C, Renzoni E, Sestini P, et al. Endothelin axis polymorphisms in patients with scleroderma. Arthritis Rheum. 2006;54(9):3034–3042. doi:10.1002/art.22036

267. Penn H, Quillinan N, Khan K, et al. Targeting the endothelin axis in scleroderma renal crisis: rationale and feasibility. QJM. 2013;106(9):839–848. doi:10.1093/qjmed/hct111

268. Stratton RJ, Coghlan JG, Pearson JD, et al. Different patterns of endothelial cell activation in renal and pulmonary vascular disease in scleroderma. QJM. 1998;91(8):561–566. doi:10.1093/qjmed/91.8.561

269. Chighizola CB, Pregnolato F, Meroni PL, Denton CP, Ong VH. N-terminal pro Brain Natriuretic Peptide as predictor of outcome in scleroderma renal crisis. Clin Exp Rheumatol. 2016;34 Suppl 100(5):122–128.

270. Penn H, Howie AJ, Kingdon EJ, et al. Scleroderma renal crisis: patient characteristics and long-term outcomes. QJM. 2007;100(8):485–494. doi:10.1093/qjmed/hcm052

271. Tormey VJ, Bunn CC, Denton CP, Black CM. Anti-fibrillarin antibodies in systemic sclerosis. Rheumatology. 2001;40(10):1157–1162. doi:10.1093/rheumatology/40.10.1157

272. Codullo V, Cavazzana I, Bonino C, et al. Serologic profile and mortality rates of scleroderma renal crisis in Italy. J Rheumatol. 2009;36(7):1464–1469. doi:10.3899/jrheum.080806

273. Merashli M, Alves J, Ames PRJ. Clinical relevance of antiphospholipid antibodies in systemic sclerosis: a systematic review and meta-analysis. Semin Arthritis Rheum. 2017;46(5):615–624. doi:10.1016/j.semarthrit.2016.10.004

274. Ogawa F, Shimizu K, Hara T, et al. Autoantibody against one of the antioxidant repair enzymes, methionine sulfoxide reductase A, in systemic sclerosis: association with pulmonary fibrosis and vascular damage. Arch Dermatol Res. 2010;302(1):27–35. doi:10.1007/s00403-009-0996-9

275. Clements PJ, Lachenbruch PA, Furst DE, Maxwell M, Danovitch G, Paulus HE. Abnormalities of renal physiology in systemic sclerosis. A prospective study with 10-year followup. Arthritis Rheum. 1994;37(1):67–74. doi:10.1002/art.1780370110

276. Gigante A, Barbano B, Barilaro G, et al. Serum uric acid as a marker of microvascular damage in systemic sclerosis patients. Microvasc Res. 2016;106:39–43. doi:10.1016/j.mvr.2016.03.007

277. Reiseter S, Molberg Ø, Gunnarsson R, et al. Associations between circulating endostatin levels and vascular organ damage in systemic sclerosis and mixed connective tissue disease: an observational study. Arthritis Res Ther. 2015;17(1):231. doi:10.1186/s13075-015-0756-5

278. Akamata K, Asano Y, Taniguchi T, et al. Increased expression of chemerin in endothelial cells due to Fli1 deficiency may contribute to the development of digital ulcers in systemic sclerosis. Rheumatology. 2015;54(7):1308–1316. doi:10.1093/rheumatology/keu479

279. Takahashi T, Asano Y, Noda S, et al. A possible contribution of lipocalin-2 to the development of dermal fibrosis, pulmonary vascular involvement and renal dysfunction in systemic sclerosis. Br J Dermatol. 2015;173(3):681–689. doi:10.1111/bjd.13779

280. Aozasa N, Asano Y, Akamata K, et al. Serum apelin levels: clinical association with vascular involvements in patients with systemic sclerosis. J Eur Acad Dermatol Venereol. 2013;27(1):37–42. doi:10.1111/j.1468-3083.2011.04354.x

281. Pawlik KK, Bohdziewicz A, Chrabąszcz M, et al. Biomarkers of disease activity in systemic sclerosis. Wiad Lek. 2020;73(10):2300–2305. doi:10.36740/WLek202010137

282. Akamata K, Asano Y, Noda S, et al. An inverse correlation of serum angiogenin levels with estimated glomerular filtration rate in systemic sclerosis patients with renal dysfunction. Eur J Dermatol. 2013;23(2):269–270. doi:10.1684/ejd.2013.1924

283. Żółkiewicz J, Stochmal A, Rudnicka L. The role of adipokines in systemic sclerosis: a missing link? Arch Dermatol Res. 2019;311(4):251–263. doi:10.1007/s00403-019-01893-1

284. Stern EP, Unwin R, Burns A, Ong VH, Denton CP. Exploring molecular pathology of chronic kidney disease in systemic sclerosis by analysis of urinary and serum proteins. Rheumatol Adv Pract. 2021;5(1):rkaa083. doi:10.1093/rap/rkaa083

Creative Commons License © 2023 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.