Next Article in Journal
Genome-Wide Identification, Expansion, and Evolution Analysis of Homeobox Gene Family Reveals TALE Genes Important for Secondary Cell Wall Biosynthesis in Moso Bamboo (Phyllostachys edulis)
Next Article in Special Issue
Secondary Osteoporosis: A Still Neglected Condition
Previous Article in Journal
Controversies in the Pathogenesis, Diagnosis and Treatment of PCOS: Focus on Insulin Resistance, Inflammation, and Hyperandrogenism
Previous Article in Special Issue
Bone Fragility in Gastrointestinal Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Biological/Targeted Therapies on Bone Mineral Density in Inflammatory Arthritis

1
Department of Medical Education, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
2
Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
3
Division of Plastic Surgery, Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
4
Division of Allergy, Immunology and Rheumatology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
5
School of Medicine, Tzu Chi University, Hualien 970, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(8), 4111; https://doi.org/10.3390/ijms23084111
Submission received: 2 March 2022 / Revised: 6 April 2022 / Accepted: 7 April 2022 / Published: 8 April 2022
(This article belongs to the Special Issue Secondary Osteoporosis in Adults 2.0)

Abstract

:
Inflammatory arthritis has been reported to be associated with the development of osteoporosis. Recent research has investigated the mechanisms of bone metabolism in chronic inflammatory arthritis such as rheumatoid arthritis (RA) and spondyloarthritis (SpA). Progress in both animal and clinical studies has provided a better understanding of the osteoclastogenesis-related pathways regarding the receptor activator of nuclear factor-κB ligand (RANKL), anti-citrullinated protein antibodies (ACPAs), and Wnt signaling and Dickkopf-related protein 1 (Dkk-1). The complex interplay between inflammatory cytokines and bone destruction has been elucidated, especially that in the interleukin-17/23 (IL-17/23) axis and Janus kinase and signal transducer and activator of transcription (JAK-STAT) signaling. Moreover, advances in biological and targeted therapies have achieved essential modifications to the bone metabolism of these inflammatory arthritis types. In this narrative review, we discuss recent findings on the pathogenic effects on bone in RA and SpA. Proinflammatory cytokines, autoantibodies, and multiple signaling pathways play an essential role in bone destruction in RA and SpA patients. We also reviewed the underlying pathomechanisms of bone structure in biological and targeted therapies of RA and SpA. The clinical implications of tumor necrosis factor inhibitors, abatacept, rituximab, tocilizumab, Janus kinase inhibitors, and inhibitors of the IL-17/23 axis are discussed. Since these novel therapeutics provide new options for disease improvement and symptom control in patients with RA and SpA, further rigorous evidence is warranted to provide a clinical reference for physicians and patients.

1. Introduction

Osteoporosis is associated with inflammatory arthritis, which has had an increasing global prevalence in recent decades [1]. Patient care regarding osteoporosis requires integrated interdisciplinary care [1]. Bone involvement is the hallmark of several inflammatory arthritis types, such as rheumatoid arthritis (RA) and spondyloarthritis (SpA) [2,3]. There are three major forms of bone loss: (a) focal bone erosions at the joint margins, (b) periarticular osteolysis and new bone formation adjacent to the inflamed joints, and (c) systemic bone loss (osteoporosis). The influence of RA and SpA on bone metabolism and arthritic pathology can lead to arthralgia, joint swelling, and limited physical activity. As these bone involvements significantly affect patients’ quality of life, the underlying mechanisms of bone loss in chronic inflammatory arthritis have driven both laboratory and clinical discussions. Previous studies have investigated bone remodeling in patients with inflammatory arthritis [4,5]. The pathophysiology of bone remodeling is complex, including interactions between the inflammatory and immune systems. The associations of inflammatory cytokines with osteocytes and bone resorption are also complicated.
As the understanding of the pathophysiology of RA and SpA has increased, numerous biological/targeted therapies have been studied and approved for clinical applications. Recent studies have investigated their effects on bone mineral density and bone metabolism, which may also impact clinical symptoms and patients’ quality of life. In this narrative review, we provide an overview of the pathophysiology of RA and SpA. We primarily focused on the effect of targeted therapies, including biologics and small molecule inhibitors, on bone mineral density and osteoporosis in inflammatory arthritis. In Figure 1, we summarize the pathophysiology discussed in this article.

2. Rheumatoid Arthritis

2.1. Bone Resorption and Osteoporosis in Rheumatoid Arthritis

RA is a chronic immune-mediated disease affecting about 5 in 1000 people worldwide [6]. The development of RA involves a complex interplay among the immune system, gene predisposition, and environmental triggers [7,8]. Clinically, RA is characterized by articular bone erosion, progressive joint destruction, and deformities, resulting in disability [2,9]. Of note, RA increases the risk of osteoporosis and joint cartilage destruction [9]. Clinical evidence has also supported an increased fracture risk in patients suffering from RA [9]. Indeed, examinations using high-resolution peripheral quantitative computed tomography (HRpQCT) have found that RA patients are associated with increased cortical porosity and reduced bone strength, representing higher risks of fractures than healthy controls [10]. Recent studies have investigated the underlying pathogenesis of bone resorption in RA, which is discussed below.

2.1.1. Proinflammatory Cytokines and Synovitis

Inflammation in RA is mainly driven by cytokines, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1 (IL-1) [11]. The involved proinflammatory cytokines can directly or indirectly provoke osteoclast activation and halt osteoblast differentiation, leading to bone loss and subsequent osteoporosis [12,13]. Cytokines such as interleukin (IL)-1β, IL-6, IL-8, and IL-11 have been reported to be directly associated with osteoclastogenesis. Other cytokines such as TNF-α, IL-7, and IL-15 may indirectly affect the formation of osteoclasts or the inhibition of osteoblasts. In addition, chronic inflammation can lead to osteoporosis through the release of matrix metalloproteinases (MMPs) [14]. The receptor activator of nuclear factor-κB ligand (RANKL) is one of the major cytokines that could be modulated in the pathogenesis of RA bone loss and cartilage damage [7,15]. In patients with RA, the primary source of RANKL is synovial fibroblasts and CD4 + CD28- T -cells. Moreover, RANKL was found to produce both a positive effect on osteoclastogenesis and a detrimental effect on osteoblastic development [16]. A complicated interplay between multiple proinflammatory cytokines and RANKL-associated osteoclastogenesis is considered to have clinical effects on osteolysis.

2.1.2. Autoantibodies

Anti-citrullinated protein antibodies (ACPAs) are the most specific serological biomarkers not only for predicting the development of RA but also for disease prognosis [17,18]. ACPAs are mainly produced by plasmablasts and plasma cells [17,18]. Recent animal studies have suggested that ACPAs may stimulate osteoclast differentiation and initiate bone change [19]. ACPAs enhance osteoclast differentiation from monocyte-derived or circulating CD1c+ dendritic cells (DCs) by increasing the release of IL-8 [20]. ACPAs binding to immature DCs might be associated with the activation and differentiation toward the osteoclast lineage, facilitating bone erosion in ACPA-positive RA [21,22]. Interestingly, immunization with citrullinated Type II mouse collagen resulted in increased ACPA levels and lowered bone quality, but these were uncoupled from the degree of inflammation [23]. Recent advances in mass spectrometry have even proposed insights into a term called “citrullinome” in RA [24]. In in vitro models, ACPAs isolated from RA patients had the potential to stimulate both murine and human osteoclast precursor cells [22,25,26]. ACPAs transferred to mice had the ability to bond to bone marrow-resident osteoclasts and osteoclast precursor cells, leading to joint pain and bone erosion [22,23]. This mechanism of bone resorption could explain why bone destruction in RA progresses with the existence of inflammation (in healthy ACPA-positive individuals or patients achieving sustained clinical remission) [22,25]. Further analyses have identified extracellular IL-8 as the key mediator of ACPA-triggered osteoclastogenesis and bone remodeling in mouse models [23,26]. Most importantly, blocking IL-8 activity could reverse the pathogenic effects of ACPAs both in vitro and in vivo [22,25,26]. Future translational research is needed to clarify its clinical importance.

2.1.3. The Wnt Signaling Pathway and Dickkopf-Related Protein 1 (Dkk-1)

Wnt proteins are palmitoylated and glycosylated ligands that have a pivotal role in the regulation of bone remodeling [27]. Wnt signaling in osteoblasts regulates the expression of RANKL [28]. Furthermore, Wnt/β-catenin signaling directly affects the regulation of osteoclastogenesis [29,30,31]. The Wnt and the osteoprotegerin (OPG)-RANKL-RANK signaling systems, as critical mediators, interact in subchondral bone remodeling [32,33]. Dkk-1, a Wnt signaling inhibitor, is a key regulator of bone remodeling [34]. Increased serum Dkk-1 was associated with a higher risk of bone destruction and osteoporosis in patients with RA [35]. The levels of Dkk-1 expression were elevated in the synovial fluid of RA patients [36,37]. Interestingly, the expression of Dkk-1 by synovial fibroblasts leads to the inhibition of osteoblast differentiation and new bone formation [38]. Understanding the crosstalk between the Wnt pathway and RANKL-associated bone effects may facilitate current pharmacologic developments.
Wnt signaling is also regulated by proinflammatory pathways, namely TNF-α and IL-1 β (through the induction of Dkk-1 and sclerostin), IL-6, and IL-17 [39]. Additionally, Wnt signaling is inhibited by plasmablasts and plasma cells through the expression of Dkk-1 [34,39], which influences the expression of RANKL. Studies on this are still in the laboratory setting without further confirmation in patients with RA.

2.2. Bone Effect of Biological/Targeted Therapies in Rheumatoid Arthritis

Effective treatment is necessary for the bothersome symptoms related to RA bone involvement. According to European League Against Rheumatism (EULAR) recommendations, synthetic and biologic disease-modifying antirheumatic drugs (DMARDs) are the mainstay in RA treatment [40]. Apart from conventional synthetic DMARDs (methotrexate, leflunomide, sulfasalazine, and hydroxychloroquine) and glucocorticoids, the development of biological (b) DMARDs and targeted synthetic (ts) DMARDs has brought RA management into a new era. In clinical trials, the clinical performance of biologics and target therapies were favorable, with tolerable adverse effects.
B/tsDMARD therapy directly targets or indirectly modulates cytokines and halts the inflammatory cascade; moreover, the pathways that biologics targets also interact with the innate and adaptive immune system. The application of b/tsDMARD therapies has demonstrated anti-inflammatory effects for RA patients. Since the advent of b/tsDMARDs, the regimens have been found to alleviate clinical symptoms, improve the quality of life, and decelerate joint damage. To date, cumulative evidence has discussed the effect of b/tsDMARDs on bone metabolism, and we summarized its pathophysiology in Figure 1.

2.2.1. Tumor Necrosis Factor Inhibitors

Tumor necrosis factor (TNF) is a protein that is mainly produced by macrophages or monocytes, which participates in immune response regulation [14]. TNF-α plays an essential role in RA, which affects the action of synovial cells, macrophages, T-cells, B-cells, and endothelial cells [14]. The binding of TNF and TNF receptors activates caspase-dependent death signaling pathways with anti-apoptotic and proinflammatory responses [41]. TNF inhibitors are engineered monoclonal antibodies developed to competitively bind to TNF receptors and induce cell lysis by activation of complement-dependent or antibody-dependent cellular cytotoxicity [41]. The effects of TNF inhibitors (adalimumab, certolizumab pegol, etanercept, golimumab, and infliximab) on bone mineral density and osteoporosis in RA patients have been studied. TNF blockades may slow down generalized bone resorption, in association with clinical improvements [42]. In mouse models, inhibition of TNF showed a positive effect on bone formation and decreased osteoclastogenesis [43]. Anti-TNF therapy was also associated with elevated bone formation markers (e.g., procollagen Type I N-terminal propeptide (PINP)) and a decrease in bone resorption markers (e.g., C-terminal telopeptide of Type I collagen (CTX-I) and C-terminal cross-linked telopeptide of Type I collagen (ICTP)) in serum [44,45]. The effects on bone remodeling produced decreased levels of DKK-1 and increased levels of intact PINP [46]. A systematic review concluded that TNF inhibition might have a protective effect on the cartilage in the joint microenvironment [47].
In clinical observations, adalimumab has the potential to reduce bone damage and halt hand bone loss [48,49]. Infliximab has been found not only to increase BMD [50] but also to prevent arthritis-related osteoporosis and suppress tendon inflammation, thus alleviating tendon-related pain [51]. In patients with RA treated with infliximab, spine and hip bone damage was arrested, whereas metacarpal cortical hand bone destruction was not stopped [52]. Nevertheless, a recent study concluded that long-term use of infliximab did not affect bone microstructure and morphology in rats in the absence of an inflammatory condition [53]. Moreover, TNF inhibitors may suppress joint destruction and reduce the joint soreness caused by synovitis in a study with a 12-month follow-up period [47]. TNF inhibitors provoked a short-term rise in PTH levels and an early increase in bone turnover [54,55]. However, the evidence regarding fracture risk among RA patients receiving biologics is conflicting [56,57].

2.2.2. Abatacept

Abatacept is a cytotoxic T lymphocyte-associated antigen 4 immunoglobulin fusion protein (CTLA-4-Ig) that can bind to CD80 or CD86, and subsequently prevent the signaling between T-cells and antigen-presenting cells [58]. Abatacept also competes with CD28 for CD80 or CD86 binding, and selectively regulates T-cell activation [59]. The inhibition of CD28-mediated T-cell activation effectively controls inflammation and inhibits bone erosion during RA [60]. By interfering with intracellular calcium oscillations in bone marrow macrophages, abatacept directly inhibits osteoclastogenesis [61]. Additionally, abatacept promoted T-cell Wnt protein production and prevented bone loss in a mouse model [62,63].
Clinically, serum OPG was significantly elevated and serum Dkk-1 was considerably lower 6 months after the introduction of abatacept [64]. The efficacy of abatacept for increasing BMD at the femoral neck was better than that of other bDMARDs [65]. Abatacept also had good efficacy for improving BMD at the femoral neck in patients with RA [66]. In RA patients with more severe disease activity and higher anti-CCP2 concentrations, treatment with abatacept was associated with more significant improvements in patient-reported outcomes over the following 6 months [67]. Hence, abatacept may improve RA patients’ quality of life and daily activity through mitigating bone pain and joint pain.

2.2.3. Rituximab

Rituximab is a monoclonal antibody against CD20 that selectively targets B-cells and is approved as a second-line therapy for RA patients [68,69]. Since CD20 molecules are involved in complement activation, rituximab therefore induces complement-mediated cytotoxicity [68]. Rituximab may also cause structural changes and apoptosis of inflammatory cells [68,69]. Such B-cell depletion therapy has been reported to have a direct effect on bone remodeling in mouse models [70]. Rituximab and its associated immune response may have an essential role in regulating osteoblasts and osteoclasts [71,72]. Moreover, rituximab has the benefit of abrogating joint destruction in RA by inhibiting osteoclastogenesis [73]. Rituximab treatment is reported to be associated with the suppression of synovial osteoclast precursors and RANKL expression and a decrease in the serum RANKL/OPG ratio [73].
In a clinical study, rituximab treatment was associated with a significant improvement in femoral BMD. The application of rituximab further reduced bone pain and prevented patients from developing osteoporosis or fractures. Additionally, there was a substantial increase in P1NP and bone-specific alkaline phosphatase (BAP) [74]. Future rigorous trials are needed to provide solid evidence of the association between clinical symptoms and the use of rituximab.

2.2.4. Tocilizumab

IL-6, a glycopeptide whose gene is located on chromosome 7, is involved in both T-cell and B-cell proliferation and differentiation [75]. Tocilizumab is a monoclonal antibody that binds to soluble and membrane-bound IL-6 receptors, promoting the inhibition of IL-6R signal transduction to inflammatory mediators [75,76]. IL-6 inhibitors also prevent bone damage and cartilage degeneration in RA patients [77,78]. Furthermore, IL-6 inhibition retards bone loss progression independently of its impact on disease activity [79]. In mouse models of collagen-induced and antigen-induced arthritis, IL-6 inhibition slowed down the progression of arthritis but did not ameliorate arthritis [80]. IL-6 inhibitor management also prevented mechanical hyperalgesia and suppressed calcitonin gene-related peptide (CGRP) expression in osteoporotic models [81].
In a 1-year prospective study, RA patients receiving tocilizumab exhibited a decrease in serum DKK-1 concentrations and an increase in bone formation markers without a significant change in BMD [82]. In contrast, tocilizumab increased total hip BMD with denosumab therapy for osteoporotic patients with RA [83]. Tocilizumab also stabilized BMD in a multicenter single-arm study [84] and increased the BMD of patients who had osteopenia at baseline [85]. In ACPA-positive patients, 2 years of tocilizumab treatment reduced bone resorption and increased femoral BMD [86].

2.2.5. Janus Kinase Inhibitors

Janus kinase (JAK) inhibitors are small molecules that inhibit the JAK family enzymes (i.e., JAK1, JAK2, JAK3, and tyrosine kinase 2) that have a crucial role in the cell signaling processes leading to the immune and inflammation responses observed in RA [87,88]. JAK inhibitors induced bone repair by altering gene expression and increasing the activity of osteoblasts, supporting the use of inhibitors as potential anabolics [89]. Baricitinib, a selective inhibitor of JAK1 and JAK2, has been reported to have osteoprotective effects, increasing mineralization in bone-forming cells [90,91]. Osteoclastogenesis was also said to be suppressed by baricitinib via reducing RANKL expression in osteoblasts [91]. Baricitinib (4 mg once daily) in patients with moderate to severe RA inhibited the progression of radiographic joint destruction and relieved clinical symptoms such as joint swelling and tenderness. In addition, the application of tofacitinib in RA has been shown to affect osteoclasts directly and to inhibit osteoclast differentiation and proliferation [92]. Tofacitinib was also clinically effective in stabilizing BMD and lowering multiple bone markers such as P1NP and RANKL [93].

3. Spondyloarthritis

3.1. Bone Remodeling and Osteoporosis in Spondyloarthritis

Pathological bone formation is one of the most iconic hallmarks of spondyloarthritis (SpA), including axial-SpA and psoriatic arthritis (PsA) [94]. In patients suffering from long-term axial-SpA, bone formations that manifest as entheseal bone formation, periostitis, and spinal syndesmophytes are strongly correlated with the burden of the disease and resulting disability [94]. The adverse effects caused by bone losses in spondyloarthritis may impair the patients’ quality of life and activities of daily living [94]. Previous observational studies have evaluated the association between SpA and osteopenia or osteoporosis, with conflicting results [95]. A previous meta-analysis concluded that patients with psoriatic diseases have a greater risk of developing fragility fractures compared with controls [96]. However, this higher risk of fractures may not necessarily correlate with lower BMD or a higher risk of osteoporosis [96]. Another meta-analysis provided more in-depth evidence about the bone microstructure and bone strength in psoriatic disease [97]. Psoriatic patients had lower volumetric BMD than non-psoriatic controls. On the other hand, osteoporosis is a frequent complication in patients with ankylosing spondylitis (AS) [94]. Low BMD, osteoporosis, and fractures are reported to be associated with AS [98]. Bone health has proven to be a crucial issue in patients with SpA [94,95]. The following paragraphs further describe the roles of distinct pathways and biologic therapeutics in bone metabolism.

3.1.1. Proinflammatory Cytokines

Proinflammatory cytokines (such as TNF-α and IL-17) are involved in the bone remodeling process of SpA [99]. Extensive experimental and clinical evidence has linked TNF-α to osteoclast development, but the role of osteoblast formation has remained somewhat controversial [100]. Surprisingly, the unique bone phenotype that occurs in PsA and AS coexists with both systemic bone destruction and new bone formation, which is likely to be the result of the actions of IL-23 and/or IL-17 on osteocytes [101,102]. IL-17, the prevailing inflammatory cytokine in many SpA patients, has been found to promote not only osteoclasts for bone resorption but also osteoblasts for bone formation [103,104]. IL-23 is another distinctive cytokine that is abundantly present in the affected skin or joints of PsA patients [103]. Generally, IL-23 is overexpressed in PsA, resulting in IL-22 upregulation and osteoblast-related gene induction. This process eventually contributes to both osteoblast expansion and enthesophyte formation [105,106].

3.1.2. Autoantibodies

As RANKL and OPG play a pivotal role in the formation of osteoporosis, studies on autoantibodies against OPG have provided further knowledge. In a recent cohort, anti-OPG antibodies have been isolated in SpA patients, and a correlation with low BMD values and fractures was found [107,108]. In patients with RA, OPG levels were elevated and independently associated with disease severity; that is, OPG levels were higher in severe RA than in mild RA [109]. Moreover, genetic analysis has suggested that the OPG SNP haplotype was associated with HLA-B27 negativity in AS patients [110].

3.1.3. The Wnt Signaling Pathway and Dkk-1

Dkk-1 is involved in the Wnt signaling pathway and also has evidence regarding bone metabolism in patients with SpA. In a study conducted by Rossini et al., Dkk-1 levels were found to correlate with low BMD and the prevalence of vertebral fractures among AS patients [111]. Nevertheless, Dkk-1 serum levels were inversely correlated with lumbar spine Z-score BMD, and higher serum levels of Dkk-1 were associated with a higher prevalence of one or more vertebral fractures. An association between Dkk-1 and PTH was observed, and higher levels of PTH and lower levels of Dkk-1 were also measured in AS patients. Future clinical and animal studies are warranted to investigate the Wnt signaling pathway and bone involvement in SpA.

3.2. Effects on Bone of Biological/Targeted Therapies in Spondyloarthritis

Apart from low back pain, patients with SpA may experience different degrees of bone pain or bone loss in their entire disease course. Evidence-based recommendations for the treatment of axial SpA and PsA were updated in 2019 [112,113]. Targeted therapies against TNF, IL-17, IL-23, and the downstream pathways appear to be of significant clinical meaning according to the promising therapeutic results. However, the exact underlying mechanism regarding the application of these biologics in the inflammatory process is not yet clear [114]. Whether these novel biologics alleviate bone symptoms in different clinical presentations of SpA patients has also not been elucidated. The effects of biologics in SpA on bone mineral density have been reported, but the existing literature has been far less than that of RA. The impact of biologic therapies on bone metabolism is illustrated in Figure 1.

3.2.1. Tumor Necrosis Factor Inhibitors

TNF inhibitors, such as adalimumab, certolizumab pegol, etanercept, golimumab, and infliximab, have been utilized in the management of SpA. Preclinical research had indicated that TNFα inhibitors could increase collagen synthesis in osteoblasts and inhibit osteoclast production [115]. In synovial fibroblasts, TNF inhibitors may also downregulate angiogenesis by activating the transcription factors and the NF-κB signal transduction pathway [116]. In patients with AS, the application of TNF-targeting therapies has been reported to slow radiographic progression and reduce disease activity in recent observational studies [117]. Lumbar and hip BMD have improved after using TNF inhibitors [118]. Furthermore, TNF inhibitors also decrease CRP and act inversely on Dkk-1 and SOST in patients with AS [47]. A recent meta-analysis concluded that TNF inhibitor use for more than 4 years was associated with delayed structural progression [119]. Joint tenderness may be alleviated by TNF inhibitors due to their effects on reducing synovial inflammation [116].
PsA-related bone diseases were better controlled by bDMARDs such as TNF inhibitors [120]. A post-hoc analysis of adalimumab in PsA revealed that the inhibition of radiographic destruction was greater than expected in the control rather than in clinical disease activity alone [121]. Another open-label study of adalimumab also showed similar results, namely that the improvement in PsA disease activity was not correlated with that of bone erosion [122]. Moreover, adalimumab improves enthesitis and may reduce joint aches as well as enhancing the range of motion in patients with PsA in magnetic resonance imaging [123]. By contrast, bone density measured by peripheral quantitative computed tomography did not show significant changes after treatment with TNF inhibitors [124].

3.2.2. IL-17 Inhibitors

IL-17A, a member of the IL-17 superfamily of cytokines, is known to be involved in the pathomechanisms of SpA manifestations in skin, joints, and entheses. As evident in synovial samples, IL-17A signaling pathways are related to the actions of natural killer cells, tissue-resident memory T-cells, and innate lymphoid cells [125]. Secukinumab, ixekizumab, and brodalumab are IL-17-targeting therapeutical options. Their clinical performance is favorable, and the adverse effects seem to be tolerable. Both short-term and long-term clinical studies have indicated that patients treated with IL-17A inhibitors might reduce synovial inflammation and the destruction in bony structures versus a placebo [126]. Compelling evidence to date has shown that secukinumab improves arthralgia, swelling, physical function, and quality of life in SpA patients [126]. The PSARTROS study demonstrated that secukinumab used in psoriatic patients for 24 weeks showed no progression of catabolic or anabolic joint alterations [127,128]. The functional strength remained stable with a reduction in disease activity as measured by Disease Activity in PSoriatic Arthritis [128]. Experimentally, IL-17A promoted local mesenchymal stem cells to differentiate into osteoblasts and also increased mineralization in AS [129]. In an animal model of pathogenic SpA, treatment with anti-IL-17A delayed the effect of bone loss and reduced pathological bone formation [130].

3.2.3. Janus Kinase Inhibitors

JAK inhibitors are novel biologics for patients with SpA and have been studied in several clinical trials. Nevertheless, data regarding the effect of JAK inhibitors on bone resorption are scarce in SpA. The rationale for using JAK inhibitors in SpA came from the inhibition of various signaling pathways [131]. As JAK-dependent cytokines are involved in the pathogenesis of SpA, including IFNγ, IL-7, IL-12, IL-15, IL-22, and IL-23, blockade of the JAK-STAT pathway may inhibit the cellular function of a broad range of innate and adaptive cell types in SpA [131,132]. JAK inhibitors have been found to significantly reduce the secretion of the proinflammatory mediators MCP-1 and IL-6 in ex vivo studies [132]. The effect of JAK inhibitors on reducing inflammation in arthritis is promising; however, the data on bone mineral density are limited. More evidence is necessary to elucidate the clinical implications of JAK inhibitors for patients with SpA.

3.2.4. IL-23 Inhibitors

IL-23, a proinflammatory cytokine associated with the production of IL-17, IL-22, and TNF, was found to play a vital role in both innate and adaptive immunity [133]. The potential pathophysiologic effects of IL-23 in chronic inflammatory diseases have been identified in animal studies [134]. Data from preclinical IL-23 knockout models demonstrated the importance of IL-23 in the pathogenesis of arthritis [135]. Moreover, evidence from genetic analyses has suggested an association between the IL-23/IL-17 pathway and AS [136]. However, these findings in laboratory settings did not generally translate into therapeutic effects in SpA patients [137]. Two IL-23 inhibitors, risankizumab (the p19IL-23 inhibitor) and ustekinumab (the p40IL-12/23 inhibitor), did not achieve the primary endpoint in AS trials [138,139].
The evidence on whether the net effect of IL-23 on bone turnover is conflicting [140]. With regard to the catabolic effects, IL-23 has been suggested to promote osteoclastogenesis in human cells independently of the osteoblasts or exogenous soluble RANKL. In experimental settings, IL-23 was found to induce the activation of precursor cells and the associated proteins in RANKL-mediated osteoclastic differentiation [141,142]. On the other hand, genetic data have also reported potential anabolic effects in IL-23-related pathways. Genetic analysis from mice has suggested protective effects on bone. IL-23 can also induce the production of granulocyte-macrophage colony-stimulating factor (GM-CSF), an inhibitor of osteoclast differentiation [143]. However, this effect is mediated by TH17 cells and the subsequent production of IL-17, suggesting a net catabolic effect on bone metabolism towards IL-23. Trials of ustekinumab in patients with PsA, which demonstrated inhibition of the progression of bone destruction [144], provided clinical evidence of this debate. Research on the associations of IL-23 with other proinflammatory cytokines and mesenchymal cells is ongoing to understand their effects on bone metabolism better.

3.2.5. PDE4 Inhibitors

Phosphodiesterase 4 (PDE4) is an enzyme in the process of cyclic adenosine monophosphate (cAMP) degradation, which involves numerous biologic responses in human cells [145]. Previous literature has suggested the pathogenic role of cAMP inhibition on diseases such as chronic obstructive pulmonary disorder, inflammatory bowel disease, and PsA [146]. As PDE4 is involved in these inflammatory processes, inhibition of PDE4 may provide profound anti-inflammatory properties.
Apremilast, an oral PDE4 inhibitor, was shown to have a good safety profile, and its clinical implications have been investigated in recent trials. Apremilast blocks several pathways involving proinflammatory cytokines and chemokines, such as TNF-α, IL-23, CXCL9, and CXCL10 [145]. In contrast to the biologics mentioned above, which neutralize proinflammatory factors at the protein level, apremilast modulates these factors at the mRNA level [147].
PDE4 inhibitors may be associated with the inhibition of osteoclastogenesis in inflammatory arthritis. An ex vivo study using synovial fluid mononuclear cells of PsA patients suggested the inhibitory role of apremilast on IL-12/IL-23p40 [145]. Apremilast could also inhibit osteoclast fusion molecules such as dendritic cell-specific transmembrane protein and osteoclast-specific transmembrane protein, preventing osteoclastogenesis [146]. Studies on another PDE4 inhibitor, rolipram, have shown a potential blocking effect on PTH-induced osteoclast formation via the inhibition of calcitonin [147]. Additionally, the effect of apremilast on IL17A-mediated osteoclastogenesis has also been indicated by experimental studies using human peripheral blood mononuclear cells [147]. More in-depth and clinical evidence may improve our knowledge of the protective effects of PDE4 inhibitors on bone destruction in inflammatory arthritis.

4. Conclusions

In this narrative review, we have discussed the recent understanding of the pathogenic effects on bone in RA and SpA. The crosstalk between inflammation and bone metabolism is complicated. Proinflammatory cytokines, autoantibodies, and multiple signaling pathways play essential roles in bone destruction in RA and SpA patients. Current evidence has suggested the effect of novel therapeutics for improving clinical symptoms as well as enhancing the quality of life and daily activities of patients with RA and SpA. Biological and targeted therapies have provided alternative options for managing RA and SpA. However, the effects of these therapies on bone are not fully understood. As the positive effects on bone mineral density and bone metabolism of inflammatory arthritis are reviewed here, more research is warranted on the underlying mechanisms and their clinical impact. Future research is encouraged to demonstrate how these biological and targeted therapies best mitigate bone destruction in chronic inflammatory arthritis.

Author Contributions

Conceptualization, T.-L.C. and K.-Y.S.; literature review, T.-L.C., K.-H.C. and K.-Y.S.; writing—original draft preparation, T.-L.C. and K.-H.C.; writing—review and editing, T.-L.C. and K.-H.C.; supervision, K.-Y.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Ethical review and approval were waived for this study because this study is a narrative review that compiled the existing evidence.

Informed Consent Statement

Patient consent was waived because this review presented data that were already publicly online.

Data Availability Statement

This review presented data that were online for the public.

Conflicts of Interest

K.-Y.S. has received speaking fees and/or honoraria from Novartis, Bristol Myers Squibb, Eisai, Chugai, AbbVie, UCB, and Pfizer.

References

  1. Smith, E.; Hoy, D.G.; Cross, M.; Vos, T.; Naghavi, M.; Buchbinder, R.; Woolf, A.D.; March, L. The global burden of other musculoskeletal disorders: Estimates from the Global Burden of Disease 2010 study. Ann. Rheum. Dis. 2014, 73, 1462–1469. [Google Scholar] [CrossRef] [PubMed]
  2. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 388, 2023–2038. [Google Scholar] [CrossRef]
  3. Sharip, A.; Kunz, J. Understanding the Pathogenesis of Spondyloarthritis. Biomolecules 2020, 10, 1461. [Google Scholar] [CrossRef]
  4. Adami, G.; Fassio, A.; Rossini, M.; Caimmi, C.; Giollo, A.; Orsolini, G.; Viapiana, O.; Gatti, D. Osteoporosis in Rheumatic Diseases. Int. J. Mol. Sci. 2019, 20, 5867. [Google Scholar] [CrossRef] [Green Version]
  5. Rotta, D.; Fassio, A.; Rossini, M.; Giollo, A.; Viapiana, O.; Orsolini, G.; Bertoldo, E.; Gatti, D.; Adami, G. Osteoporosis in Inflammatory Arthritides: New Perspective on Pathogenesis and Treatment. Front. Med. 2020, 7, 613720. [Google Scholar] [CrossRef]
  6. Aletaha, D.; Smolen, J.S. Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA 2018, 320, 1360–1372. [Google Scholar] [CrossRef]
  7. McInnes, I.B.; Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011, 365, 2205–2219. [Google Scholar] [CrossRef] [Green Version]
  8. Orange, D.E.; Yao, V.; Sawicka, K.; Fak, J.; Frank, M.O.; Parveen, S.; Blachere, N.E.; Hale, C.; Zhang, F.; Raychaudhuri, S.; et al. RNA Identification of PRIME Cells Predicting Rheumatoid Arthritis Flares. N. Engl. J. Med. 2020, 383, 218–228. [Google Scholar] [CrossRef]
  9. Harnden, K.; Pease, C.; Jackson, A. Rheumatoid arthritis. BMJ 2016, 352, i387. [Google Scholar] [CrossRef]
  10. Klose-Jensen, R.; Tse, J.J.; Keller, K.K.; Barnabe, C.; Burghardt, A.J.; Finzel, S.; Tam, L.S.; Hauge, E.M.; Stok, K.S.; Manske, S.L. High-Resolution Peripheral Quantitative Computed Tomography for Bone Evaluation in Inflammatory Rheumatic Disease. Front. Med. 2020, 7, 337. [Google Scholar] [CrossRef]
  11. Chen, Z.; Bozec, A.; Ramming, A.; Schett, G. Anti-inflammatory and immune-regulatory cytokines in rheumatoid arthritis. Nat. Rev. Rheumatol. 2019, 15, 9–17. [Google Scholar] [CrossRef]
  12. Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Amarasekara, D.S.; Yun, H.; Kim, S.; Lee, N.; Kim, H.; Rho, J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw. 2018, 18, e8. [Google Scholar] [CrossRef] [PubMed]
  14. Amarasekara, D.S.; Yu, J.; Rho, J. Bone Loss Triggered by the Cytokine Network in Inflammatory Autoimmune Diseases. J. Immunol. Res. 2015, 2015, 832127. [Google Scholar] [CrossRef] [PubMed]
  15. Nanke, Y.; Kobashigawa, T.; Yago, T.; Kawamoto, M.; Yamanaka, H.; Kotake, S. RANK Expression and Osteoclastogenesis in Human Monocytes in Peripheral Blood from Rheumatoid Arthritis Patients. Biomed. Res. Int. 2016, 2016, 4874195. [Google Scholar] [CrossRef]
  16. Komatsu, N.; Takayanagi, H. Immune-bone interplay in the structural damage in rheumatoid arthritis. Clin. Exp. Immunol. 2018, 194, 1–8. [Google Scholar] [CrossRef] [Green Version]
  17. Li, S.; Yu, Y.; Yue, Y.; Liao, H.; Xie, W.; Thai, J.; Mikuls, T.R.; Thiele, G.M.; Duryee, M.J.; Sayles, H.; et al. Autoantibodies from Single Circulating Plasmablasts React with Citrullinated Antigens and Porphyromonas gingivalis in Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 614–626. [Google Scholar] [CrossRef] [Green Version]
  18. Harre, U.; Georgess, D.; Bang, H.; Bozec, A.; Axmann, R.; Ossipova, E.; Jakobsson, P.J.; Baum, W.; Nimmerjahn, F.; Szarka, E.; et al. Induction of osteoclastogenesis and bone loss by human autoantibodies against citrullinated vimentin. J. Clin. Investig. 2012, 122, 1791–1802. [Google Scholar] [CrossRef]
  19. Kurowska, W.; Slowinska, I.; Krogulec, Z.; Syrowka, P.; Maslinski, W. Antibodies to Citrullinated Proteins (ACPA) Associate with Markers of Osteoclast Activation and Bone Destruction in the Bone Marrow of Patients with Rheumatoid Arthritis. J. Clin. Med. 2021, 10, 1778. [Google Scholar] [CrossRef]
  20. Krishnamurthy, A.; Ytterberg, A.J.; Sun, M.; Sakuraba, K.; Steen, J.; Joshua, V.; Tarasova, N.K.; Malmström, V.; Wähämaa, H.; Réthi, B.; et al. Citrullination Controls Dendritic Cell Transdifferentiation into Osteoclasts. J. Immunol. 2019, 202, 3143–3150. [Google Scholar] [CrossRef] [Green Version]
  21. Rivollier, A.; Mazzorana, M.; Tebib, J.; Piperno, M.; Aitsiselmi, T.; Rabourdin-Combe, C.; Jurdic, P.; Servet-Delprat, C. Immature dendritic cell transdifferentiation into osteoclasts: A novel pathway sustained by the rheumatoid arthritis microenvironment. Blood 2004, 104, 4029–4037. [Google Scholar] [CrossRef] [PubMed]
  22. Krishnamurthy, A.; Joshua, V.; Haj Hensvold, A.; Jin, T.; Sun, M.; Vivar, N.; Ytterberg, A.J.; Engström, M.; Fernandes-Cerqueira, C.; Amara, K.; et al. Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann. Rheum. Dis. 2016, 75, 721–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Catrina, A.; Krishnamurthy, A.; Rethi, B. Current view on the pathogenic role of anti-citrullinated protein antibodies in rheumatoid arthritis. RMD Open 2021, 7, e001228. [Google Scholar] [CrossRef]
  24. Fert-Bober, J.; Darrah, E.; Andrade, F. Insights into the study and origin of the citrullinome in rheumatoid arthritis. Immunol. Rev. 2020, 294, 133–147. [Google Scholar] [CrossRef] [PubMed]
  25. Wigerblad, G.; Bas, D.B.; Fernades-Cerqueira, C.; Krishnamurthy, A.; Nandakumar, K.S.; Rogoz, K.; Kato, J.; Sandor, K.; Su, J.; Jimenez-Andrade, J.M.; et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann. Rheum. Dis. 2016, 75, 730–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kleyer, A.; Finzel, S.; Rech, J.; Manger, B.; Krieter, M.; Faustini, F.; Araujo, E.; Hueber, A.J.; Harre, U.; Engelke, K.; et al. Bone loss before the clinical onset of rheumatoid arthritis in subjects with anticitrullinated protein antibodies. Ann. Rheum. Dis. 2014, 73, 854–860. [Google Scholar] [CrossRef] [Green Version]
  27. Kobayashi, Y.; Uehara, S.; Udagawa, N.; Takahashi, N. Regulation of bone metabolism by Wnt signals. J. Biochem. 2016, 159, 387–392. [Google Scholar] [CrossRef]
  28. Spencer, G.J.; Utting, J.C.; Etheridge, S.L.; Arnett, T.R.; Genever, P.G. Wnt signalling in osteoblasts regulates expression of the receptor activator of NFkappaB ligand and inhibits osteoclastogenesis in vitro. J. Cell Sci. 2006, 119, 1283–1296. [Google Scholar] [CrossRef] [Green Version]
  29. Kumar, A.; Bala, I.; Bhukal, I.; Singh, H. Spinal anaesthesia with lidocaine 2% for caesarean section. Can. J. Anaesth. 1992, 39, 915–919. [Google Scholar] [CrossRef] [Green Version]
  30. Weivoda, M.M.; Ruan, M.; Hachfeld, C.M.; Pederson, L.; Howe, A.; Davey, R.A.; Zajac, J.D.; Kobayashi, Y.; Williams, B.O.; Westendorf, J.J.; et al. Wnt Signaling Inhibits Osteoclast Differentiation by Activating Canonical and Noncanonical cAMP/PKA Pathways. J. Bone Miner. Res. 2016, 31, 65–75. [Google Scholar] [CrossRef] [Green Version]
  31. Kobayashi, Y.; Uehara, S.; Koide, M.; Takahashi, N. The regulation of osteoclast differentiation by Wnt signals. Bonekey Rep. 2015, 4, 713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kovács, B.; Vajda, E.; Nagy, E.E. Regulatory Effects and Interactions of the Wnt and OPG-RANKL-RANK Signaling at the Bone-Cartilage Interface in Osteoarthritis. Int. J. Mol. Sci. 2019, 20, 4653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kobayashi, Y.; Maeda, K.; Takahashi, N. Roles of Wnt signaling in bone formation and resorption. Jpn. Dent. Sci. Rev. 2008, 44, 76–82. [Google Scholar] [CrossRef] [Green Version]
  34. Pinzone, J.J.; Hall, B.M.; Thudi, N.K.; Vonau, M.; Qiang, Y.W.; Rosol, T.J.; Shaughnessy, J.D., Jr. The role of Dickkopf-1 in bone development, homeostasis, and disease. Blood 2009, 113, 517–525. [Google Scholar] [CrossRef] [Green Version]
  35. Ma, Y.; Zhang, X.; Wang, M.; Xia, Q.; Yang, J.; Wu, M.; Han, R.; Chen, M.; Hu, X.; Yuan, Y.; et al. The serum level of Dickkopf-1 in patients with rheumatoid arthritis: A systematic review and meta-analysis. Int. Immunopharmacol. 2018, 59, 227–232. [Google Scholar] [CrossRef]
  36. Zheng, L.; Hu, F.; Bian, W.; Li, Y.; Zhang, L.; Shi, L.; Ma, X.; Liu, Y.; Zhang, X.; Li, Z. Dickkopf-1 perpetuated synovial fibroblast activation and synovial angiogenesis in rheumatoid arthritis. Clin. Rheumatol. 2021, 40, 4279–4288. [Google Scholar] [CrossRef]
  37. Miao, C.G.; Yang, Y.Y.; He, X.; Li, X.F.; Huang, C.; Huang, Y.; Zhang, L.; Lv, X.W.; Jin, Y.; Li, J. Wnt signaling pathway in rheumatoid arthritis, with special emphasis on the different roles in synovial inflammation and bone remodeling. Cell Signal. 2013, 25, 2069–2078. [Google Scholar] [CrossRef]
  38. Juarez, M.; McGettrick, H.M.; Scheel-Toellner, D.; Yeo, L.; Spengler, J.; de Paz, B.; Hardy, R.; Cooper, M.; Raza, K.; Buckley, C.D.; et al. DKK1 expression by synovial fibroblasts in very early rheumatoid arthritis associates with lymphocyte adhesion in an in vitro flow co-culture system. Arthritis Res. Ther. 2016, 18, 14. [Google Scholar] [CrossRef] [Green Version]
  39. Cici, D.; Corrado, A.; Rotondo, C.; Cantatore, F.P. Wnt Signaling and Biological Therapy in Rheumatoid Arthritis and Spondyloarthritis. Int. J. Mol. Sci. 2019, 20, 5552. [Google Scholar] [CrossRef] [Green Version]
  40. Smolen, J.S.; Landewé, R.B.M.; Bijlsma, J.W.J.; Burmester, G.R.; Dougados, M.; Kerschbaumer, A.; McInnes, I.B.; Sepriano, A.; van Vollenhoven, R.F.; de Wit, M.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis. 2020, 79, 685–699. [Google Scholar] [CrossRef] [Green Version]
  41. Barnabe, C.; Hanley, D.A. Effect of tumor necrosis factor alpha inhibition on bone density and turnover markers in patients with rheumatoid arthritis and spondyloarthropathy. Semin. Arthritis Rheum. 2009, 39, 116–122. [Google Scholar] [CrossRef] [PubMed]
  42. Sakthiswary, R.; Das, S. The effects of TNF α antagonist therapy on bone metabolism in rheumatoid arthritis: A systematic review. Curr. Drug Targets 2013, 14, 1552–1557. [Google Scholar] [CrossRef] [PubMed]
  43. Zwerina, J.; Tuerk, B.; Redlich, K.; Smolen, J.S.; Schett, G. Imbalance of local bone metabolism in inflammatory arthritis and its reversal upon tumor necrosis factor blockade: Direct analysis of bone turnover in murine arthritis. Arthritis Res. Ther. 2006, 8, R22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Thudium, C.S.; Nielsen, S.H.; Sardar, S.; Mobasheri, A.; van Spil, W.E.; Lories, R.; Henriksen, K.; Bay-Jensen, A.C.; Karsdal, M.A. Bone phenotypes in rheumatology—There is more to bone than just bone. BMC Musculoskelet Disord 2020, 21, 789. [Google Scholar] [CrossRef] [PubMed]
  45. Chopin, F.; Garnero, P.; le Henanff, A.; Debiais, F.; Daragon, A.; Roux, C.; Sany, J.; Wendling, D.; Zarnitsky, C.; Ravaud, P.; et al. Long-term effects of infliximab on bone and cartilage turnover markers in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2008, 67, 353–357. [Google Scholar] [CrossRef] [PubMed]
  46. Fassio, A.; Adami, G.; Gatti, D.; Orsolini, G.; Giollo, A.; Idolazzi, L.; Benini, C.; Vantaggiato, E.; Rossini, M.; Viapiana, O. Inhibition of tumor necrosis factor-alpha (TNF-alpha) in patients with early rheumatoid arthritis results in acute changes of bone modulators. Int. Immunopharmacol. 2019, 67, 487–489. [Google Scholar] [CrossRef]
  47. Chisari, E.; Yaghmour, K.M.; Khan, W.S. The effects of TNF-alpha inhibition on cartilage: A systematic review of preclinical studies. Osteoarthr. Cartil. 2020, 28, 708–718. [Google Scholar] [CrossRef]
  48. Ørnbjerg, L.M.; Østergaard, M.; Jensen, T.; Hørslev-Petersen, K.; Stengaard-Pedersen, K.; Junker, P.; Ellingsen, T.; Ahlquist, P.; Lindegaard, H.; Linauskas, A.; et al. Hand bone loss in early rheumatoid arthritis during a methotrexate-based treat-to-target strategy with or without adalimumab-a substudy of the optimized treatment algorithm in early RA (OPERA) trial. Clin. Rheumatol. 2017, 36, 781–789. [Google Scholar] [CrossRef]
  49. Hoff, M.; Kvien, T.K.; Kälvesten, J.; Elden, A.; Kavanaugh, A.; Haugeberg, G. Adalimumab reduces hand bone loss in rheumatoid arthritis independent of clinical response: Subanalysis of the PREMIER study. BMC Musculoskelet Disord 2011, 12, 54. [Google Scholar] [CrossRef] [Green Version]
  50. Sole, K. Infliximab increases bone mineral density in patients with rheumatoid arthritis. Nat. Clin. Pract. Rheumatol. 2006, 2, 120. [Google Scholar] [CrossRef]
  51. Poutoglidou, F.; Pourzitaki, C.; Manthou, M.E.; Samoladas, E.; Saitis, A.; Malliou, F.; Kouvelas, D. Infliximab prevents systemic bone loss and suppresses tendon inflammation in a collagen-induced arthritis rat model. Inflammopharmacology 2021, 29, 661–672. [Google Scholar] [CrossRef] [PubMed]
  52. Vis, M.; Havaardsholm, E.A.; Haugeberg, G.; Uhlig, T.; Voskuyl, A.E.; van de Stadt, R.J.; Dijkmans, B.A.; Woolf, A.D.; Kvien, T.K.; Lems, W.F. Evaluation of bone mineral density, bone metabolism, osteoprotegerin and receptor activator of the NFkappaB ligand serum levels during treatment with infliximab in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2006, 65, 1495–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Poutoglidou, F.; Pourzitaki, C.; Manthou, M.E.; Samoladas, E.; Malliou, F.; Saitis, A.; Kouvelas, D. Effects of Long-Term Methotrexate, Infliximab, and Tocilizumab Administration on Bone Microarchitecture and Tendon Morphology in Healthy Wistar Rats. Cureus 2021, 13, e14696. [Google Scholar] [CrossRef]
  54. Adami, G.; Orsolini, G.; Adami, S.; Viapiana, O.; Idolazzi, L.; Gatti, D.; Rossini, M. Effects of TNF Inhibitors on Parathyroid Hormone and Wnt Signaling Antagonists in Rheumatoid Arthritis. Calcif. Tissue Int. 2016, 99, 360–364. [Google Scholar] [CrossRef] [PubMed]
  55. Orsolini, G.; Adami, G.; Adami, S.; Viapiana, O.; Idolazzi, L.; Gatti, D.; Rossini, M. Short-Term Effects of TNF Inhibitors on Bone Turnover Markers and Bone Mineral Density in Rheumatoid Arthritis. Calcif. Tissue Int. 2016, 98, 580–585. [Google Scholar] [CrossRef]
  56. Ozen, G.; Pedro, S.; Wolfe, F.; Michaud, K. Medications associated with fracture risk in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2019, 78, 1041–1047. [Google Scholar] [CrossRef]
  57. Shin, A.; Park, E.H.; Dong, Y.H.; Ha, Y.J.; Lee, Y.J.; Lee, E.B.; Song, Y.W.; Kang, E.H. Comparative risk of osteoporotic fracture among patients with rheumatoid arthritis receiving TNF inhibitors versus other biologics: A cohort study. Osteoporos. Int. 2020, 31, 2131–2139. [Google Scholar] [CrossRef]
  58. Alenazy, M.F.; Saheb Sharif-Askari, F.; Omair, M.A.; El-Wetidy, M.S.; Omair, M.A.; Mitwalli, H.; Al-Muhsen, S.; Al-Masri, A.; Hamid, Q.; Halwani, R. Abatacept enhances blood regulatory B cells of rheumatoid arthritis patients to a level that associates with disease remittance. Sci. Rep. 2021, 11, 5629. [Google Scholar] [CrossRef]
  59. Genovese, M.C.; Becker, J.C.; Schiff, M.; Luggen, M.; Sherrer, Y.; Kremer, J.; Birbara, C.; Box, J.; Natarajan, K.; Nuamah, I.; et al. abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N. Engl. J. Med. 2005, 353, 1114–1123. [Google Scholar] [CrossRef] [Green Version]
  60. Cutolo, M.; Nadler, S.G. Advances in CTLA-4-Ig-mediated modulation of inflammatory cell and immune response activation in rheumatoid arthritis. Autoimmun. Rev. 2013, 12, 758–767. [Google Scholar] [CrossRef]
  61. Okada, H.; Kajiya, H.; Omata, Y.; Matsumoto, T.; Sato, Y.; Kobayashi, T.; Nakamura, S.; Kaneko, Y.; Nakamura, S.; Koyama, T.; et al. CTLA4-Ig Directly Inhibits Osteoclastogenesis by Interfering With Intracellular Calcium Oscillations in Bone Marrow Macrophages. J. Bone Miner. Res. 2019, 34, 1744–1752. [Google Scholar] [CrossRef]
  62. Roser-Page, S.; Vikulina, T.; Zayzafoon, M.; Weitzmann, M.N. CTLA-4Ig-induced T cell anergy promotes Wnt-10b production and bone formation in a mouse model. Arthritis Rheumatol. 2014, 66, 990–999. [Google Scholar] [CrossRef] [Green Version]
  63. Nagao, N.; Wakabayashi, H.; Miyamura, G.; Kato, S.; Naito, Y.; Sudo, A. CTLA-4Ig Improves Hyperalgesia in a Mouse Model of Osteoporosis. Int. J. Mol. Sci. 2020, 21, 9479. [Google Scholar] [CrossRef]
  64. Kawashiri, S.Y.; Endo, Y.; Nishino, A.; Okamoto, M.; Tsuji, S.; Takatani, A.; Shimizu, T.; Sumiyoshi, R.; Koga, T.; Iwamoto, N.; et al. Association between serum bone biomarker levels and therapeutic response to abatacept in patients with rheumatoid arthritis (RA): A multicenter, prospective, and observational RA ultrasound cohort study in Japan. BMC Musculoskelet Disord. 2021, 22, 506. [Google Scholar] [CrossRef]
  65. Tada, M.; Inui, K.; Sugioka, Y.; Mamoto, K.; Okano, T.; Koike, T. Abatacept might increase bone mineral density at femoral neck for patients with rheumatoid arthritis in clinical practice: AIRTIGHT study. Rheumatol. Int. 2018, 38, 777–784. [Google Scholar] [CrossRef] [Green Version]
  66. Tokumoto, H.; Tominaga, H.; Arishima, Y.; Jokoji, G.; Akimoto, M.; Ohtsubo, H.; Taketomi, E.; Sunahara, N.; Nagano, S.; Ishidou, Y.; et al. Association between Bone Mineral Density of Femoral Neck and Geriatric Nutritional Risk Index in Rheumatoid Arthritis Patients Treated with Biological Disease-Modifying Anti-Rheumatic Drugs. Nutrients 2018, 10, 234. [Google Scholar] [CrossRef] [Green Version]
  67. Harrold, L.R.; Bryson, J.; Lehman, T.; Zhuo, J.; Gao, S.; Han, X.; Schrader, A.; Rebello, S.; Pappas, D.A.; Sommers, T.; et al. Association Between Baseline Anti-cyclic Citrullinated Peptide Antibodies and 6-Month Clinical Response Following Abatacept or TNF Inhibitor Treatment: A Real-World Analysis of Biologic-Experienced Patients with RA. Rheumatol. Ther. 2021, 8, 937–953. [Google Scholar] [CrossRef]
  68. Kerschbaumer, A.; Sepriano, A.; Smolen, J.S.; van der Heijde, D.; Dougados, M.; van Vollenhoven, R.; McInnes, I.B.; Bijlsma, J.W.J.; Burmester, G.R.; de Wit, M.; et al. Efficacy of pharmacological treatment in rheumatoid arthritis: A systematic literature research informing the 2019 update of the EULAR recommendations for management of rheumatoid arthritis. Ann. Rheum. Dis. 2020, 79, 744–759. [Google Scholar] [CrossRef] [Green Version]
  69. Humby, F.; Durez, P.; Buch, M.H.; Lewis, M.J.; Rizvi, H.; Rivellese, F.; Nerviani, A.; Giorli, G.; Mahto, A.; Montecucco, C.; et al. rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, phase 4 randomised controlled trial. Lancet 2021, 397, 305–317. [Google Scholar] [CrossRef]
  70. Kolomansky, A.; Kaye, I.; Ben-Califa, N.; Gorodov, A.; Awida, Z.; Sadovnic, O.; Ibrahim, M.; Liron, T.; Hiram-Bab, S.; Oster, H.S.; et al. Anti-CD20-Mediated B Cell Depletion Is Associated With Bone Preservation in Lymphoma Patients and Bone Mass Increase in Mice. Front. Immunol. 2020, 11, 561294. [Google Scholar] [CrossRef]
  71. Sun, W.; Meednu, N.; Rosenberg, A.; Rangel-Moreno, J.; Wang, V.; Glanzman, J.; Owen, T.; Zhou, X.; Zhang, H.; Boyce, B.F.; et al. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat. Commun. 2018, 9, 5127. [Google Scholar] [CrossRef] [PubMed]
  72. Kitaura, H.; Marahleh, A.; Ohori, F.; Noguchi, T.; Shen, W.R.; Qi, J.; Nara, Y.; Pramusita, A.; Kinjo, R.; Mizoguchi, I. Osteocyte-Related Cytokines Regulate Osteoclast Formation and Bone Resorption. Int. J. Mol. Sci. 2020, 21, 5169. [Google Scholar] [CrossRef] [PubMed]
  73. Boumans, M.J.; Thurlings, R.M.; Yeo, L.; Scheel-Toellner, D.; Vos, K.; Gerlag, D.M.; Tak, P.P. Rituximab abrogates joint destruction in rheumatoid arthritis by inhibiting osteoclastogenesis. Ann. Rheum. Dis. 2012, 71, 108–113. [Google Scholar] [CrossRef]
  74. Wheater, G.; Elshahaly, M.; Naraghi, K.; Tuck, S.P.; Datta, H.K.; van Laar, J.M. Changes in bone density and bone turnover in patients with rheumatoid arthritis treated with rituximab, results from an exploratory, prospective study. PLoS ONE 2018, 13, e0201527. [Google Scholar] [CrossRef]
  75. Singh, J.A.; Beg, S.; Lopez-Olivo, M.A. Tocilizumab for rheumatoid arthritis. Cochrane Database Syst. Rev. 2010, 7, Cd008331. [Google Scholar] [CrossRef]
  76. Bijlsma, J.W.J.; Welsing, P.M.J.; Woodworth, T.G.; Middelink, L.M.; Pethö-Schramm, A.; Bernasconi, C.; Borm, M.E.A.; Wortel, C.H.; Ter Borg, E.J.; Jahangier, Z.N.; et al. Early rheumatoid arthritis treated with tocilizumab, methotrexate, or their combination (U-Act-Early): A multicentre, randomised, double-blind, double-dummy, strategy trial. Lancet 2016, 388, 343–355. [Google Scholar] [CrossRef]
  77. Murakami, M.; Nishimoto, N. IL-6 inhibitors prevent bone loss and cartilage degeneration in rheumatoid arthritis. Clin. Calcium 2015, 25, 1851–1857. [Google Scholar]
  78. Yip, R.M.L.; Yim, C.W. Role of Interleukin 6 Inhibitors in the Management of Rheumatoid Arthritis. J. Clin. Rheumatol. 2019, 27, e516. [Google Scholar] [CrossRef]
  79. Smolen, J.S.; Avila, J.C.; Aletaha, D. Tocilizumab inhibits progression of joint damage in rheumatoid arthritis irrespective of its anti-inflammatory effects: Disassociation of the link between inflammation and destruction. Ann. Rheum. Dis. 2012, 71, 687–693. [Google Scholar] [CrossRef] [Green Version]
  80. Choy, E.H.; De Benedetti, F.; Takeuchi, T.; Hashizume, M.; John, M.R.; Kishimoto, T. Translating IL-6 biology into effective treatments. Nat. Rev. Rheumatol. 2020, 16, 335–345. [Google Scholar] [CrossRef] [Green Version]
  81. Wakabayashi, H.; Miyamura, G.; Nagao, N.; Kato, S.; Naito, Y.; Sudo, A. Functional Block of Interleukin-6 Reduces a Bone Pain Marker but Not Bone Loss in Hindlimb-Unloaded Mice. Int. J. Mol. Sci. 2020, 21, 3521. [Google Scholar] [CrossRef]
  82. Briot, K.; Rouanet, S.; Schaeverbeke, T.; Etchepare, F.; Gaudin, P.; Perdriger, A.; Vray, M.; Steinberg, G.; Roux, C. The effect of tocilizumab on bone mineral density, serum levels of Dickkopf-1 and bone remodeling markers in patients with rheumatoid arthritis. Jt. Bone Spine 2015, 82, 109–115. [Google Scholar] [CrossRef]
  83. Suzuki, T.; Nakamura, Y.; Kato, H. Effects of denosumab on bone metabolism and bone mineral density with anti-TNF inhibitors, tocilizumab, or abatacept in osteoporosis with rheumatoid arthritis. Ther. Clin. Risk Manag. 2018, 14, 453–459. [Google Scholar] [CrossRef] [Green Version]
  84. Abu-Shakra, M.; Zisman, D.; Balbir-Gurman, A.; Amital, H.; Levy, Y.; Langevitz, P.; Tishler, M.; Molad, Y.; Aamar, S.; Roser, I.; et al. Effect of Tocilizumab on Fatigue and Bone Mineral Density in Patients with Rheumatoid Arthritis. Isr. Med. Assoc. J. 2018, 20, 239–244. [Google Scholar]
  85. Kume, K.; Amano, K.; Yamada, S.; Kanazawa, T.; Ohta, H.; Hatta, K.; Amano, K.; Kuwaba, N. The effect of tocilizumab on bone mineral density in patients with methotrexate-resistant active rheumatoid arthritis. Rheumatology 2014, 53, 900–903. [Google Scholar] [CrossRef] [Green Version]
  86. Chen, Y.M.; Chen, H.H.; Huang, W.N.; Liao, T.L.; Chen, J.P.; Chao, W.C.; Lin, C.T.; Hung, W.T.; Hsieh, C.W.; Hsieh, T.Y.; et al. tocilizumab potentially prevents bone loss in patients with anticitrullinated protein antibody-positive rheumatoid arthritis. PLoS ONE 2017, 12, e0188454. [Google Scholar] [CrossRef]
  87. Schwartz, D.M.; Kanno, Y.; Villarino, A.; Ward, M.; Gadina, M.; O’Shea, J.J. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 2017, 17, 78. [Google Scholar] [CrossRef] [Green Version]
  88. Pope, J.; Sawant, R.; Tundia, N.; Du, E.X.; Qi, C.Z.; Song, Y.; Tang, P.; Betts, K.A. Comparative Efficacy of JAK Inhibitors for Moderate-To-Severe Rheumatoid Arthritis: A Network Meta-Analysis. Adv. Ther. 2020, 37, 2356–2372. [Google Scholar] [CrossRef] [Green Version]
  89. Adam, S.; Simon, N.; Steffen, U.; Andes, F.T.; Scholtysek, C.; Müller, D.I.H.; Weidner, D.; Andreev, D.; Kleyer, A.; Culemann, S.; et al. JAK inhibition increases bone mass in steady-state conditions and ameliorates pathological bone loss by stimulating osteoblast function. Sci. Transl. Med. 2020, 12, eaay4447. [Google Scholar] [CrossRef]
  90. Emery, P.; Durez, P.; Hueber, A.J.; de la Torre, I.; Larsson, E.; Holzkämper, T.; Tanaka, Y. Baricitinib inhibits structural joint damage progression in patients with rheumatoid arthritis—A comprehensive review. Arthritis Res. Ther. 2021, 23, 3. [Google Scholar] [CrossRef]
  91. van der Heijde, D.; Schiff, M.; Tanaka, Y.; Xie, L.; Meszaros, G.; Ishii, T.; Casillas, M.; Ortmann, R.A.; Emery, P. Low rates of radiographic progression of structural joint damage over 2 years of baricitinib treatment in patients with rheumatoid arthritis. RMD Open 2019, 5, e000898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Gaber, T.; Brinkman, A.C.K.; Pienczikowski, J.; Diesing, K.; Damerau, A.; Pfeiffenberger, M.; Lang, A.; Ohrndorf, S.; Burmester, G.R.; Buttgereit, F.; et al. Impact of Janus Kinase Inhibition with Tofacitinib on Fundamental Processes of Bone Healing. Int. J. Mol. Sci. 2020, 21, 865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hamar, A.; Szekanecz, Z.; Pusztai, A.; Czókolyová, M.; Végh, E.; Pethő, Z.; Bodnár, N.; Gulyás, K.; Horváth, Á.; Soós, B.; et al. Effects of one-year tofacitinib therapy on bone metabolism in rheumatoid arthritis. Osteoporos Int. 2021, 32, 1621–1629. [Google Scholar] [CrossRef] [PubMed]
  94. Lim, M.J.; Kang, K.Y. A Contemporary View of the Diagnosis of Osteoporosis in Patients With Axial Spondyloarthritis. Front. Med. 2020, 7, 569449. [Google Scholar] [CrossRef]
  95. Law, L.; Beckman Rehnman, J.; Deminger, A.; Klingberg, E.; Jacobsson, L.; Forsblad-d’Elia, H. Factors related to health-related quality of life in ankylosing spondylitis, overall and stratified by sex. Arthritis Res. Ther. 2018, 20, 284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Chen, T.L.; Lu, J.W.; Huang, Y.W.; Wang, J.H.; Su, K.Y. Bone Mineral Density, Osteoporosis, and Fracture Risk in Adult Patients with Psoriasis or Psoriatic Arthritis: A Systematic Review and Meta-Analysis of Observational Studies. J. Clin. Med. 2020, 9, 3712. [Google Scholar] [CrossRef]
  97. Huang, Y.W.; Lu, J.W.; Chen, T.L. Volumetric Bone Mineral Density Measured by HR-pQCT in Patients with Psoriasis or Psoriatic Arthritis: A Systematic Review and Meta-Analysis with Trial Sequential Analysis. Healthcare 2021, 9, 1056. [Google Scholar] [CrossRef]
  98. Hinze, A.M.; Louie, G.H. Osteoporosis Management in Ankylosing Spondylitis. Curr. Treat. Options Rheumatol. 2016, 2, 271–282. [Google Scholar] [CrossRef] [Green Version]
  99. Poddubnyy, D.; Sieper, J. Mechanism of New Bone Formation in Axial Spondyloarthritis. Curr. Rheumatol. Rep. 2017, 19, 55. [Google Scholar] [CrossRef]
  100. Briot, K.; Roux, C. Inflammation, bone loss and fracture risk in spondyloarthritis. RMD Open 2015, 1, e000052. [Google Scholar] [CrossRef]
  101. Taams, L.S.; Steel, K.J.A.; Srenathan, U.; Burns, L.A.; Kirkham, B.W. IL-17 in the immunopathogenesis of spondyloarthritis. Nat. Rev. Rheumatol. 2018, 14, 453–466. [Google Scholar] [CrossRef] [Green Version]
  102. Yeremenko, N.; Paramarta, J.E.; Baeten, D. The interleukin-23/interleukin-17 immune axis as a promising new target in the treatment of spondyloarthritis. Curr Opin Rheumatol. 2014, 26, 361–370. [Google Scholar] [CrossRef]
  103. Chisălău, B.A.; Crînguș, L.I.; Vreju, F.A.; Pârvănescu, C.D.; Firulescu, S.C.; Dinescu, Ș.C.; Ciobanu, D.A.; Tica, A.A.; Sandu, R.E.; Siloși, I.; et al. New insights into IL-17/IL-23 signaling in ankylosing spondylitis (Review). Exp. Ther. Med. 2020, 20, 3493–3497. [Google Scholar] [CrossRef]
  104. Groen, S.S.; Sinkeviciute, D.; Bay-Jensen, A.C.; Thudium, C.S.; Karsdal, M.A.; Thomsen, S.F.; Schett, G.; Nielsen, S.H. Exploring IL-17 in spondyloarthritis for development of novel treatments and biomarkers. Autoimmun. Rev. 2021, 20, 102760. [Google Scholar] [CrossRef]
  105. Vecellio, M.; Hake, V.X.; Davidson, C.; Carena, M.C.; Wordsworth, B.P.; Selmi, C. The IL-17/IL-23 Axis and Its Genetic Contribution to Psoriatic Arthritis. Front. Immunol. 2020, 11, 596086. [Google Scholar] [CrossRef]
  106. Boutet, M.A.; Nerviani, A.; Gallo Afflitto, G.; Pitzalis, C. Role of the IL-23/IL-17 Axis in Psoriasis and Psoriatic Arthritis: The Clinical Importance of Its Divergence in Skin and Joints. Int. J. Mol. Sci. 2018, 19, 530. [Google Scholar] [CrossRef] [Green Version]
  107. Hauser, B.; Zhao, S.; Visconti, M.R.; Riches, P.L.; Fraser, W.D.; Piec, I.; Goodson, N.J.; Ralston, S.H. Autoantibodies to Osteoprotegerin are Associated with Low Hip Bone Mineral Density and History of Fractures in Axial Spondyloarthritis: A Cross-Sectional Observational Study. Calcif. Tissue Int. 2017, 101, 375–383. [Google Scholar] [CrossRef] [Green Version]
  108. Clunie, G.; Horwood, N. Loss and gain of bone in spondyloarthritis: What drives these opposing clinical features? Ther. Adv. Musculoskelet Dis. 2020, 12, 1759720x20969260. [Google Scholar] [CrossRef]
  109. Titanji, K. Beyond Antibodies: B Cells and the OPG/RANK-RANKL Pathway in Health, Non-HIV Disease and HIV-Induced Bone Loss. Front. Immunol. 2017, 8, 1851. [Google Scholar] [CrossRef] [Green Version]
  110. Wang, C.M.; Tsai, S.C.; Lin, J.C.; Wu, Y.J.; Wu, J.; Chen, J.Y. Association of Genetic Variants of RANK, RANKL, and OPG with Ankylosing Spondylitis Clinical Features in Taiwanese. Mediat. Inflamm. 2019, 2019, 8029863. [Google Scholar] [CrossRef] [Green Version]
  111. Rossini, M.; Viapiana, O.; Idolazzi, L.; Ghellere, F.; Fracassi, E.; Troplini, S.; Povino, M.R.; Kunnathully, V.; Adami, S.; Gatti, D. Higher Level of Dickkopf-1 is Associated with Low Bone Mineral Density and Higher Prevalence of Vertebral Fractures in Patients with Ankylosing Spondylitis. Calcif. Tissue Int. 2016, 98, 438–445. [Google Scholar] [CrossRef] [PubMed]
  112. Ward, M.M.; Deodhar, A.; Gensler, L.S.; Dubreuil, M.; Yu, D.; Khan, M.A.; Haroon, N.; Borenstein, D.; Wang, R.; Biehl, A.; et al. 2019 Update of the American College of Rheumatology/Spondylitis Association of America/Spondyloarthritis Research and Treatment Network Recommendations for the Treatment of Ankylosing Spondylitis and Nonradiographic Axial Spondyloarthritis. Arthritis Rheumatol. 2019, 71, 1599–1613. [Google Scholar] [CrossRef] [PubMed]
  113. The Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) Treatment Recommendations 2021. Available online: https://www.grappanetwork.org/ (accessed on 20 December 2021).
  114. Sieper, J.; Braun, J.; Dougados, M.; Baeten, D. Axial spondyloarthritis. Nat. Rev. Dis. Primers 2015, 1, 15013. [Google Scholar] [CrossRef]
  115. Koo, B.S.; Oh, J.S.; Park, S.Y.; Shin, J.H.; Ahn, G.Y.; Lee, S.; Joo, K.B.; Kim, T.H. Tumour necrosis factor inhibitors slow radiographic progression in patients with ankylosing spondylitis: 18-year real-world evidence. Ann. Rheum. Dis. 2020, 79, 1327–1332. [Google Scholar] [CrossRef]
  116. Molnar, C.; Scherer, A.; Baraliakos, X.; de Hooge, M.; Micheroli, R.; Exer, P.; Kissling, R.O.; Tamborrini, G.; Wildi, L.M.; Nissen, M.J.; et al. TNF blockers inhibit spinal radiographic progression in ankylosing spondylitis by reducing disease activity: Results from the Swiss Clinical Quality Management cohort. Ann. Rheum. Dis. 2018, 77, 63–69. [Google Scholar] [CrossRef]
  117. Ajrawat, P.; Touma, Z.; Sari, I.; Taheri, C.; Diaz Martinez, J.P.; Haroon, N. Effect of TNF-inhibitor therapy on spinal structural progression in ankylosing spondylitis patients: A systematic review and meta-analysis. Int. J. Rheum. Dis. 2020, 23, 728–743. [Google Scholar] [CrossRef]
  118. Siu, S.; Haraoui, B.; Bissonnette, R.; Bessette, L.; Roubille, C.; Richer, V.; Starnino, T.; McCourt, C.; McFarlane, A.; Fleming, P.; et al. Meta-analysis of tumor necrosis factor inhibitors and glucocorticoids on bone density in rheumatoid arthritis and ankylosing spondylitis trials. Arthritis Care Res. 2015, 67, 754–764. [Google Scholar] [CrossRef] [Green Version]
  119. Landewé, R.; Ritchlin, C.T.; Aletaha, D.; Zhang, Y.; Ganz, F.; Hojnik, M.; Coates, L.C. Inhibition of radiographic progression in psoriatic arthritis by adalimumab independent of the control of clinical disease activity. Rheumatology 2019, 58, 1025–1033. [Google Scholar] [CrossRef] [Green Version]
  120. Simon, D.; Kleyer, A.; Bayat, S.; Tascilar, K.; Kampylafka, E.; Meinderink, T.; Schuster, L.; Petrov, R.; Liphardt, A.M.; Rech, J.; et al. Effect of disease-modifying anti-rheumatic drugs on bone structure and strength in psoriatic arthritis patients. Arthritis Res. Ther. 2019, 21, 162. [Google Scholar] [CrossRef] [Green Version]
  121. Poggenborg, R.P.; Wiell, C.; Bøyesen, P.; Boonen, A.; Bird, P.; Pedersen, S.J.; Sørensen, I.J.; Madsen, O.R.; Slot, O.; Møller, J.M.; et al. No overall damage progression despite persistent inflammation in adalimumab-treated psoriatic arthritis patients: Results from an investigator-initiated 48-week comparative magnetic resonance imaging, computed tomography and radiography trial. Rheumatology 2014, 53, 746–756. [Google Scholar] [CrossRef] [Green Version]
  122. Yanaba, K.; Sadaoka, A.; Yonenaga, T.; Saeki, H.; Umezawa, Y.; Tojo, S.; Ito, T.; Kikuchi, S.; Fukuda, K.; Nakagawa, H. Adalimumab markedly improves enthesitis in patients with psoriatic arthritis: Evaluation with a magnetic resonance imaging scoring system. J. Dermatol. 2015, 42, 1153–1159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. McGonagle, D.G.; McInnes, I.B.; Kirkham, B.W.; Sherlock, J.; Moots, R. The role of IL-17A in axial spondyloarthritis and psoriatic arthritis: Recent advances and controversies. Ann. Rheum. Dis. 2019, 78, 1167–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Juhász, B.; Gulyás, K.; Horváth, Á.; Végh, E.; Pusztai, A.; Szentpétery, Á.; Pethő, Z.; Bodnár, N.; Hamar, A.; Bodoki, L.; et al. Peripheral quantitative computed tomography in the assessment of bone mineral density in anti-TNF-treated rheumatoid arthritis and ankylosing spondylitis patients. BMC Musculoskelet. Disord. 2021, 22, 817. [Google Scholar] [CrossRef] [PubMed]
  125. McInnes, I.B.; Behrens, F.; Mease, P.J.; Kavanaugh, A.; Ritchlin, C.; Nash, P.; Masmitja, J.G.; Goupille, P.; Korotaeva, T.; Gottlieb, A.B.; et al. Secukinumab versus adalimumab for treatment of active psoriatic arthritis (EXCEED): A double-blind, parallel-group, randomised, active-controlled, phase 3b trial. Lancet 2020, 395, 1496–1505. [Google Scholar] [CrossRef]
  126. Kampylafka, E.; d’Oliveira, I.; Linz, C.; Lerchen, V.; Stemmler, F.; Simon, D.; Englbrecht, M.; Sticherling, M.; Rech, J.; Kleyer, A.; et al. Resolution of synovitis and arrest of catabolic and anabolic bone changes in patients with psoriatic arthritis by IL-17A blockade with secukinumab: Results from the prospective PSARTROS study. Arthritis Res. Ther. 2018, 20, 153. [Google Scholar] [CrossRef] [Green Version]
  127. Kampylafka, E.; Simon, D.; d’Oliveira, I.; Linz, C.; Lerchen, V.; Englbrecht, M.; Rech, J.; Kleyer, A.; Sticherling, M.; Schett, G.; et al. Disease interception with interleukin-17 inhibition in high-risk psoriasis patients with subclinical joint inflammation-data from the prospective IVEPSA study. Arthritis Res. Ther. 2019, 21, 178. [Google Scholar] [CrossRef] [Green Version]
  128. Tang, M.; Lu, L.; Yu, X. Interleukin-17A Interweaves the Skeletal and Immune Systems. Front. Immunol. 2020, 11, 625034. [Google Scholar] [CrossRef]
  129. van Tok, M.N.; van Duivenvoorde, L.M.; Kramer, I.; Ingold, P.; Pfister, S.; Roth, L.; Blijdorp, I.C.; van de Sande, M.G.H.; Taurog, J.D.; Kolbinger, F.; et al. Interleukin-17A Inhibition Diminishes Inflammation and New Bone Formation in Experimental Spondyloarthritis. Arthritis Rheumatol. 2019, 71, 612–625. [Google Scholar] [CrossRef]
  130. Veale, D.J.; McGonagle, D.; McInnes, I.B.; Krueger, J.G.; Ritchlin, C.T.; Elewaut, D.; Kanik, K.S.; Hendrikx, T.; Berstein, G.; Hodge, J.; et al. The rationale for Janus kinase inhibitors for the treatment of spondyloarthritis. Rheumatology 2019, 58, 197–205. [Google Scholar] [CrossRef] [Green Version]
  131. Gao, W.; McGarry, T.; Orr, C.; McCormick, J.; Veale, D.J.; Fearon, U. Tofacitinib regulates synovial inflammation in psoriatic arthritis, inhibiting STAT activation and induction of negative feedback inhibitors. Ann. Rheum. Dis. 2016, 75, 311–315. [Google Scholar] [CrossRef]
  132. Tsukazaki, H.; Kaito, T. The Role of the IL-23/IL-17 Pathway in the Pathogenesis of Spondyloarthritis. Int. J. Mol. Sci. 2020, 21, 6401. [Google Scholar] [CrossRef]
  133. Sherlock, J.P.; Joyce-Shaikh, B.; Turner, S.P.; Chao, C.C.; Sathe, M.; Grein, J.; Gorman, D.M.; Bowman, E.P.; McClanahan, T.K.; Yearley, J.H.; et al. IL-23 induces spondyloarthropathy by acting on ROR-γt+ CD3+CD4-CD8- entheseal resident T cells. Nat. Med. 2012, 18, 1069–1076. [Google Scholar] [CrossRef] [PubMed]
  134. Najm, A.; McInnes, I.B. IL-23 orchestrating immune cell activation in arthritis. Rheumatology 2021, 60, iv4–iv15. [Google Scholar] [CrossRef] [PubMed]
  135. Brown, M.A.; Wordsworth, B.P. Genetics in ankylosing spondylitis—Current state of the art and translation into clinical outcomes. Best Pract. Res. Clin. Rheumatol. 2017, 31, 763–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Mease, P.; van den Bosch, F. IL-23 and axial disease: Do they come together? Rheumatology 2021, 60, iv28–iv33. [Google Scholar] [CrossRef]
  137. Baeten, D.; Østergaard, M.; Wei, J.C.; Sieper, J.; Järvinen, P.; Tam, L.S.; Salvarani, C.; Kim, T.H.; Solinger, A.; Datsenko, Y.; et al. Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: Results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-finding phase 2 study. Ann. Rheum. Dis. 2018, 77, 1295–1302. [Google Scholar] [CrossRef] [Green Version]
  138. Deodhar, A.; Gensler, L.S.; Sieper, J.; Clark, M.; Calderon, C.; Wang, Y.; Zhou, Y.; Leu, J.H.; Campbell, K.; Sweet, K.; et al. Three Multicenter, Randomized, Double-Blind, Placebo-Controlled Studies Evaluating the Efficacy and Safety of Ustekinumab in Axial Spondyloarthritis. Arthritis Rheumatol. 2019, 71, 258–270. [Google Scholar] [CrossRef] [Green Version]
  139. Gravallese, E.M.; Schett, G. Effects of the IL-23-IL-17 pathway on bone in spondyloarthritis. Nat. Rev. Rheumatol. 2018, 14, 631–640. [Google Scholar] [CrossRef]
  140. Chen, L.; Wei, X.Q.; Evans, B.; Jiang, W.; Aeschlimann, D. IL-23 promotes osteoclast formation by up-regulation of receptor activator of NF-kappaB (RANK) expression in myeloid precursor cells. Eur. J. Immunol. 2008, 38, 2845–2854. [Google Scholar] [CrossRef]
  141. Shin, H.S.; Sarin, R.; Dixit, N.; Wu, J.; Gershwin, E.; Bowman, E.P.; Adamopoulos, I.E. Crosstalk among IL-23 and DNAX activating protein of 12 kDa-dependent pathways promotes osteoclastogenesis. J. Immunol. 2015, 194, 316–324. [Google Scholar] [CrossRef]
  142. Quinn, J.M.; Sims, N.A.; Saleh, H.; Mirosa, D.; Thompson, K.; Bouralexis, S.; Walker, E.C.; Martin, T.J.; Gillespie, M.T. IL-23 inhibits osteoclastogenesis indirectly through lymphocytes and is required for the maintenance of bone mass in mice. J. Immunol. 2008, 181, 5720–5729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kavanaugh, A.; Ritchlin, C.; Rahman, P.; Puig, L.; Gottlieb, A.B.; Li, S.; Wang, Y.; Noonan, L.; Brodmerkel, C.; Song, M.; et al. Ustekinumab, an anti-IL-12/23 p40 monoclonal antibody, inhibits radiographic progression in patients with active psoriatic arthritis: Results of an integrated analysis of radiographic data from the phase 3, multicentre, randomised, double-blind, placebo-controlled PSUMMIT-1 and PSUMMIT-2 trials. Ann. Rheum. Dis. 2014, 73, 1000–1006. [Google Scholar] [CrossRef] [PubMed]
  144. Kragstrup, T.W.; Adams, M.; Lomholt, S.; Nielsen, M.A.; Heftdal, L.D.; Schafer, P.; Deleuran, B. IL-12/IL-23p40 identified as a downstream target of apremilast in ex vivo models of arthritis. Ther. Adv. Musculoskelet Dis. 2019, 11, 1759720x19828669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Ishii, T.; Ruiz-Torruella, M.; Ikeda, A.; Shindo, S.; Movila, A.; Mawardi, H.; Albassam, A.; Kayal, R.A.; Al-Dharrab, A.A.; Egashira, K.; et al. OC-STAMP promotes osteoclast fusion for pathogenic bone resorption in periodontitis via up-regulation of permissive fusogen CD9. FASEB J. 2018, 32, 4016–4030. [Google Scholar] [CrossRef] [Green Version]
  146. Miyamoto, K.; Nishioka, T.; Waki, Y.; Nomura, M.; Katsuta, H.; Yokogawa, K.; Amano, H. Phosphodiesterase 4 inhibitor rolipram potentiates the inhibitory effect of calcitonin on osteoclastogenesis. J. Bone Miner. Metab. 2006, 24, 260–265. [Google Scholar] [CrossRef] [Green Version]
  147. Degboé, Y.; Sunzini, F.; Sood, S.; Bozec, A.; Sokolova, M.V.; Zekovic, A.; McInnes, I.B.; Schett, G.; Goodyear, C.S. Apremilast inhibits inflammatory osteoclastogenesis. Rheumatology 2021, 61, 452–461. [Google Scholar] [CrossRef]
Figure 1. The mechanism of the effect of biological/targeted therapies on bone metabolism in inflammatory arthritis. bDAMARDs, biologic disease-modifying antirheumatic drugs (DMARDs); IL, interleukin; JAKi, Janus kinase inhibitors; TNF-α, tumor necrosis factor-α.
Figure 1. The mechanism of the effect of biological/targeted therapies on bone metabolism in inflammatory arthritis. bDAMARDs, biologic disease-modifying antirheumatic drugs (DMARDs); IL, interleukin; JAKi, Janus kinase inhibitors; TNF-α, tumor necrosis factor-α.
Ijms 23 04111 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chen, T.-L.; Chang, K.-H.; Su, K.-Y. Effects of Biological/Targeted Therapies on Bone Mineral Density in Inflammatory Arthritis. Int. J. Mol. Sci. 2022, 23, 4111. https://doi.org/10.3390/ijms23084111

AMA Style

Chen T-L, Chang K-H, Su K-Y. Effects of Biological/Targeted Therapies on Bone Mineral Density in Inflammatory Arthritis. International Journal of Molecular Sciences. 2022; 23(8):4111. https://doi.org/10.3390/ijms23084111

Chicago/Turabian Style

Chen, Tai-Li, Kai-Hung Chang, and Kuei-Ying Su. 2022. "Effects of Biological/Targeted Therapies on Bone Mineral Density in Inflammatory Arthritis" International Journal of Molecular Sciences 23, no. 8: 4111. https://doi.org/10.3390/ijms23084111

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

Article Metrics

Back to TopTop