You are on page 1of 16

Autoimmunity Reviews 20 (2021) 102760

Contents lists available at ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Review

Exploring IL-17 in spondyloarthritis for development of novel treatments


and biomarkers
Solveig Skovlund Groen a, b, *, Dovile Sinkeviciute a, c, Anne-Christine Bay-Jensen a,
Christian S. Thudium a, Morten A. Karsdal a, Simon Francis Thomsen b, e, Georg Schett f,
Signe Holm Nielsen a, d
a
Immunoscience, Nordic Bioscience, Herlev, Denmark
b
Biomecial Sciences, University of Copenhagen, Copenhagen, Denmark
c
Department of Clinical Sciences Lund, University of Lund, Lund, Sweden
d
Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
e
Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
f
Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen,
Erlangen, Germany

A R T I C L E I N F O A B S T R A C T

Keywords: Spondyloarthritis (SpA) is an umbrella term describing a family of chronic inflammatory rheumatic diseases.
Biomarker These diseases are characterised by inflammation of the axial skeleton, peripheral joints, and entheseal insertion
Spondyloarthritis sites throughout the body which can lead to structural joint damage including formation of axial syndesmophytes
IL-17
and peripheral osteophytes. Genetic evidence, preclinical and clinical studies indicate a clear role of interleukin
pathogenesis
treatment
(IL)- 23 and IL-17 as mediators in SpA pathogenesis. Targeting the IL-23/− 17 pathways seems an efficient
strategy for treatment of SpA patients, and despite the remaining challenges the pathway holds great promise for
further advances and improved therapeutic opportunities. Much research is focusing on serological markers and
imaging strategies to correctly diagnose patients in the early stages of SpA. Biomarkers may facilitate person­
alised medicine tailored to each patient’s specific disease to optimise treatment efficacy and to monitor thera­
peutic response. This narrative review focuses on the IL-17 pathway in SpA-related diseases with emphasis on its
role in pathogenesis, current approved IL-17 inhibitors, and the need for biomarkers reflecting core disease
pathways for early diagnosis and measurement of disease activity, prognosis, and response to therapy.

1. Introduction ankylosis including syndesmophytes formation in the spine. The in­


flammatory processes and new bone formation take place in the
Spondyloarthritis (SpA) represents a group of related but pheno­ entheses, axial skeleton, and peripheral joints through mechanisms that
typically distinct inflammatory autoimmune diseases. SpA includes axial are poorly understood [2]. Extra-articular manifestations can appear
SpA (axSpA, summarizing radiographic (ankylosing spondylitis, AS); with anterior uveitis, as well as co-occurrence of psoriasis (PsO) and
=”” axialSpA) and non-radiographic disease, peripheral SpA (pSpA) and inflammatory bowel disease (IBD) [4]. As mentioned, axSpA consists of
psoriatic arthritis (PsA) [1,2]. These subforms of SpA have overlapping two diagnostic entities, non-radiographic axSpA (nr-axSpA) and AS [5].
clinical features that reflect shared genetic risk factors and pathophys­ The latter is the prototype of axSpA and is characterised by definite
iology. Genetics plays a role in the aetiology of SpA sharing a close as­ radiographic damage in the sacroiliac joints and/or in the spine as
sociation with the major histocompatibility complex (MHC) class I defined by the Assessment of SpondyloArthritis international Society
surface antigen HLA-B27 (human leukocyte antigen B27), which is (ASAS) classification criteria [5,6]. On x-ray, patients with nr-axSpA are
believed to be the major genetic susceptibility factor [3]. visualised without characteristic damage and may represent an early
AxSpA involves inflammation, bone and cartilage loss which is fol­ form of AS. However, some patients may never develop structural
lowed by new local bone formation that can lead to progressive damage on radiographs However, some patients may never develop

* Corresponding author at: Nordic Bioscience, Herlev Hovedgade 205–207, 2730 Herlev, Denmark.
E-mail address: ssg@nordicbio.com (S.S. Groen).

https://doi.org/10.1016/j.autrev.2021.102760
Received 6 November 2020; Accepted 14 November 2020
Available online 22 January 2021
1568-9972/© 2021 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

Fig. 1. Venn diagram illustrating the categorisation and overlapping relationships between the Spondyloarthritis subsets. AS, Ankylosing Spondylitis; PsA, Psoriatic
Arthritis; ReA, Reactive Arthritis; IBD-SpA, inflammatory bowel disease (IBD) associated spondyloarthritis (IBD-SpA). Figure modified from [6].

Fig. 2. Healthy spine compared to spine with ankylosing spondylitis. Severe inflammation can lead to fusion of vertebrae bodies which can result in a less flexible
spine with loss of normal curvature seen in a healthy S-curved spine. Additionally, ankylosing spondylitis is characterised by inflammation in the sacroiliac joints.
Figure has been modified from [23].

structural damage on radiographs [7]. The pSpA category covers the aberrant new bone formation which can lead to fusion of the
inflammation predominantly in peripheral joints rather than axial, sacroiliac joints and zygapophyseal joints, including outgrowth of syn­
commonly affecting the lower extremities asymmetrically. pSpA also desmophytes in the spine bridging the vertebral bodies resulting in
includes enthesitis, anterior uveitis and/or dactylitis. Patients diagnosed ankylosis [17] (Fig. 2). Until now, no treatment has shown improvement
with reactive arthritis and IBD-associated arthritis often fit into the pSpA in long-term outcomes in axSpA, particularly loss of movement caused
category [6] (Fig. 1). In addition, while PsA is usually considered a by bone ankylosis [18]. Hence, treatment that only suppresses inflam­
separate disease entity, PsA often fulfils the criteria for pSpA. mation may not resolve bone loss and aberrant bone formation.
AxSpA affects 0.5–1.5% of the western population [8], and it has In the past decade, strong evidence from genetic, animal, trans­
been suggested that the entire spectrum of SpA may be more prevalent lational and clinical studies has confirmed the role of the IL-23/− 17 axis
than rheumatoid arthritis (RA) in the United States [9]. Although TNF in the pathogenesis of SpA. Clinical evidence suggests that TNF blockade
inhibitor therapy improves symptoms, functional mobility and reduces in AS patients does not influence the IL-23/− 17 axis [19]. Instead,
disease activity [10–14], almost 40% of axSpA patients do not suffi­ therapeutics have been directed towards the IL-23 and IL-17 which are
ciently respond to anti-TNF treatment [15,16]. Therapies directed considered as key disease-modifying targets in the treatment of SpA.
against TNF may be successful in suppressing the inflammation, how­ This has recently contributed to development of a range of novel ther­
ever, axSpA involves both inflammation, bone loss and new bone apeutics targeting IL-17 and IL-23 pathway in SpA, PsA and PsO [20]. IL-
formation. 17 inhibitors have shown beneficial effects on symptoms by significantly
Bone erosion seen in AS is a frequent complication but is often of improving psoriatic skin lesions, reducing pain and inflammatory joint
limited severity. Eventually, the principal cause of structural damage is lesions [21]. Furthermore, recent results have showed that IL-17

2
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

Fig. 3. IL-17 signalling pathway. IL-17 binds the


heterodimeric IL-17RA and IL-17RC receptor complex
mediating downstream signalling. The recruitment
and binding of Act1 to the cytoplasmic tail of IL-17R
together with the binding of TRANF6 mediates the
canonical pathway where C/EBP, AP-1 and NFKB
pathways can be activated resulting in transcription
of inflammatory genes. Additionally, Act1 interacts
with the TRAF2/TRAF5 complex modulating the
noncanonical pathway where the mRNA stability is
controlled through multiple RNA-binding proteins,
including HuR and Arid5a. TRAF3 can bind directly
to the IL-17R to block the formation of the IL-17R-
Act1-TRAF6 complex. Furthermore, TRAF4 can
compete with TRAF6 for the TRAF binding site on
Act1 and inhibit TRAF6 signalling. IL-17R, IL-17 re­
ceptor; TRAF, TNF receptor associated factor; SEFIR,
similar expression of fibroblast growth factor and IL-
17Rs; SEFEX, SEFIR-extension; HuR, Human antigen
R; Arid5a, AT-rich interactive domain-containing
protein; TAK1, transforming growth factor β-acti­
vated kinase 1; NFKB, nuclear factor kappa light
chain enhancer of activated B cells; MAPK/AP1,
mitogen-activated protein kinase activator protein-1.
Figure modified from [27,28,39].

inhibition arrests the progression of catabolic and anabolic bone often co-produced with IL-17A by Th17 cells [26]. Both IL-17A and IL-
changes in the joints of PsA patients and may potentially slow disease 17F exist as disulphide-linked homodimers or as IL-17A/-17F hetero­
progression [22]. Yet, comprehensive evidence is still needed to deter­ dimers. All forms of IL-17A and IL-17F cytokines signal through a het­
mine whether these therapeutics control the SpA disease and succeed as erodimeric IL-17RA and IL-17RC receptor complex. The cytoplasmic tail
effective disease-modifying anti-rheumatic drugs (DMARDs). of IL-17RA possesses discrete signalling motifs, a conserved SEFIR
In recent years, studies have focused on clinically applicable bio­ domain (similar expression of fibroblast growth factor and IL-17Rs) with
logical markers to overcome the challenges in the management of SpA an additional ‘SEFIR-extension’ domain (SEFEX) which are both
related to unclear pathogenesis, lack of early diagnosis and accurate required for functional downstream IL-17 signalling [27]. Upon IL-17A
monitoring of disease activity and inability to predict joint damage or binding to the IL-17R complex, the IL-17R undergoes a conformational
response to treatment. Applying biomarkers to address the many chal­ change enabling interactions between the signalling adaptor protein
lenging aspects of SpA may enhance the development of novel treat­ Act1 and SEFIR domains mediating downstream events [28]. The
ments which may offer more specialised and effective treatment of recruitment of Act1 represents a hallmark of IL-17 signalling and is an
patients [24]. essential signalling component of the IL-17R signalling event, not seen in
This narrative review will explore the role of IL-17 in homeostasis any other known class of receptors [29]. Act1 contains a TNF receptor
and its heterogeneous effects in inflammation and structural damage to associated factor (TRAF)-binding site which enables association with
justify for the potential advantageous effect of IL-17 inhibition in AS and different TRAF family proteins. Hereof, Act1 operates as a docking sta­
PsA. Furthermore, the review will assess the current challenges in the tion for the different TRAF proteins being able to activate several in­
management of SpA and summarise promising biomarkers of inflam­ dependent signalling pathways. The engagement with TRAF6 drives the
mation and tissue remodelling including parameters of angiogenesis and activation of the canonical pathway (de novo gene transcription) which
autoantibodies that may improve the diagnosis, prognosis and treatment includes the nuclear factor kappa light chain enhancer of activated B
outcomes in SpA. cells (NFKB) pathways activated by the transforming growth factor
β-activated kinase 1 (TAK1), mitogen-activated protein kinase activator
2. IL-17 signalling and production protein-1 (MAPK/AP1) pathways and C/EBP pathways [30].
Collectively, these IL-17A/-17F-activated pathways initiate tran­
The IL-17 superfamily consists of six structurally related proin­ scriptional induction of target genes resulting in release of proin­
flammatory cytokines IL-17A, IL-17B, IL-17C, IL-17D, IL-17E and IL- flammatory cytokines (IL-1β, IL-6, GM-CSF, G-CSF and TNF-α),
17F, which are involved in the immune-mediated antimicrobial chemokines (CXCL1, CXCL2, CXCL8, CCL20 and many others), antimi­
response. Excessive production of IL-17 during chronic inflammation crobial peptides (AMPs; mucins, defensins), tissue remodelling matrix
has been associated with development of inflammatory diseases [20]. IL- metalloproteinases (MMPs; MMP -1, − 2, − 3, − 8, − 9, and − 13), and
17A is the best characterised member of the IL-17 family and generally additional inflammatory effectors (acute-phase proteins and comple­
referred to as IL-17. Of the six cytokine family members, IL-17F is the ment) [31,32]. Furthermore, the IL-17A/-17F-mediated production of
closet related member to IL-17A with 55% sequence identity [25] and IL-1β, IL-6, and TNF-α contributes to a positive feedback loop leading to

3
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

enhanced IL-17 signalling [31,33]. Besides activating the transcription response have been strongly associated with the development of AS
of inflammatory genes, the binding of TRAF2 and TRAF5 to Act1 acti­ [79], PsO [80], and IBD [81]. Additionally, a genome-wide association
vates the noncanonical pathway, which mediates post-transcriptional study (GWAS) in AS has identified several variants in genes including
mRNA stabilisation, particularly CXCL1 mRNA encoding chemokines CARD9, IL-12B, PTGER4, IL-6R, IL-27, TYK2 and STAT3, which are
[34]. The post-transcriptional stabilisation is controlled through multi­ implicated directly or indirectly in the IL-23/− 17 axis [82,83].
ple RNA-binding proteins, including the human antigen R (HuR) and AT-
rich interactive domain-containing protein 5a (Arid5a) [35,36]. The 3.2. Evidence from preclinical studies
stabilised transcripts are deposited in cytoplasmic granules, where they
can be translated or degraded instantly [27]. The IL-17-mediated sig­ The importance of IL-17 in the pathogenesis of SpA has further been
nalling not only activates proinflammatory cascades but also promotes defined in several animal studies. In 2012, Sherlock et al. systemically
numerous regulatory pathways. Upon IL-17A stimulation, TRAF4 which overexpressed IL-23 in transgenic mice and identified a unique popu­
acts as a negative modulator of IL-17-mediated signalling, can be lation of CD3+/CD4-/CD8- T cells in the enthesis which expressed the
recruited and compete with TRAF6 for Act-1 binding [36,37]. Addi­ Th17 transcription factor RAR-related orphan receptor-γt (ROR-γt), and
tionally, TRAF3 also acts as a negative regulator by similar competing the IL-23 receptor. Upon activation with IL-23, these T cells produced IL-
action on the IL-17 signalling [36,38] (Fig. 3). The IL-17 signal trans­ 17A, IL-17F, IL-6, and IL-22 resulting in enhanced axial and peripheral
duction overall consists of many non-redundant mechanisms to fine- enthesis and sacroiliitis followed by IL-22 dependent osteoproliferation
tune IL-17-induced inflammation, however, these mechanisms are not in the absence of synovial inflammation. In addition, blockade of IL-17
fully understood [27]. and IL-22 with neutralising antibodies significantly diminished the
disease severity, especially when the two antibodies were administered
2.1. Effects of IL-17 on inflammation together [84]. The existence of this unique T cell population may explain
the generation of the SpA phenotype in the early phases of the disease,
In close cooperation with the other IL-17 cytokine family members, without activating CD8+ T cell mediated autoreactivity or other IL-17
IL-17A plays a prominent role in host defence to clear off pathogens at secreting cells [18]. IL-17-deficient mice have shown to exhibit less
epithelial and mucosal barriers in the skin, intestinal tract and airways inflammation in a type II collagen-induced arthritis (CIA) model [85]
[40–42]. The inflammation is predominantly induced through the and the IL-17R-deficient mice also demonstrated reduced secretion of
ability of IL-17 to induce the secretion of chemotactic factors which proinflammatory cytokines into the synovium preventing cartilage
leads to recruitment of neutrophils to the site of infection [43,44]. IL-17 degradation [86].
also stimulates the release of chemokine CCL20 which promotes the
migration of immature dendritic cells and Th17 cells [45]. Moreover, IL- 3.3. Evidence from clinical studies
17 stimulates various cell types, including keratinocytes [46–49],
chondrocytes [50–52], fibroblasts [53,54], osteoclasts [55], osteoblasts IL-17 and IL-23 levels have been measured in serum of AS patients
[56], endothelial cells [57–59], macrophages [60–63] and epithelial and healthy controls by enzyme linked immunosorbent assay (ELISA).
cells [64]. Interaction between migrating Th17 cells and local mesen­ Results demonstrated significantly elevated IL-17 and IL-23 levels in AS
chymal cells contributes to an immense production of IL-17 [65]. On its patients compared to controls [87–89]. Furthermore, flow cytometry
own, IL-17 is a weak inducer of inflammation compared to other measurements showed increased numbers of circulating Th17 cells
proinflammatory molecules, such as TNF-α. Nonetheless, IL-17 can act found in the peripheral blood (PB) of PsA and AS compared to RA pa­
synergistically with TNF-α and other proinflammatory cytokines and tients, which showed no increase of circulating Th17 cells in PB [90,91].
inflammatory effectors, generating a powerful and rapid inflammatory In addition, Th17 levels in synovial fluid samples from PsA patients were
response [27,66–68]. analysed by flow cytometry. In PsA patients, levels of Th17 cells were
The cellular production of IL-17 is complex and heterogeneous. Both significantly elevated compared to RA patients and correlated with
innate cell populations and different adaptive T cell subsets can produce disease activity measures and progression of radiographic joint erosion
IL-17. Notably, naïve CD4+ T cells differentiated into Th17 cells are the [91]. Increased mRNA expressions of IL-17 and IL-22 were found in skin
main producing cells of IL-17. However, during inflammation most IL-17 lesions from PsO patients including elevated levels of Th17 cells in PB
is secreted by innate immune cells including innate-acting lymphocytes measured by fluorescence-activated cell sorting (FACS) [92]. In mast
such as γδ T cells, invariant natural killer T (iNKT) cells [69], lymphoid cells, IL-17 levels measured by ELISA were upregulated in the synovium
tissue inducer (LTi) cells [70], type 3 innate lymphoid cells (ILC3s) [71], of peripheral joints in patients with SpA [93]. By immunohistochemical
macrophages [72], neutrophils [73] and Paneth cells [74]. Additionally, analysis, enhanced expression levels of IL-17 were found in CD15+
IL-17 can be secreted by Tc17 cells subsets of CD8+ T cells [75,76] and neutrophils located in the subchondral bone marrow of inflamed spine
resident memory T cells [77]. Several animal and human studies have from SpA patients with low abundance of Th17 cells indicating that IL-
demonstrated that these cells produce IL-17 in response to inflamma­ 17 producing cells other than the Th17 cells are relevant in local
tion, but their respective contributions to the SpA pathogenesis remain inflammation in SpA [94]. IL-23, but not IL-17, has been found over­
unexplored [30]. expressed in inflamed ileum in AS patients analysed by RT-PCR [95]. IL-
Although the pathogenic mechanisms underlying SpA are not fully 23 positive cells within the subchondral bone marrow and fibrous tissue
elucidated, multiple areas of investigation have recently converged to in facet joints of AS patients were significantly elevated compared to
provide strong evidence implicating the IL-23/− 17 axis in rheumatic osteoarthritis (OA) patients measured by immunohistochemistry [96].
diseases including AS and PsA. The following section will describe Altogether, these results are strong evidence of both the innate and
several lines of evidence from genetics, preclinical and clinical studies adaptive mechanisms including the IL-23/− 17 axis may drive the
emphasising a pivotal role of the IL-23/− 17 axis in SpA. pathogenesis of SpA.
In 2011, Bowness et al. found elevated numbers of CD4+ Th17 cells
3. Role of the IL-23/¡17 axis in SpA expressing the receptor KIR3DL2 in PB and synovial fluid from AS and
SpA patients. KIR3DL2 responds to the HLA-B27 homodimer molecule
3.1. Genetic evidence by promoting survival of Th17 cells and upregulation of IL-17 secretion
in AS patients [97]. Nevertheless, levels of CD4+ Th17 cells were not
IL-23 is responsible for promoting the expansion and survival of found elevated in other studies [94,98] including the expression of
Th17 subsets including the production of IL-17 from Th17 cells [78]. IL- KIR3DL2 [99], whereas IL-23R+ γδ T cells were observed instead [100].
23R gene polymorphisms with functional relevance in T cell immune γδ T cells have shown to be involved in large productions of IL-17 in a

4
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

murine model accumulating enthesitis with enhanced bone regeneration formation to maintain bone homeostasis [121].
[101,102]. Analysis of the PB and synovial fluid in SpA has been Abnormal bone turnover is a feature shared by several rheumatic
considered inadequate to uncover whether pathologic processes were diseases including AS and PsA, and these bone alterations are displayed
present or not in the sacroiliac joints, vertebral bodies, zygapophyseal in different forms [17,120]. Bone turnover in AS and PsA is distin­
joints including bone marrow next to the axial inflammatory lesions. For guished from RA by occurrence of prominent focal areas of new bone
instance, TNF-α serum levels were not consistently elevated in SpA pa­ formation involving endochondral ossification. This process is normally
tients and even found to be lower than in healthy controls [18]. Thus, active during the period of skeleton growth and development and in­
this viability of the T cell subtypes seems to be an important factor volves the replacement of the cartilaginous matrix with mature miner­
compared to TNF-α levels and may be an essential therapeutic target. alised bone. However, during bone healing after e.g. fracture or
The important role of IL-23/− 17 in SpA has not only been evidenced pathological bone growth associated with the ankylosing process,
as mentioned above, but the significant clinical efficacy of IL-17 in­ endochondral bone formation can be reactivated through a series of cell
hibitors in treating AS, PsA and PsO has also been acknowledged and differentiation steps ultimately leading to new postnatal bone forma­
will be discussed in the next section. tion. Although the mechanism behind the reactivation of endochondral
bone formation is incompletely understood, it is believed that the
4. Targeting the IL-23/¡17 axis in SpA mesenchymal progenitor cells commit to chondrogenic differentiation
ending up differentiating into hypertrophic chondrocytes which begin to
As evidence from genetic, animal and human studies support the calcify the cartilage tissue matrix. Consequently, through direct bone
importance of IL-23 and IL-17 in the pathogenesis of SpA, therapeutics formation osteoblast precursor cells invade and progressively degrade
inhibiting IL-23 would be expected to show similar clinical efficacy as the matrix and replace it with bone [122]. In addition, hypertrophic
drugs inhibiting IL-17 in SpA. Interestingly, clinical studies with IL-12/ chondrocytes have been shown to transdifferentiate into osteoblasts in
IL-23 inhibitor, ustekinumab, showed lack of efficacy in axSpA, while endochondral bone during development [123]. This indicates that
effective in PsA and PsO [103]. Furthermore, the efficacy of risankizu­ endochondral and direct bone formation are both involved in the pro­
mab, an IL-23 p19 subunit inhibitor, was evaluated in patients with gressive ankylosis with the outlay of a cartilage template as a key
active AS in a phase II study. Risankizumab did not reduce the signs and element in the development of syndesmophytes [122]. This aberrant
symptoms of AS patients compared to placebo, questioning whether IL- new bone arises at specific localised anatomical sites and may be the
23 is a relevant driver of AS pathogenesis and symptoms [104]. effect of prolonged entheseal inflammation [124]. Both IL-23 and IL-17
Conversely, IL-17A inhibition has demonstrated efficacy in AS, PsA, and are involved in the pathway that expands entheseal inflammation and
PsO in clinical trials. A fully human anti-IL-17A monoclonal antibody, are closely linked to the new bone formation by promoting catabolic and
secukinumab, has been approved for treatment of AS, PsA and PsO based anabolic bone alterations [124].
on several large randomized controlled trials [105–110]. Another IL-
17A inhibitor, ixekizumab, a humanized anti-IL-17A antibody has 5.2. The effects of IL-17 on bone
been approved for treatment of PsA and PsO and has shown significant
efficacy in AS in two large phase III trials [111–115]. These results Bone loss seen in PsA, and in some AS patients, is believed to be
suggest that IL-17A is a key cytokine facilitating disease pathogenesis in related to activation of the IL-23/− 17 axis by stimulating differentiation
AS, PsA and PsO. In AS, IL-17 seems to be produced in a largely IL-23- of osteoclasts. Kotake et al. have demonstrated that IL-17 influences
independent way. Furthermore, in human interspinous entheseal tis­ bone homeostasis by showing that IL-17 promoted osteoclastogenesis in
sue IL-23 receptor positive and negative subpopulations of γδ T cells an osteoblast-osteoclast co-culture model [125]. IL-17 can induce the
were found signifying the role of IL-23-independent IL-17 production expression of receptor activator of NFKB ligand (RANKL) on osteoblasts
[116]. A broader understanding of the divergent roles of IL-23 and IL-17 which stimulates RANK signalling on osteoclastic precursors vital for the
in the pathophysiology of axSpA is still needed. Evidence towards production of mature osteoclasts [126–128]. Several studies have
anatomical and immunological differences between axial and peripheral demonstrated pro- osteoclastogenic effect of IL-17 [129,130] including
disease may be one of the reasons for the IL-23/− 17 axis pathway Th17 cells being the main T cell subset to stimulate osteoclast iffer­
divergence. For instance, more peripheral enthesis is observed in PsA entiation by releasing IL-17 and increasing expression of RANKL on
with inflammation in the entheseal soft tissue [117] while in AS, osteoblasts [131]. During chronic inflammation, IL-17 has been shown
inflammation is observed in the peri-entheseal bone in the spine and to activate MMPs and promote matrix turnover leading to cartilage loss
more severe axial bone edema has been observed with much higher in joint tissue [132]. Chabaud et al. have demonstrated that adding IL-
frequency in HLA-B27 positive patients [118]. 17 to human bone samples increased the production of bone-
destructive cytokines and bone resorption in vitro [133]. In contrast,
5. Implication of IL-17 in bone damage the inhibition of IL-17 showed an anti-inflammatory effect and reduced
bone destruction and an even more efficient anti-inflammatory effect
Currently, the focus has primarily been on the mechanisms that was obtained when IL-17 was inhibited in combination with TNF-α and
trigger chronic inflammation in joints and entheses. However, as a result IL-6 [67]. These studies suggest that IL-17 activity affects the bone ho­
of chronic inflammation, structural damage evolves which is a critical meostasis by inducing osteoclastogenesis which partially explains the
contributor to morbidity in patients suffering from AS and PsA [119]. development of destructive bone lesions and loss seen in PsA and AS.
In addition to the effects of IL-17 on bone loss, IL-17 also critically
5.1. Bone homeostasis and abnormal turnover orchestrates the activation and differentiation of osteoblasts. Studies
have shown that IL-17 enhances osteogenic differentiation of mesen­
The bone remodelling cycle is a tightly regulated balance between chymal stem cells (MSCs) [134–136]. Furthermore, Osta et al. have
osteoclasts, which are responsible for bone resorption involving removal demonstrated that IL-17 combined with TNF-α can stimulate osteogenic
of mineralised bone matrix, and osteoblasts, which form and replace the differentiation of human MScs [137]. Although these findings suggest
resorbed bone [120]. To maintain bone homeostasis, the resorption and that IL-17 is involved in the osteogenesis, the role of IL-17 on osteogenic
formation processes are tightly coupled ensuring that the amount of MSCs is still not clear.
bone removed equals the amount of bone formed. Within the bone Besides the effects of IL-17 on bone remodelling, the development of
matrix, bone growth factors, such as transforming growth factor-β (TGF- enthesitis and entheseal new bone formation have been shown to
β) and bone morphogenetic proteins (BMPs) are stored. These growth depend on IL-22 [138]. Entheseal resident T cells producing IL-22 have
factors are released during the resorptive phase promoting bone been shown to activate osteoblast-mediated bone remodelling [84].

5
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

Fig. 4. Overview of the bone erosion and new bone formation in AS and PsA activated by the IL-23/− 17 axis. IL-23 stimulates Th17 to secrete IL-17 and IL-22.
Subsequently, IL-17 induces the expression of RANKL on osteoblasts which initiates osteoclast differentiation via upregulating of RANK, the receptor for RANKL.
In combination with other proinflammatory cytokines such as TNF-α, IL-1, and IL-6, IL-17 can also directly promote osteoclastogenesis leading to bone erosion.
Moreover, the pathological bone growth includes both direct bone formation and reactivation of the endochondral bone formation. IL-17 and IL-22 promote dif­
ferentiation of bone marrow-derived mesenchymal stem cells (MSCs) which further differentiate either into hypertrophic chondrocytes that form calcified cartilage
tissue matrix, or osteoblasts promoting osteoproliferation. Eventually, the calcified cartilage will be progressively degraded and replaced with bone matrix by os­
teoblasts. This exaggerated new bone formation can result in ankylosis. RANK, receptor activator of NFKB; RANKL, receptor activator of NFKB ligand. Adapted
from [18,122,124].

Furthermore, in vitro IL-22 has been shown to enhance MSC osteo­ compared to primary erosive forms of arthritis such as rheumatoid
genesis and increase proliferation and migration of human MSCs in in­ arthritis (RA). Although synovial inflammation in the peripheral joints
flammatory environments [139]. Hereof, IL-22 may play an important of patients with AS exhibits histopathologic characteristics similar to
role of the IL-23/− 17 axis in bone remodelling providing a novel those seen in the RA joints, differences have been observed in the excess
pathway for studying the pathological and inflammatory osteogenesis in bone formation and repair pattern [120]. IL-17 has showed to promote
SpA [30]. excessive osteogenesis at sites of entheses under conditions of mechan­
ical stress, resulting in formation of new bone and syndesmophytes, the
key difference between RA and AS [146]. Determining the heteroge­
5.3. Aberrant bone formation in AS neous effects of IL-17 in new bone formation remains a central avenue of
future research.
AS is mainly characterised by ankylosis of the sacroiliac joints and
vertebral bodies which involves excessive bone formation encompassing
both endochondral and direct bone formation [17]. Inflamed sacroiliac 5.4. Bone erosion and formation in PsA
joint tissue from patients with AS has been examined by Braun et al. who
observed localised nodules of endochondral ossification within the Whereas AS is mainly characterised by extensive aberrant bone
inflamed tissue [140]. TGF-β and BMPs have been proposed as potential formation primarily in the axial joints, PsA manifests in the peripheral
key players of the enhanced bone formation in AS. The endochondral joints with both increased bone destruction and increased bone forma­
bone formation during development and postnatal fracture repair are tion [147] and differs from RA by its characteristic calcification of
induced by TGF-β and BMPs and have been implicated in this process entheses [148]. Despite main involvement of peripheral joints, certain
[141,142]. TGF-β and BMPs signal through the wingless (Wnt) pathway PsA patients may have axial involvement with inflammatory back pain
which is a main regulator of skeletal patterning and post-natal bone and sacroiliitis. In PsA, the bone remodelling homeostasis is disrupted by
remodelling [143]. The activation of the Wnt/ β-catenin pathway results several cytokines and growth factors altering the differentiation and
in transcriptional activation of the osteoprotegerin (OPG) gene, which is function of osteoclasts and osteoblasts [149]. These cytokines include
a potent inhibitor of RANK ligand, the principal regulator of osteoclast both IL-17, IL-6, TNF-α, RANKL, and the growth factors BMPs and TGF-
differentiation and activation [144]. β, similar to AS [150].
Lories et al. have elaborated on the concept of a potential relation Prolonged effect of inflammatory cytokines such as IL-17 has been
between biomechanical factors and the ankylosing process [122]. The linked to increased fracture risk in PsA patients [151]. As previously
skeleton consists of mechanosensitive tissue which can adapt biome­ mentioned, IL-17 induces osteoclast differentiation and facilitates bone
chanical loading controlled by the bone remodelling cycle. In axSpA erosion [126–128,131,152] and these observations provide basis for
patients, osteitis in vertebral bodies and sacroiliac joints influences the focusing on IL-17 inhibition in PsA to protect from bone erosion.
normal bone homeostasis by cytokine-mediated osteoclast activation Consistent with these results, clinical studies have showed that PsA
increasing net bone loss. Lories et al. suggested that increased bone net patients treated with IL-17A inhibitors, exhibit significantly reduced
loss leads to instability of the axial skeleton and that the loss of stability inflammation and structural damage versus placebo in the short- and
triggers tissue remodelling which includes syndesmophyte formation long-term [106,153,154]. Recently, the PSARTROS study demonstrated
[122]. Studies have suggested that the bone may not be able to no progression of catabolic or anabolic alterations in the joints of PsA
compensate for the bone loss in a normal physiological way due to patients treated with secukinumab for 24 weeks [22].
osteoblast activation being suppressed by proinflammatory cytokines or Comparing RA and PsA, similar cytokine expressions have been
bone formation inhibited by the Wnt signalling antagonist Dickkopf-1 observed suggesting related mechanisms involved in joint inflammation
(Dkk1) driven by TNF-α [145]. and destruction [155,156]. However, significant differences between
In summary, bone remodelling seen in AS seems much more complex RA and PsA are seen in the bone resorptive phenotype. In RA, the

6
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

Fig. 5. IL-17 therapy options for management of SpA and upcoming treatment. Overview of approved IL-17 inhibiting therapies and novel drugs investigated in the
development phases for the treatment of patients with psoriasis (PsO), ankylosing spondylitis (AS), and psoriatic arthritis (PsA). mAb, monoclonal antibody; IL-17R,
IL-17 receptor. Data adapted from [177].

phalangeal joints are most frequently involved in a symmetric manner Yet, the connection between inflammation and new bone formation is
with U-shaped lesions involving bone destruction and insufficient repair not clearly understood. In mouse models of PsA, BMP signalling in DBA/
leading to local and systemic bone loss [157,158]. In contrast, PsA ex­ 1 mice showed to trigger spontaneously PsA-like arthritis indicating that
hibits a tubular or omega-like asymmetric bone erosion pattern less BMP signalling may be related to the abnormal development of bone in
severe in size and depth [159]. This suggests that the bone repair PsA [141,163].
mechanism and pathogenesis may be different between PsA and RA. Unlike RA, decreased levels of Dkk1 have been reported in PsA serum
New bone formation has been observed in PsA patients with enthe­ compared to healthy subjects which agrees with the increased bone
seophytes and ankylosis mainly manifested in the peripheral joints. formation and indicates that Wnt/β-catenin signalling plays an impor­
However, bone formation in the axial skeleton has also been observed in tant role in directing osteoblast differentiation and function in PsA
PsA thus, axial disease can occur both in AS and PsA patients. Likewise, [164]. Interestingly, in serum samples from AS patients high levels of
PsO can occur in AS patients which has initiated a debate on whether Dkk1 showed to protect from developing syndesmophytes [165]. Other
axial PsA is essentially AS with PsO, or if these conditions are two studies have reported additional high levels of Dkk1 in PsA patients
separate diseases with overlapping features [160]. Like in AS, the highlighting the complexity of PsA as a disease involving the combina­
mechanisms behind the new bone formation are currently unknown. tion of extensive bone destruction and aberrant bone formation
Hereof, the aberrant bone formation seems to be like AS, but they differ [166,167] (Fig. 4).
somewhat in their pathology [161].
According to radiographic differences between AS and PsA, the 6. IL-17 therapy options for management of SpA
syndesmophytes are symmetrically distributed with a primary marginal
shape in AS, while the distribution in PsA is rather asymmetric along the In early 2015, secukinumab was approved in the US and the EU as
spine with equal amounts of marginal and paramarginal syndesmo­ the first IL-17A inhibiting drug developed for treatment of patients with
phytes. From radiographs, the axial severity appears to be worse in AS plaque PsO [168] and in 2016 secukinumab was approved as treatment
than in PsA by the number of syndesmophytes and the Bath AS Radi­ of AS and PsA patients [169]. Subsequently, another IL-17 inhibitor,
ology Index (BASRI) [162]. Like in AS, anabolic growth factors are ixekizumab, was approved for the treatment of plaque PsO and PsA and
considered to play one of the key roles in the bone formation in PsA by is yet under investigation for treatment of AS [170]. In 2017, the latest
mediating the formation of hypertrophic chondrocytes and osteoblasts. IL-17 receptor inhibitor class medication, brodalumab, was approved for

7
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

treatment of patients with plaque PsO. Brodalumab, however, has been 7.1. CRP
associated with increased risk of suicidal behaviour [171].
Several novel drug candidates of anti-IL-17 are currently investi­ CRP is an acute-phase protein which reflects systemic inflammation.
gated in the development phase. Netakimab, tackling IL-17A is under Although CRP is not elevated in a substantial proportion of patients with
investigation for treatment of plaque PsO and is now in phase III clinical active SpA, it is commonly used as a reliable acute inflammatory marker
trials [172]. Bimekizumab, tackling both IL-17 A and IL-17F has shown of disease activity. Levels of CRP are frequently higher in patients with
promising results in phase II trial in AS, PsA and PsO and is currently full-blown radiographic forms of AS compared to nr-axSpA. CRP cor­
undergoing phase III trials [173]. Another bispecific agent, afaseviku­ relates with Ankylosing Spondylitis Disease Activity Score (ASDAS) and
mab, which neutralises IL-17A and IL-17F, is in early phases of clinical moderately correlates with MRI inflammation which is the preferred
trials [174]. One more novel approach is the development of the method to assess disease activity in SpA, though very costly [184]. In a
nanoantibody, ALX-0761, which is undergoing phase II clinical trials for study by Poddubnyy et al. elevated levels of CRP were observed as a
treatment of PsO [175] (Fig. 5). For future treatment of SpA, bispecific solid predictor of radiographic sacroiliitis progression in AS and nr-
biologic therapies which inhibit both TNF-α and IL-17 are of great in­ axSpA patients [185]. Subsequently, in 2016 Poddubnyy et al. investi­
terest. Dual inhibition of these cytokines in a type II collagen-induced gated the link between several disease activity parameters (CRP, ASDAS
arthritis model has been shown to improve protection against joint and BASDAI) and radiographic spinal progression in early AS patients.
damage compared to TNF-α inhibition alone [68,176]. Thus, the com­ Here, five time points of the mentioned parameters were available
bination of blocking TNF-α and IL-17 may provide additional thera­ during 2 years of follow up. The study confirmed an association between
peutic value for treatment of SpA. Dual IL-17/TNF-α inhibitors, such as high disease activity and development of structural damage in the spine
remtolumab and COVA322, are in phase II clinical trials [173]. in patients with early AS [186]. In several studies, SpA patients have
been treated with TNF inhibitors which has been shown to decrease the
7. Biomarkers in drug discovery and development elevated CRP levels, and thus, CRP has been demonstrated as a reliable
biomarker for therapeutic response [187–189]. In a study by Pedersen
Over the past decade it has become clear that biomarkers constitute a et al. the development of new syndesmophytes resulted in a more sig­
high priority for the drug discovery process and understanding of the nificant decrease in CRP levels while chronic systemic inflammation was
pathogenesis of SpA. Although the ASAS classification and response associated with absence of radiographic progression throughout a brief
criteria are useful clinical tools, they are built on patient-reported out­ period [188]. These findings support the hypothesis that resolved
comes including questions about pain and stiffness which is purely inflammation results in an increased rate of ossification of the repaired
subjective and can have high levels of individual variability and bias. tissue whereas persistent inflammation prevents the formation of syn­
Hereof, the usage of more objective measures should be considered in desmophytes [184].
combination with the ASAS criteria [170].
The rheumatoid factor (RF), anti-citrullinated protein antibodies
(ACPA), erythrocyte sedimentation rate (ESR) and C-reactive protein 7.2. Anti-CD74
(CRP) levels are well-established serological biomarkers to evaluate
disease activity in RA patients [178]. However, SpA patient are sero­ CD74, also known as human lymphocyte antigen (HLA) class II
negative by the lack of induced levels of serum autoantibodies including gamma chain or invariant chain, is involved in the assembly and
RF and ACPA, and ESR and CRP only are used as serum biomarkers, transport of human histocompatibility complex class II (MHC II) mole­
directly reflecting inflammation. CRP is a relative insensitive marker in cules [190]. It is believed to mediate B cell proliferation and bind the
SpA diseases and correlates poorly with disease activity and is not pre­ macrophage migration inhibitory factor (MIF) leading to NFKB activa­
sent in all AS patients thus, may not be useful [179,180]. The onset of tion and thus, expression of pro-inflammatory mediators [191]. ELISA
disease in SpA patients typically manifests between the ages of 20–30 studies performed by Baerlecken et al. 2013 observed antibody reac­
years with slow progression and is associated with vague symptoms tivity against full-length human CD74 in 56% of 41 patients sera with AS
[181]. Altogether, with the lack of confirmatory blood tests and an and 5% of 100 healthy subjects. Furthermore, antibody reactivity
average delay in diagnosis of AS of 5–10 years, there is a growing in­ against CLIP domain of the CD74 protein was observed in 67% of the
terest in the discovery of new biomarkers [181]. Earlier diagnosis and 216 SpA patients with axial (n = 156) and peripheral (n = 60) SpA.
initiation of appropriate therapy would improve the success rates of Antibody reactivity was also observed in 45% of 40 PsA patients, 11% of
clinical remission and may prevent the development of structural lesions the 80 RA patients, 15% of the 40 patients with systemic lupus, and 0.8%
including ankylosis. of 125 healthy blood donors. In the 67% of SpA patients, 97% experi­
In recent years, several new and expensive therapeutic options such enced inflammatory back pain of <1 year and hereafter, the reactivity
as TNF and IL-17 inhibitors have been approved for treatment of PsA and declined by 65% after >20 years after onset and no association with
AS. These agents have shown to improve symptoms, quality of life, and disease activity was observed indicating a potential biomarker for early
slow down radiographic progression. However, new data on the genetic diagnosis of SpA [192]. In an additional European study, similar results
profiles of the different types of arthritis have appeared with clear evi­ were observed with reactivity in 85.1% of 94 axSpA patients and in 7.8%
dence of little or no genetic overlap between the different types of of 51 controls [193]. However in another study, anti-CD74 IgG and IgA
arthritis, suggesting that therapies may not be efficient on all these forms autoantibodies have no diagnostic value in early axSpA demonstrating
[182]. For example, despite the positive effect of IL-17A inhibition in AS reactivity with IgG antibodies in 46.4% of 274 axSpA patients and
and PsA, clinically secukinumab showed to be ineffective in SpA patients 47.9% of 319 non-SpA chronic back pain controls [194]. This points
with IBD and even exacerbated the symptoms in some patients [183]. towards two opposite conclusions in the role of anti-CD74 autoantibody
Due to the improved understanding of AS and PsA together with the new in the diagnosis of SpA. Furthermore, anti-CD74 was measured by ELISA
therapeutic options, more accurate analysis tools are needed to predict in sera from 71 Chinese SpA patients and 70 controls. The results
early diagnosis, disease activity, disease progression and response to demonstrated reactivity of 14.1% in SpA patients and 2.9% in healthy
therapy significantly increasing the need for new biological markers. controls. Although these results showed higher reactivity in SpA
Several biomarkers have been proposed for SpA but they are still in the compared to controls, the prevalence of anti-CD74 was lower than in the
research phase and not yet widely utilised in clinical practice. This European studies. This suggests that CD74 may not be a useful diag­
section reviews well-known and new promising biological markers. nostic biomarkers in an Asian population [195]. Based on these results,
further investigation is needed to test the efficiency of CD74 as a good
target biomarker molecule for axSpA diagnostics.

8
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

7.3. PPM1A prospective laboratory tool to assess disease activity in axSpA patients.
However, this study had several limitations such as the lack of whole-
Protein phosphatase magnesium-dependent 1A (PPM1A) is a serine/ body MRI in AS and nr-axSpA to examine the correlations between
threonine protein phosphatase contributing to the formation of bone CRPM and local inflammation [205].
including syndesmophytes which is a key feature of AS. PPM1A partic­
ipates in the bone formation by regulating the BMP and Wnt signalling 7.6. Connective tissue remodelling markers
pathways, including suppressing TGF-β signalling [196,197]. By using
high-density protein microarrays, antibody reactivity to PPM1A was In inflammatory joints disease, the tissue remodelling homeostasis is
tested in sera from patients with PsA (n = 34), PsO (n = 6), and AS (n = disrupted leading to altered extracellular matrix (ECM) remodelling.
16), RA (n = 21), juvenile idiopathic arthritis (n = 15), and pulmonary The ECM consists of several collagens including type I, II, III and IV
artery hypertension (n = 23). Here, higher levels of antibodies against collagens and when degraded by proteases, it generates new collagen
PPM1A were detected in AS patients compared to the other autoimmune metabolites fragments called neoepitopes, which have been suggested as
diseases. Moreover, in AS patients with grade 3 and 4 radiographic useful future serological biomarkers of SpA [206].
sacroiliitis, higher levels of anti-PPM1A autoantibody were observed The neoepitope biomarker of type I collagen (C1M) is associated with
compared to those with grade 2 radiographic sacroiliitis. Furthermore, destruction of soft tissue and high levels have been observed in patients
the AS patients were treated with TNF inhibitor which resulted in a with AS [207]. The neoepitope biomarker of type II collagen (C2M)
significant decrease in the serum levels of anti-PPM1A, which also reflects the degradation of cartilage [208], and metabolite of type III
showed to correlate positively with degree of sacroiliitis in AS by the collagen is linked to degradation of soft tissue (C3M). Elevated levels of
Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) score. C2M and C3M have been observed in patients with AS compared to
PPM1A was also found to be highly expressed in synovial tissue taken healthy subjects [207]. In addition, C3M has been found in inflamed
from AS patients and driving osteoblast differentiation, which suggests tissue of liver fibrosis [209]. The neoepitope biomarker of type IV
that PPM1A may participate in the pathogenesis of AS. In Korean pa­ collagen (C4M) has also been found elevated in soft tissue destruction
tients, significantly higher levels of anti-PPM1A were also observed in [210]. Recently, C1M, C2M, and C3M have been related with the
AS patients (n = 45) compared to RA patients (n = 20), and healthy response to biologic therapy in AS [206] and have shown correlation
controls (n = 30). In rats, transgenic for HLA-B27 and human β2- with CRP, ESR, and radiographic severity [211]. Thus, it has been pro­
microglobulin, anti-PPM1A levels were also increased in sera [198]. posed that the pathological process in AS involves a higher ECM turn­
Despite these remarkable observations and the potential association over with elevated levels of MMP-cleaved collagen products. In a study
with SpA pathogenesis, more research is needed to examine PPM1A as a by Hušáková et al. C1M, C2M, C3M and C4M were quantified by ELISA
diagnostic biomarker. in serum from AS (n = 72) and nr-axSpA (n = 121) and controls (n =
100). All collagen metabolite biomarkers showed elevated levels in
7.4. miRNA axSpA patients compared to the controls. Furthermore, the serum levels
of C1M, C3M and C4M were significant higher in AS compared to nr-
Diagnostic biomarker candidates have been considered from gene axSpA. The best biomarker to distinguish between AS and nr-axSpA
expression of transcriptomic markers. A meta-analysis performed on was C3M, but more investigation of C3M is needed before validation
gene expression arrays discovered 905 cases of differentially expressed as a diagnostic tool within axSpA. Both C1M, C3M, and C4M were
genes in axSpA patients compared to healthy subjects. Here, the most correlated with ASDAS-CRP in AS and nr-axSpA reflecting association
significantly expressed genes represented pathways linked to antigen with disease activity. Altogether, the current study showed that sero­
processing and presentation [199]. In another report, RNA sequence logical MMP-mediated metabolites from collagen may have potential as
analysis revealed upregulated expression of microRNA (miR)-146a-5p, novel disease activity biomarkers in axSpA however, more compre­
miR-125a-5p, miR-151a-3p and miR-22-3p, and downregulated hensive studies are needed to confirm these results [212]. In another
expression of miR-150-5p and miR-451a in AS (n = 53) compared to study, Gudmann et al. measured the same serological neoepitope bio­
healthy subjects (n = 57). Furthermore, syndesmophyte development in markers, C1M and C3M, in serum from patients with axSpA (n = 110),
patients with AS was associated with the expression of miR-125a-5p, PsA (n = 101), and healthy controls (n = 120). The C1M and C3M levels
miR-151a-3p, miR-150-5p and miR-451a [200]. Another miRNA, miR- were both significantly elevated in axSpA and PsA patients compared to
10b-5p, has shown to be a Th17 regulator present in Th17 cells from controls. Furthermore, C1M and C3M correlated with ASDAS and Dis­
AS patients. Thus, miR-10b-5p decreases the IL-17A production by ease Activity Score 28 (DAS28). In summary, this study demonstrated
inhibiting the mitogen-activated protein kinase 7 (MAP3K7) in CD4+ T that C1M and C3M could be used to distinguish between diseased and
lymphocytes [201]. Other miRNAs found in diseased tissue of hip liga­ healthy subjects and the increase in biomarker levels was associated
ments from AS patients showed differential expression of 22 miRNAs with disease activity in axSpA and PsA [213]. In addition, Siebuhr et al.
from AS patients compared to health controls [202]. Altogether, these measured C1M, C2M and C3M including CRP, ESR, CRPM in serum from
miRNAs could be attractive promising biomarker candidates for AS AS patients during their first year of treatment with etanercept, a TNF
diagnosis which may help to clarify the disease pathogenesis, however, inhibitor. Study showed that ESR, CRP, BASDAI and C1M were signifi­
further validation is required. cantly decreased with treatment. Furthermore, C1M and CRP showed to
predict treatment efficacy. Overall, this study demonstrated that quan­
7.5. CRPM tification of acute inflammation and connective tissue remodelling can
determine which AS patients will most likely benefit from the etanercept
The MMP-cleaved protein fragment of CRP (CRPM) has been asso­ treatment [206].
ciated with local inflammation [203] and appears to be a more specific Besides measuring tissue biomarkers from collagen degradation,
inflammation biomarker for AS compared to measuring the full-length PRO-C2, a marker of mature cartilage collagen type IIB formation, and
CRP [204]. CRPM was measured in serum from axSpA patients (nr- C-Col10, a marker of type X collagen turnover exclusively expressed by
axSpA = 121, AS = 72) and CRPM levels were higher compared to hypertrophic chondrocytes, were measured in 2016 by Gudmann et al.
asymptomatic controls (n = 100). In addition, CRPM levels were The study aimed to investigate the cartilage metabolism in axSpA (n =
increased in AS patients compared to nr-axSpA, indicating that CRPM 110) and PsA (n = 101) patients by measuring and evaluating the
reflects axSpA associated inflammation. CRPM correlated with ASDAS- diagnostic utility of PRO-C2 and C-Col10. The PRO-C2 levels showed to
CRP in AS patients and with CRP in nr-axSpA patients. In summary, be significantly increased in serum samples of both axSpA and PsA
this study indicated that the CRPM biomarker seems to have potential as compared to healthy subjects. Likewise, C-Col10 was significantly

9
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

BIOMARKER DESCRIPTION OF BIOMARKER PARTICIPANTS PRIMARY STUDY OUTCOMES REFS

DIAGNOSTIC BIOMARKERS
192
ANTI-CD74 Human lymphocyte angen P=216, C=325 Anbody reacvity against CD74 in 56 %
(HLA) class II gamma chain of axSpA paents.
(CD74), involved in the Anbody reacvity against CLIP domain
assembly and transport of of the CD74 protein in 67 % of SpA
human histocompability paents.
complex class II (MHC II)
molecules. 193
P=94, C=51 Anbody reacvity against CD74 in 85.1
% of 94 axSpA paents.
198
ANTI-PPM1A Protein phosphatase P=16, C=99 Anbody reacvity against PPM1A in AS
magnesium-dependent 1A paents was higher compared to other
(PPM1A), parcipate in the autoimmune diseases. Higher levels of
formaon of bone including an-PPM1A autoanbody were
syndesmophytes by regulang observed in AS paents with severe
the BMP and Wnt signalling radiographic sacroiliis compared to
pathways, including suppressing those with mild radiographic sacroiliis.
TGF-beta signalling.
200
MICRO-RNAS microRNAs (miRNAs), a group of P=53, C=57 Upregulated expression of (miR)-146a-
single-stranded noncoding 5p, miR-125a-5p, miR-151a-3p and miR-
RNAs. miRNA has been 22-3p, and lower expression of miR-
appreciated to be important in 150-5p, and miR-451a were observed in
pathologies systemic rheumac AS. miR-125a-5p, miR-151a-3p, miR-
diseases and inflammaon by 150-5p and miR-451a were associated
regulang the post- with development of syndesmophytes
transcriponal control of gene in paents with AS.
expression. 201
P=4, C=4 miR-10b-5p showed to be a Th17
regulator present in Th17 cells from AS
paents.
DISEASE ACTIVITY
185
CRP C-reacve protein (CRP), acute P=210 Elevated levels of CRP were a strong
phase reactant. Widely used in predictor of radiographic sacroiliis
the clinic. Elevated levels of progression in AS and nr-axSpA
CRP are a direct indicator of paents.
186
inflammaon and is used as a P=178 An associaon was observed between
predictor of clinical response to disease acvity parameters (ASDAS,
an-TNF treatment. BASDAI and CRP) and radiographic
spinal progression in paents with early
AS.
187
P=201, C=78 AS paents treated with infliximab
resulted in significant reducon in CRP
levels compared to placebo.

Fig. 6. Summary of potential biomarkers for diagnosis, monitoring disease activity, treatment response, and prognostics in patients with spondyloarthritis. P, Pa­
tients; C, Controls.

increased in PsA patients compared to healthy subjects, and a similar increased rate of ossification which is seen by the increase in C-Col10
trend was seen in axSpA patients. Interestingly, a small group of axSpA levels reflecting hypertrophic chondrocytes. In conclusion, these find­
patients (n = 34) naïve to anti-TNF therapy were significantly lower in ings suggest that PRO-C2 and C-Col10 may be promising markers for
C-Col10 levels compared to the axSpA patients undergoing TNF inhib­ investigating the cartilage collagen metabolism in SpA patients [214].
iting treatment. This supports the previously mentioned theory that
resolved inflammation as a result of TNF inhibiting treatment leads to an

10
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

188
P=60 Treatment with either infliximab,
etanercept or adalimumab, gave a
decrease in MRI inflammaon scores
and CRP levels in SpA paents.
Formaon of syndesmophytes was
associated with larger decrease in CRP.
189
P=155 Elevated baseline CRP levels was
observed in AS paents treated with
either etanercept or infliximab.
205
CRPM MMP-mediated metabolite of P=193, C=100 CRPM levels were higher in axSpA and
CRP associated with local increased in AS compared to nr-axSpA,
inflammaon. indicang that CRPM reflects axSpA
associated inflammaon. CRPM
correlated with ASDAS-CRP in AS
paents and with CRP in nr-axSpA
paents.
212
COLLAGEN MMP-degraded type I, II, III, IV P=193, C=100 Increased concentraons of C1M, C2M,
METABOLITE collagen markers, C1M, C2M, C3M and C4M were measured in axSpA.
S C3M, and C4M. Both C1M, C3M, and C4M correlated
The extracellular matrix with ASDAS-CRP.
remodelling is altered in joint 213
P=211, C=120 Increased concentraons of C1M and
degenerave diseases. C3M in PsA and axSpA paents. C1M
and C3M correlated with ASDAS and
DAS28.
206
P=22 AS paents treated with etanercept
showed significant decreased levels of
C1M in response to treatment.
214
Type II collagen formaon P=211, C=118 The PRO-C2 was significantly increased
markers, PRO-C2 and PRO-C10. in axSpA and PsA compared to healthy
subjects. Likewise, C-Col10 was
significantly increased in PsA compared
to healthy subjects, and similar trend
was seen in axSpA paents. axSpA
paents naïve to an-TNF therapy were
significantly lower in C-Col10 levels
compared to axSpA paents undergoing
TNF inhibiting treatment.
DISEASE PROGRESSION
215
SCLEROSTIN Wnt signalling inhibitor, P=204, C=80 Lower sclerosn levels were found in AS
suppresses bone formaon. serum and correlated with high CRP
levels.
216
P=46, C=50 Low sclerosn serum levels have been
found in paents with AS and

Fig. 6. (continued).

7.7. Markers of bone remodelling expression in patients with AS compared to healthy controls [215,216].
Low serum levels of sclerostin have been related to the development of
As previously mentioned, the Wnt/β-catenin signalling pathway syndesmophytes in AS patients [216]. AS patients treated with anti-TNF
including its inhibitors, Dkk proteins and sclerostin, are believed to be therapy showed gradually increased levels of sclerostin over 12 months,
involved in the pathogenesis of SpA. Dkk1 and sclerostin have been while the sclerostin levels stayed substantially lower in AS patients
suggested as markers, however, conflicting data exists with regards to compared to controls [217]. Higher baseline CRP levels were observed
bone formation, Dkk1 and sclerostin correlation to disease progression. in AS patients with lower serum sclerostin. Hereof, AS patients with low
Sclerostin has shown to be significantly lower in serum and in local serum sclerostin had increased risk of having high CRP levels after 12

11
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

associated with the formaon of new


syndesmophytes.
217
P=30, C=30 Low baseline levels of sclerosn in
serum are associated with high CRP
levels in AS paents receiving an-TNF
therapy.
215
DKK1 Dickkopf-1 (Dkk1). Wnt P=204, C=80 Low Dkk1 levels are correlated with
signalling inhibitor, affecng high CRP levels in AS paents.
165
bone formaon where P=65 Higher levels of Dkk1 in paents with no
dysregulaon of Dkk1 leads to formaon of syndesmophyte compared
bone pathologies seen in AS. to those with clear syndesmophytes
progression.
PROGNOSTIC BIOMARKERS
221
VISFATIN Adipokine involved in bone P=86, C=25 Visfan levels were higher in AS
metabolism, shown to promote paents than in healthy subjects.
angiogenesis.
222
LEPTIN Adipokine involved in bone P=120 Serum levels of lepn are inversely
metabolism. related to radiographic spinal growth in
paents with AS.
223
P=42, C=42 Higher serum lepn levels in men with
AS. Lepn levels correlated with serum
IL-6 levels and BASDAI.
225
MIF Macrophage migraon P=147, C=61 Serum levels of MIF were elevated in AS
inhibitory factor (MIF), paents, parcularly in paents with
proinflammatory cytokine. disease progression and with appearing
Proposed to promote syndesmophytes. Increased levels of
mineralisaon of osteoblasts. MIF were found in synovial fluid
collected from inflamed peripheral
joints of paents with AS.

Fig. 6. (continued).

months of treatment compared to patients with higher baseline levels of of visfatin have shown to predict radiographic spinal damage progres­
sclerostin [215,217]. These results may indicate that baseline levels of sion in AS over 2 years [221]. Leptin, another adipokine, together with
sclerostin could serve treatment predictive potential of anti-TNF therapy high molecular weight form of adiponectin (HMW-APN) showed to
and most likely other types of treatment. Serum levels of Dkk1 have significantly predict protection from spinal radiographic progression in
shown to be lower in AS patients compared to healthy controls AS patients. In general, females with AS displayed higher levels of leptin
[145,218]. In AS patients with no syndesmophyte growth, the Dkk1 and HMW-APN compared to males, which may explain the decreased
levels were significantly higher compared to patients with formation of structural damage in the spine compared to male patients with AS [222].
syndesmophytes over 2 years. These results suggest that high serum In addition, serum leptin levels were higher in men with AS compared to
levels of Dkk1 are protective by reducing Wnt signalling and inhibiting healthy male controls and significantly correlated with serum IL-6 levels
new bone growth, subsequently syndesmophyte growth and spinal and BASDAI [223]. From these results, leptin might have a protective
ankylosis [165]. Thus, low serum levels of sclerostin and Dkk1 may effect by inhibition of the extensive aberrant bone formation and a
explain the syndesmophyte growth and ankyloses. However, conflicting possible role in the inflammatory reactions of AS.
data exists of the Dkk1 and sclerostin levels. In contrast to the low levels
of Dkk1 observed in AS patients, in another study high levels of serum 7.9. MIF
Dkk1 were found in AS patients compared to healthy subjects including
other types of arthritis. Furthermore, in a study employing meta-analysis Macrophage migration inhibitory factor (MIF) is a pro-inflammatory
the serum levels of sclerostin in AS patients were not significantly cytokine which has previously been shown to be elevated in serum from
different to controls [219]. More studies are needed before knowing AS patients [224]. In a study by Ranganathan et al. serum levels of MIF
whether sclerostin and Dkk1 levels can be used as potential biomarkers were significantly elevated in AS patients compared to healthy controls,
for structural progression in patients with AS. particularly in patients with disease progression and with appearing
syndesmophytes. In addition, increased levels of MIF were found in
7.8. Adipokines synovial fluid collected from inflamed peripheral joints of patients with
AS. Furthermore, MIF have showed to increase the TNF-α secretion by
The adipokine, visfatin, is released by various cells and can affect monocytes and mediated stabilisation of β-catenin and promoted min­
bone metabolism by directly activating osteoblasts and promoting eralisation of osteoblasts. Thus, MIF has been proposed to have a direct
angiogenesis [220]. In a study by Syrbe et al., increased baseline levels role on the mineralisation of osteoblasts [225]. More studies are needed

12
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

to investigate the prognostic biomarker potential of MIF. clinical trials, and the comparison of the Caucasian and Chinese population. Eur J
Orthop Surg Traumatol 2013;23:497–506.
Although these biomarkers may have potential value in axSpA, most
[13] Van Der Heijde D, et al. Efficacy and safety of adalimumab in patients with
of these studies have not yet been reproduced or evaluated in prospec­ ankylosing spondylitis: Results of a multicenter, randomized, double-blind,
tive studies for the validation as gold standard biomarkers (Fig. 6). placebo-controlled trial. Arthritis Rheum 2006;54:2136–46.
[14] Van Der Heijde D, et al. Efficacy and safety of infliximab in patients with
ankylosing spondylitis: Results of a randomized, placebo-controlled trial
8. Conclusion (ASSERT). Arthritis Rheum 2005;52:582–91.
[15] Haroon N, et al. The impact of tumor necrosis factor α inhibitors on radiographic
progression in ankylosing spondylitis. Arthritis Rheum 2013;65:2645–54.
TNF inhibition has long been the only targeted approach to treat
[16] Xueyi L, et al. Levels of circulating Th17 cells and regulatory T cells in ankylosing
SpA, showing good efficacy in a fraction of SpA patients yielding mixed spondylitis patients with an inadequate response to antiTNF-α therapy. J Clin
results in preventing ossification of damaged bone or cartilage [226]. Immunol 2013;33:151–61.
[17] Lories RJ, Haroon N. Bone formation in axial spondyloarthritis. Best Pract Res
This situation has created an emerging need to discover new treatment
Clin Rheumatol 2014;28:765–77.
targets for development of novel therapies for axSpA patients. Conse­ [18] Sikora KA, Layh-Schmitt G, Colbert RA. Etiology and pathogenesis of
quently, the role of IL-17 in inflammatory process and structural damage spondyloarthritis. Kelley Firestein’s Textbook Rheumatol 2017;74:1245–55. e4.
in axSpA has opened new opportunities to treat SpA. Secukinumab, the [19] Milanez FM, et al. IL-23/Th17 axis is not influenced by TNF-blocking agents in
ankylosing spondylitis patients. Arthritis Res Ther 2016;18:52.
first IL-17 inhibiting agent approved for treatment of axSpA patients, has [20] Le Goff B, et al. Implication of IL-17 in bone loss and structural damage in
showed high clinical efficacy. Subsequently, several other IL-17 block­ inflammatory rheumatic diseases. Mediators Inflamm 2019;2019:8659302.
ing agents such as ixekizumab, brodalumab, bimekizumab, netakimab, [21] Kampylafka E, et al. Disease interception with interleukin-17 inhibition in high-
risk psoriasis patients with subclinical joint inflammation - Data from the
remtolumab, and COVA322 are licensed or under development and prospective IVEPSA study. Arthritis Res Ther 2019;21:178.
expected to be available for treatment in the next couple of years due to [22] Kampylafka E, et al. Resolution of synovitis and arrest of catabolic and anabolic
positive results observed of phase II/III trials. The introduction of anti- bone changes in patients with psoriatic arthritis by IL-17A blockade with
secukinumab: Results from the prospective PSARTROS study. Arthritis Res Ther
IL-17 therapy has expanded the number of options for targeting cyto­ 2018;20:153.
kine pathways in SpA beyond TNF and marked a step forward in the [23] Servier Medical Art. Bones Archives. Available at: https://smart.servier.com/cate
management of SpA. However, despite the positive results of IL-17 in­ gory/anatomy-and-the-human-body/locomotor-system/bones/; 2020 (Accessed:
18th January 2020).
hibitors on the symptoms which are mostly related to pain of SpA, only
[24] Danve A, O’Dell J. The ongoing quest for biomarkers in Ankylosing Spondylitis.
limited data exist on their effect on such drugs on the inflammatory Int J Rheum Dis 2015;18:826–34.
disease process in itself and the related structural damage, especially in [25] Dubin PJ, Kolls JK. Interleukin-17A and Interleukin-17F: a tale of two cytokines.
Immunity 2009;30:9–11.
axial disease. To approach these challenges, a search for objective bio­
[26] Akimzhanov AM, Yang XO, Dong C. Chromatin remodeling of interleukin-17 (IL-
markers reflecting disease activity including dynamic structural pro­ 17)-IL-17F cytokine gene locus during inflammatory helper T cell differentiation.
gression are of key interest. Moreover, combinations of biomarkers J Biol Chem 2007;282:5969–72.
together with imaging data may provide a future analysis platform for [27] Amatya N, Garg AV, Gaffen SL. IL-17 Signaling: The Yin and the Yang. Trends
Immunol 2017;38:310–22.
novel drug discovery, improvement of clinical trial design, and patient [28] Monin L, Gaffen SL. Interleukin 17 family cytokines: signaling mechanisms,
inclusion when testing new treatments, which can aid in the improve­ biological activities, and therapeutic implications. Cold Spring Harb Perspect Biol
ment of individualised care. 2018;10:a028522.
[29] Seon HC, Park H, Dong C. Act1 adaptor protein is an immediate and essential
signaling component of interleukin-17 receptor. J Biol Chem 2006;281:35603–7.
[30] Chyuan IT, Chen JY. Role of interleukin- (IL-) 17 in the pathogenesis and targeted
Declaration of Competing Interest therapies in spondyloarthropathies. Mediators Inflamm 2018;2018(2403935).
[31] Gaffen SL. An overview of IL-17 function and signaling. Cytokine 2008;43:402–7.
Solveig Skovlund Groen, Dovile Sinkeviciute, Simon Francis Thom­ [32] Onishi RM, Gaffen SL. Interleukin-17 and its target genes: mechanisms of
interleukin-17 function in disease. Immunology 2010;129:311–21.
sen, Georg Schett have no competing interest. Morten A. Karsdal, Anne- [33] Shen F, Gaffen SL. Structure-function relationships in the IL-17 receptor:
Christine Bay-Jensen, Christian S. Thudium and Signe Holm Nielsen are implications for signal transduction and therapy. Cytokine 2008;41:92–104.
employees of Nordic Bioscience. Morten A. Karsdal and Anne-Christine [34] Sun D, et al. Treatment with IL-17 prolongs the half-life of chemokine CXCL1
mRNA via the adaptor TRAF5 and the splicing-regulatory factor SF2 (ASF). Nat
Bay-Jensen hold stock in Nordic Bioscience.
Immunol 2011;12:853–60.
[35] McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL-17A
References in axial spondyloarthritis and psoriatic arthritis: recent advances and
controversies. Ann Rheum Dis 2019;78:1167–78.
[36] Swaidani S, Liu C, Zhao J, Bulek K. Li, X. TRAF regulation of IL-17 cytokine
[1] Stolwijk C, van Onna M, Boonen A, van Tubergen A. Global prevalence of
signaling. Front Immunol 2019;10:1293.
spondyloarthritis: a systematic review and meta-regression analysis. Arthritis
[37] Zepp JA, et al. Cutting edge: TNF receptor-associated factor 4 restricts IL-
Care Res 2016;68:1320–31.
17–mediated pathology and signaling processes. J Immunol 2012;189:33–7.
[2] Simone D, Al Mossawi MH, Bowness P. Progress in our understanding of the
[38] Zhu S, et al. Modulation of experimental autoimmune encephalomyelitis through
pathogenesis of ankylosing spondylitis. Rheumatology (Oxford) 2018;57:vi4–9.
TRAF3-mediated suppression of interleukin 17 receptor signaling. J Exp Med
[3] Bodis G, Toth V, Schwarting A. Role of human leukocyte antigens (HLA) in
2010;207:2647–62.
autoimmune diseases. Methods Mol Biol 2018;1802:11–29.
[39] McGeachy MJ, Cua DJ, Gaffen SL. The IL-17 family of cytokines in health and
[4] Machado P, et al. Ankylosing spondylitis patients with and without psoriasis do
disease. Immunity 2019;50:892–906.
not differ in disease phenotype. Ann Rheum Dis 2013;72:1104–7.
[40] Ishigame H, et al. Differential roles of Interleukin-17A and -17F in host defense
[5] Lipton S, Deodhar A. The new ASAS classification criteria for axial and peripheral
against mucoepithelial bacterial infection and allergic responses. Immunity 2009;
spondyloarthritis: promises and pitfalls. Int J Clin Rheumatol 2012;7:675–82.
30:108–19.
[6] Raychaudhuri SP, Deodhar A. The classification and diagnostic criteria of
[41] Cho JS, et al. IL-17 is essential for host defense against cutaneous Staphylococcus
ankylosing spondylitis. J Autoimmun 2014;48-49:128–33.
aureus infection in mice. J Clin Invest 2010;120:1762–73.
[7] Aouad K, Ziade N, Baraliakos X. Structural progression in axial spondyloarthritis.
[42] Aujla SJ, Dubin PJ, Kolls JK. Th17 cells and mucosal host defense. Semin
Joint Bone Spine 2019;87:131–6.
Immunol 2007;19:377–82.
[8] Danve A, Deodhar A. IL-17 Inhibition in Axial Spondyloarthritis. Curr Treatm
[43] Garraud K, et al. Differential role of the interleukin-17 axis and neutrophils in
Options Rheumatol 2015;1:221–30.
resolution of inhalational anthrax. Infect Immun 2012;80:131–42.
[9] Akkoc N. Are spondyloarthropathies as common as rheumatoid arthritis
[44] Ye P, et al. Requirement of interleukin 17 receptor signaling for lung CXC
worldwide? A review. Curr Rheumatol Rep 2008;10:371–8.
chemokine and granulocyte colony-stimulating factor expression, neutrophil
[10] Landewé R, et al. Efficacy of certolizumab pegol on signs and symptoms of axial
recruitment, and host defense. J Exp Med 2001;194:519–27.
spondyloarthritis including ankylosing spondylitis: 24-week results of a double-
[45] Chabaud M, Page G, Miossec P. Enhancing effect of IL-1, IL-17, and TNF-α on
blind randomised placebo-controlled Phase 3 study. Ann Rheum Dis 2014;73:
Macrophage Inflammatory Protein-3α production in rheumatoid arthritis:
39–47.
regulation by soluble receptors and Th2 Cytokines. J Immunol 2001;167:
[11] Inman RD, et al. Efficacy and safety of golimumab in patients with ankylosing
6015–20.
spondylitis: Results of a randomized, double-blind, placebo-controlled, phase III
[46] Pfaff CM, Marquardt Y, Fietkau K, Baron JM, Lüscher B. The psoriasis-associated
trial. Arthritis Rheum 2008;24:49–54.
IL-17A induces and cooperates with IL-36 cytokines to control keratinocyte
[12] Li ZH, Zhang Y, Wang J, Shi ZJ. Etanercept in the treatment of ankylosing
differentiation and function. Sci Rep 2017;7:15631.
spondylitis: A meta-analysis of randomized, double-blind, placebo-controlled

13
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

[47] Chiricozzi A, et al. Integrative responses to IL-17 and TNF-α in human [79] Zhong L, Wang W, Song H. Complex role of IL-23R polymorphisms on ankylosing
keratinocytes account for key inflammatory pathogenic circuits in psoriasis. spondylitis: a meta-analysis. Expert Rev Clin Immunol 2018;14:635–43.
J Invest Dermatol 2011;131:677–87. [80] Nair RP, et al. Genome-wide scan reveals association of psoriasis with IL-23 and
[48] Muromoto R, et al. IL-17A plays a central role in the expression of psoriasis NF-κB pathways. Nat Genet 2009;41:199–204.
signature genes through the induction of IκB-ζ in keratinocytes. Int Immunol [81] Duerr RH, et al. A genome-wide association study identifies IL23R as an
2016;28:443–52. inflammatory bowel disease gene. Science 2006;314:1461–3.
[49] Nograles KE, et al. Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct [82] Evans DM, et al. Interaction between ERAP1 and HLA-B27 in ankylosing
inflammatory and keratinocyte-response pathways. Br J Dermatol 2008;159: spondylitis implicates peptide handling in the mechanism for HLA-B27 in disease
1092–102. susceptibility. Nat. Genet. 2011;43:761–7.
[50] Koshy PJ, et al. Interleukin 17 induces cartilage collagen breakdown: Novel [83] Cortes A, et al. Identification of multiple risk variants for ankylosing spondylitis
synergistic effects in combination with proinflammatory cytokines. Ann Rheum through high-density genotyping of immune-related loci. Nat Genet 2013;45:
Dis 2002;61:704–13. 730–8.
[51] Shalom-Barak T, Quach J, Lotz M. Interleukin-17-induced gene expression in [84] Sherlock JP, et al. IL-23 induces spondyloarthropathy by acting on ROR-γt + CD3
articular chondrocytes is associated with activation of mitogen-activated protein + CD4 - CD8 - entheseal resident T cells. Nat Med 2012;18:1069–76.
kinases and NF-κB. J Biol Chem 1998;273:27467–73. [85] Nakae S, Nambu A, Sudo K, Iwakura Y. Suppression of immune induction of
[52] Honorati MC, Bovara M, Cattini L, Piacentini A, Facchini A. Contribution of collagen-induced arthritis in IL-17-deficient mice. J Immunol 2003;171:6173–7.
interleukin 17 to human cartilage degradation and synovial inflammation in [86] Koenders MI, et al. Interleukin-17 receptor deficiency results in impaired synovial
osteoarthritis. Osteoarthr Cartil 2002;10:799–807. expression of interleukin-1 and matrix metalloproteinases 3, 9, and 13 and
[53] Ha H-L, et al. IL-17 drives psoriatic inflammation via distinct, target cell-specific prevents cartilage destruction during chronic reactivated streptococcal cell wall-
mechanisms. Proc Natl Acad Sci 2014;111:E3422–31. induced arthritis. Arthritis Rheum 2005;52:3239–47.
[54] Hwang SY, et al. IL-17 induces production of IL-6 and IL-8 in rheumatoid arthritis [87] Mei Y, et al. Increased serum IL-17 and IL-23 in the patient with ankylosing
synovial fibroblasts via NF-kappaB- and PI3-kinase/Akt-dependent pathways. spondylitis. Clin Rheumatol 2011;30:269–73.
Arthritis Res Ther 2004;6:R120–8. [88] Wendling D, Cedoz JP, Racadot E, Dumoulin G. Serum IL-17, BMP-7, and bone
[55] Zhang JR, et al. Different modulatory effects of IL-17, IL-22, and IL-23 on turnover markers in patients with ankylosing spondylitis. Joint Bone Spine 2007;
osteoblast differentiation. Mediators Inflamm 2017;2017:5950395. 74:304–5.
[56] Song L, et al. Interleukin-17A facilitates osteoclast differentiation and bone [89] Chen WS, et al. Association of serum interleukin-17 and interleukin-23 levels with
resorption via activation of autophagy in mouse bone marrow macrophages. Mol disease activity in Chinese patients with ankylosing spondylitis. J Chin Med Assoc
Med Rep 2019;19:4743–52. 2012;75:303–8.
[57] Griffin GK, et al. IL-17 and TNF- Sustain Neutrophil Recruitment during [90] Jandus C, et al. Increased numbers of circulating polyfunctional Th17 memory
Inflammation through Synergistic Effects on Endothelial Activation. J Immunol cells in patients with seronegative spondylarthritides. Arthritis Rheum 2008;58:
2012;188:6287–99. 2307–17.
[58] Yuan S, et al. Interleukin-17 stimulates STAT3-mediated endothelial cell [91] Menon B, et al. Interleukin-17+CD8+ T cells are enriched in the joints of patients
activation for neutrophil recruitment. Cell Physiol Biochem 2015;36:2340–56. with psoriatic arthritis and correlate with disease activity and joint damage
[59] Hot A, Lenief V, Cazalis M-A, Miossec P. Pathogenic role of IL-17 in endothelial progression. Arthritis Rheumatol (Hoboken NJ) 2014;66:1272–81.
dysfunction, a link between rheumatoid arthritis and atherosclerosis. Ann Rheum [92] Lowes MA, et al. Psoriasis vulgaris lesions contain discrete populations of Th1 and
Dis 2010;69. A44 LP-A45. Th17 T cells. J Invest Dermatol 2008;128:1207–11.
[60] Barin JG, et al. Macrophages participate in IL-17-mediated inflammation. Eur J [93] Noordenbos T, et al. Interleukin-17-positive mast cells contribute to synovial
Immunol 2012;42:726–36. inflammation in spondylarthritis. Arthritis Rheum 2012;64:99–109.
[61] Chen J, et al. IL-17A induces pro-inflammatory cytokines production in [94] Appel H, et al. Analysis of IL-17+ cells in facet joints of patients with
macrophages via MAPKinases, NF-κB and AP-1. Cell Physiol Biochem 2013;32: spondyloarthritis suggests that the innate immune pathway might be of greater
1265–74. relevance than the Th17-mediated adaptive immune response. Arthritis Res Ther
[62] Nakai K, et al. IL-17A induces heterogeneous macrophages, and it does not alter 2011;13:R95.
the effects of lipopolysaccharides on macrophage activation in the skin of mice. [95] Ciccia F, et al. Overexpression of interleukin-23, but Not interleukin-17, as an
Sci Rep 2017;7:1–14. immunologic signature of Subclinical intestinal inflammation in ankylosing
[63] Jovanovic DV, et al. IL-17 stimulates the production and expression of spondylitis. Arthritis Rheum 2009;60:955–65.
proinflammatory cytokines, IL-beta and TNF-alpha, by human macrophages. [96] Appel H, et al. In situ analysis of interleukin-23- and interleukin-12-positive cells
J Immunol 1998;160:3513–21. in the spine of patients with ankylosing spondylitis. Arthritis Rheum 2013;65:
[64] Liu L, et al. Effect of pro-inflammatory interleukin-17A on epithelial cell 1522–9.
phenotype inversion in HK-2 cells in vitro. Eur Cytokine Netw 2016;27:34–40. [97] Bowness P, et al. Th17 cells expressing KIR3DL2+ and responsive to HLA-B27
[65] Noack M, Ndongo-Thiam N, Miossec P. Interaction among activated lymphocytes homodimers are increased in Ankylosing Spondylitis Europe PMC Funders Group.
and mesenchymal cells through podoplanin is critical for a high IL-17 secretion. J Immunol 2011;186:2672–80.
Arthritis Res Ther 2016;18:148. [98] Bautista-caro MB, et al. Decreased Th17 and Th1 cells in the peripheral blood of
[66] Hartupee J, Liu C, Novotny M, Li X, Hamilton T. IL-17 Enhances Chemokine Gene patients with early non-radiographic axial spondyloarthritis: A marker of disease
Expression through mRNA Stabilization. J Immunol 2007;179:4135–41. activity in HLA-B27+ patients. Rheumatol (UK) 2013;52:352–62.
[67] Chabaud M, Miossec P. The combination of tumor necrosis factor α blockade with [99] D.T.S.L.J, et al. IL-17-producing CD4+ T cells are increased in early, active axial
interleukin-1 and interleukin-17 blockade is more effective for controlling spondyloarthritis including patients without imaging abnormalities.
synovial inflammation and bone resorption in an ex vivo model. Arthritis Rheum Rheumatology 2014;52:352–62.
2001;44:1293–303. [100] Kenna TJ, et al. Enrichment of circulating interleukin-17-secreting interleukin-23
[68] Fischer JAA, et al. Combined inhibition of tumor necrosis factor α and receptor-positive γ/δ T cells in patients with active ankylosing spondylitis.
interleukin-17 as a therapeutic opportunity in rheumatoid arthritis: Development Arthritis Rheum 2012;64:1420–9.
and characterization of a novel bispecific antibody. Arthritis Rheumatol 2015;67: [101] Ono T, et al. IL-17-producing γδT cells enhance bone regeneration. Nat Commun
51–62. 2016;7:10928.
[69] Michel ML, et al. Identification of an IL-17-producing NK1.1neg iNKT cell [102] Reinhardt A, et al. Interleukin-23–dependent γ/δ T cells produce Interleukin-17
population involved in airway neutrophilia. J Exp Med 2007;204:995–1001. and accumulate in the enthesis, aortic valve, and ciliary body in mice. Arthritis
[70] Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune Rheumatol 2016;68:2476–86.
system. Nat Rev Immunol 2010;10:479–89. [103] Deodhar A, et al. Three multicenter, randomized, double-blind, placebo-
[71] Zeng B, et al. ILC3 function as a double-edged sword in inflammatory bowel controlled studies evaluating the efficacy and safety of ustekinumab in axial
diseases. Cell Death Dis 2019:10. spondyloarthritis. Arthritis Rheumatol 2019;71:258–70.
[72] Song C, et al. IL-17-Producing alveolar macrophages mediate allergic lung [104] Baeten D, et al. Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis:
inflammation related to asthma. J Immunol 2008;181:6117–24. results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-
[73] Ferretti S, Bonneau O, Dubois GR, Jones CE, Trifilieff A. IL-17, Produced by finding phase 2 study. Ann Rheum Dis 2018;77:1295–302.
Lymphocytes and Neutrophils, Is Necessary for Lipopolysaccharide-Induced [105] Langley RG, et al. Secukinumab in plaque psoriasis - results of two phase 3 trials.
Airway Neutrophilia: IL-15 as a Possible Trigger. J Immunol 2003;170:2106–12. N Engl J Med 2014;371:326–38.
[74] Takahashi N, et al. IL-17 produced by Paneth cells drives TNF-induced shock. [106] Mease P, et al. Secukinumab improves active psoriatic arthritis symptoms and
J Exp Med 2008;205:1755–61. inhibits radiographic progression: primary results from the randomised, double-
[75] Teunissen MBM, et al. The IL-17A-producing CD8 + T-cell population in psoriatic blind, phase III FUTURE 5 study. Ann Rheum Dis 2018;77:890–7.
lesional skin comprises mucosa-associated invariant t cells and conventional t [107] Mease PJ, et al. Secukinumab inhibition of interleukin-17A in patients with
cells. J Invest Dermatol 2014;134:2898–907. psoriatic arthritis. N Engl J Med 2015;373:1329–39.
[76] Steel K, et al. Synovial IL-17+ CD8+ T cells are a pro-inflammatory tissue [108] McInnes IB, et al. Secukinumab, a human anti-interleukin-17A monoclonal
resident population enriched in spondyloarthritis. Ann Rheum Dis 2018;77:A8–9. antibody, in patients with psoriatic arthritis (FUTURE 2): A randomised, double-
[77] Cheuk S, et al. CD49a expression defines tissue-resident CD8+ T cells poised for blind, placebo-controlled, phase 3 trial. Lancet 2015;386:1137–46.
cytotoxic function in human skin. Immunity 2017;46:287–300. [109] Baeten D, et al. Secukinumab, an interleukin-17A inhibitor, in ankylosing
[78] McGeachy MJ, et al. The interleukin 23 receptor is essential for the terminal spondylitis. N Engl J Med 2015;373:2534–48.
differentiation of interleukin 17-producing effector T helper cells in vivo. Nat [110] Thaçi D, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects
Immunol 2009;10:314–24. with moderate to severe plaque psoriasis: CLEAR, a randomized controlled trial.
J Am Acad Dermatol 2015;73:400–9.

14
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

[111] Deodhar A, et al. Efficacy and safety of Ixekizumab in the treatment of contribute to new bone formation in spondyloarthropathies. Rheumatol (U K)
radiographic axial spondyloarthritis: sixteen-week results from a phase III 2017;56:kew384.
randomized, double-blind, placebo-controlled trial in patients with prior [140] Braun Jür, et al. Use of immunohistologic and in situ hybridization techniques in
inadequate response to or intolerance of tumor necrosis factpr inhibitors. Arthritis the examination of sacroiliac joint biopsy specimens from patients with
Rheumatol 2019;71:599–611. ankylosing spondylitis. Arthritis Rheum 1995;38:499–505.
[112] Gordon KB, et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque [141] Lories RJU, Derese I, Luyten FP. Modulation of bone morphogenetic protein
psoriasis. N Engl J Med 2016;375:345–56. signaling inhibits the onset and progression of ankylosing enthesitis. J Clin Invest
[113] Mease PJ, et al. Ixekizumab, an interleukin-17A specific monoclonal antibody, for 2005;115:1571–9.
the treatment of biologic-naive patients with active psoriatic arthritis: Results [142] Lories RJU, Derese I, Ceuppens JL, Luyten FP. Bone morphogenetic proteins 2 and
from the 24-week randomised, double-blind, placebocontrolled and active 6, expressed in arthritic synovium, are regulated by proinflammatory cytokines
(adalimumab)-controlled period of the phase III trial SPRIT-P1. Ann Rheum Dis and differentially modulate fibroblast-like synoviocyte apoptosis. Arthritis Rheum
2017;76:79–87. 2003;48:2807–18.
[114] Van der Heijde D, et al. Ixekizumab, an interleukin-17A antagonist in the [143] Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from
treatment of ankylosing spondylitis or radiographic axial spondyloarthritis in human mutations to treatments. Nat Med 2013;19:179–92.
patients previously untreated with biological disease-modifying anti-rheumatic [144] Glass DA, et al. Canonical Wnt signaling in differentiated osteoblasts controls
drugs (COAST-V): 16 week results of a phase 3 randomised, double-blind, active. osteoclast differentiation. Dev Cell 2005;8:751–64.
Lancet 2018;392:2441–51. [145] Diarra D, et al. Dickkopf-1 is a master regulator of joint remodeling. Nat Med
[115] Nash P, et al. Ixekizumab for the treatment of patients with active psoriatic 2007;13:156–63.
arthritis and an inadequate response to tumour necrosis factor inhibitors: results [146] Baum R, Gravallese EM. Impact of inflammation on the osteoblast in rheumatic
from the 24-week randomised, double-blind, placebo-controlled period of the diseases. Curr Osteoporos Rep 2014;12:9–16.
SPIRIT-P2 phase 3 trial. Lancet 2017;389:2317–27. [147] Sudoł-Szopińska I, Matuszewska G, Kwiatkowska B, Pracoń G. Diagnostic imaging
[116] R.C, et al. The VD2 subset of FDT-cells are present at healthy human enthesis and of psoriatic arthritis. Part I: etiopathogenesis, classifications and radiographic
have transcriptional and functional characteristics consistent with a capacity for features. J Ultrason 2016;16:65–77.
IL-17a production in response to IL-23. Arthritis Rheumatol 2018;70:1833. [148] Schett G, et al. Enthesitis: from pathophysiology to treatment. Nat Rev Rheumatol
[117] McGonagle D, Lories RJU, Tan AL, Benjamin M. The concept of a ‘synovio- 2017;118:108–14.
entheseal complex’ and its implications for understanding joint inflammation and [149] Paine A, Ritchlin C. Bone remodeling in psoriasis and psoriatic arthritis: an
damage in psoriatic arthritis and beyond. Arthritis Rheum 2007;56:2482–91. update. Curr Opin Rheumatol 2016;28:66–75.
[118] Castillo-Gallego C, Aydin SZ, Emery P, McGonagle DG, Marzo-Ortega H. Magnetic [150] Paine A, Ritchlin C. Altered bone remodeling in psoriatic disease: new insights
resonance imaging assessment of axial psoriatic arthritis: extent of disease relates and future directions. Calcif Tissue Int 2018;102:559–74.
to HLA-B27. Arthritis Rheum 2013;65:2274–8. [151] Kocijan R, et al. Quantitative and qualitative changes of bone in psoriasis and
[119] Strand V, Singh JA. Patient burden of axial spondyloarthritis. J Clin Rheumatol psoriatic arthritis patients. J Bone Miner Res 2015;30:1775–83.
2017;23:383–91. [152] Adamopoulos IE, et al. Interleukin-17A upregulates receptor activator of NF-κB on
[120] Goldring SR. Differential mechanisms of de-regulated bone formation in osteoclast precursors. Arthritis Res Ther 2010;12:R29.
rheumatoid arthritis and spondyloarthritis. Rheumatology 2016;55:ii56–60. [153] Van der Heijde D, et al. Brief report: secukinumab provides significant and
[121] Nistala H, Lee-Arteaga S, Siciliano G, Smaldone S, Ramirez F. Extracellular sustained inhibition of joint structural damage in a Phase III study of active
regulation of transforming growth factor β and bone morphogenetic protein psoriatic arthritis. Arthritis Rheumatol 2016;68:1914–21.
signaling in bone. In: Annals of the New York Academy of Sciences. 1192; 2010. [154] Mease PJ, et al. Secukinumab in the treatment of psoriatic arthritis: efficacy and
p. 253–6. safety results through 3 years from the year 1 extension of the randomised phase
[122] Lories RJ, Haroon N. Evolving concepts of new bone formation in axial III FUTURE 1 trial. RMD Open 2018;4:e000723.
spondyloarthritis: insights from animal models and human studies. Best Pract Res [155] Chimenti MS, Maria Morello PC. A prospective study on inflammatory cytokines
Clin Rheumatol 2017;31:877–86. and bone metabolism mediators in patients affected by rheumatoid and psoriatic
[123] Zhou X, et al. Chondrocytes transdifferentiate into osteoblasts in endochondral arthritis treated with Adalimumab. J Arthritis 2015;4(158).
bone during development, postnatal growth and fracture healing in mice. PLoS [156] Finzel S, et al. Bone anabolic changes progress in psoriatic arthritis patients
Genet 2014;10:e1004820. despite treatment with methotrexate or tumour necrosis factor inhibitors. Ann
[124] Gravallese EM, Schett G. Effects of the IL-23–IL-17 pathway on bone in Rheum Dis 2013;72:1176–81.
spondyloarthritis. Nat Rev Rheumatol 2018;14:631–40. [157] Stavre Z, Upchurch K, Kay J, Gravallese EM. Differential effects of inflammation
[125] Kotake S, et al. IL-17 in synovial fluids from patients with rheumatoid arthritis is a on bone and response to biologics in rheumatoid arthritis and spondyloarthritis.
potent stimulator of osteoclastogenesis. J Clin Invest 1999;103:1345–52. Curr Rheumatol Rep 2016:4.
[126] Miossec P. Interleukin-17 in rheumatoid arthritis: If T cells were to contribute to [158] Llopis E, Kroon HM, Acosta J, Bloem JL. Conventional radiology in rheumatoid
inflammation and destruction through synergy. Arthritis Rheum 2003;48: arthritis. Radiol Clin North Am 2017;55:917–41.
594–601. [159] Finzel S, Englbrecht M, Engelke K, Stach C, Schett G. A comparative study of
[127] Van Hamburg JP, et al. Th17 cells, but not Th1 cells, from patients with early periarticular bone lesions in rheumatoid arthritis and psoriatic arthritis. Ann
rheumatoid arthritis are potent inducers of matrix metalloproteinases and Rheum Dis 2011;70:122–7.
proinflammatory cytokines upon synovial fibroblast interaction, including [160] Feld J, Chandran V, Haroon N, Inman R, Gladman D. Axial disease in psoriatic
autocrine interleukin-17A production. Arthritis Rheum 2011;63:73–83. arthritis and ankylosing spondylitis: A critical comparison. Nat Rev Rheumatol
[128] Rossini M, et al. Focal bone involvement in inflammatory arthritis: the role of 2018;14:363–71.
IL17. Rheumatol Int 2016;36:469–82. [161] Grisar J, et al. Ankylosing spondylitis, psoriatic arthritis, and reactive arthritis
[129] Lee Y. The role of interleukin-17 in bone metabolism and inflammatory skeletal show increased bone resorption, but differ with regard to bone formation.
diseases. BMB Rep 2013;46:479–83. J Rheumatol 2002;29:1430–6.
[130] Kim KW, Kim HR, Kim BM, La Cho M, Lee SH. Th17 cytokines regulate [162] Jadon DR, et al. Axial disease in psoriatic arthritis study: defining the clinical and
osteoclastogenesis in rheumatoid arthritis. Am J Pathol 2015;67:1156. radiographic phenotype of psoriatic spondyloarthritis. Ann Rheum Dis 2017;76:
[131] Sato K, et al. Th17 functions as an osteoclastogenic helper T cell subset that links 701–7.
T cell activation and bone destruction. J Exp Med 2006;203:2673–82. [163] Lories RJU, Matthys P, De Vlam K, Derese I, Luyten FP. Ankylosing enthesitis,
[132] Moran EM, et al. Human rheumatoid arthritis tissue production of IL-17A drives dactylitis, and onychoperiostitis in male DBA/1 mice: a model of psoriatic
matrix and cartilage degradation: Synergy with tumour necrosis factor-α, arthritis. Ann Rheum Dis 2004;63:595–8.
Oncostatin M and response to biologic therapies. Arthritis Res. Ther. 2009;11: [164] Fassio A, et al. In psoriatic arthritis Dkk-1 and PTH are lower than in rheumatoid
R113. arthritis and healthy controls. Clin Rheumatol 2017;36:2377–81.
[133] Chabaud M, Lubberts E, Joosten L, Van Den Berg W, Miossec P. IL-17 derived [165] Heiland GR, et al. High level of functional dickkopf-1 predicts protection from
from juxta-articular bone and synovium contributes to joint degradation in syndesmophyte formation in patients with ankylosing spondylitis. Ann Rheum Dis
rheumatoid arthritis. Arthritis Res 2001;3:168–77. 2012;71:572–4.
[134] Liao C, Zhang C, Jin L, Yang Y. IL-17 alters the mesenchymal stem cell niche [166] Jadon DR, et al. Serum soluble bone turnover biomarkers in psoriatic arthritis and
towards osteogenesis in cooperation with osteocytes. J Cell Physiol 2020;235: psoriatic spondyloarthropathy. J Rheumatol 2015;42:21–30.
4466–80. [167] Dalbeth N, et al. Circulating mediators of bone remodeling in psoriatic arthritis:
[135] Huang H, et al. IL-17 stimulates the proliferation and differentiation of human implications for disordered osteoclastogenesis and bone erosion. Arthritis Res.
mesenchymal stem cells: implications for bone remodeling. Cell Death Differ Ther. 2010;12:R164.
2009;16:1332–43. [168] Sanford M, McKeage K. Secukinumab: first global approval. Drugs 2015;75:
[136] Noh M. Interleukin-17A increases leptin production in human bone marrow 329–38.
mesenchymal stem cells. Biochem Pharmacol 2012;83:661–70. [169] Baronaite Hansen R, Kavanaugh A. Secukinumab for the treatment of psoriatic
[137] Osta B, Lavocat F, Eljaafari A, Miossec P. Effects of interleukin-17A on osteogenic arthritis. Expert Rev Clin Immunol 2016;12:1027–36.
differentiation of isolated human mesenchymal stem cells. Front Immunol 2014; [170] Furst DE, Louie JS. Targeting inflammatory pathways in axial spondyloarthritis.
5:425. Arthritis Res Therapy 2019;21:135.
[138] Benham H, et al. Interleukin-23 mediates the intestinal response to microbial [171] Galluzzo M, et al. Spotlight on brodalumab in the treatment of plaque psoriasis:
β-1,3-glucan and the development of spondyloarthritis pathology in SKG mice. the evidence to date. Clin Cosmet Investig Dermatol 2019;12:311–21.
Arthritis Rheumatol 2014;66:1755–67. [172] Kaplon H, Muralidharan M, Schneider Z, Reichert JM. Antibodies to watch in
[139] El-Zayadi AA, et al. Interleukin-22 drives the proliferation, migration and 2020. MAbs 2020;12:1703531.
osteogenic differentiation of mesenchymal stem cells: A novel cytokine that could

15
S.S. Groen et al. Autoimmunity Reviews 20 (2021) 102760

[173] Wasilewska A, Winiarska M, Olszewska M, Rudnicka L. Interleukin-17 inhibitors. [201] Chen L, et al. MiR-10b-5p is a novel Th17 regulator present in Th17 cells from
A new era in treatment of psoriasis and other skin diseases. Postepy Dermatol ankylosing spondylitis. Ann Rheum Dis 2017;76:620–4.
Alergol 2016;33:247–52. [202] Zhang C, et al. Differentially expressed mRNAs, lncRNAs, and miRNAs with
[174] Chiricozzi A, De Simone C, Fossati B, Peris K. Emerging treatment options for the associated co-expression and ceRNA networks in ankylosing spondylitis.
treatment of moderate to severe plaque psoriasis and psoriatic arthritis: Oncotarget 2017;8:113543–57.
evaluating bimekizumab and its therapeutic potential. Psoriasis Targets Ther [203] Siebuhr AS, et al. Identification and characterisation of osteoarthritis patients
2019;9:73–4. with inflammation derived tissue turnover. Osteoarthr Cartil 2014;22:44–50.
[175] A Phase 2b Study of the Efficacy, Safety, and Tolerability of M1095 in Subjects [204] Skjøt-Arkil H, et al. Investigation of two novel biochemical markers of
With Moderate to Severe Psoriasis - Full Text View - ClinicalTrials.gov. Available inflammation, matrix metalloproteinase and cathepsin generated fragments of C-
at: https://clinicaltrials.gov/ct2/show/NCT03384745; 2020 (Accessed: 21st reactive protein, in patients with ankylosing spondylitis. Clin Exp Rheumatol
February 2020). 2012;30:371–9.
[176] Koenders MI, et al. Tumor necrosis factor-interleukin-17 interplay induces [205] Siebuhr AS, et al. Metabolites of C-reactive protein and vimentin are associated
S100A8, interleukin-1β, and matrix metalloproteinases, and drives irreversible with disease activity of axial spondyloarthritis. Clin Exp Rheumatol 2019;37:
cartilage destruction in murine arthritis: rationale for combination treatment 358–66.
during arthritis. Arthritis Rheum 2011;63:2329–39. [206] Siebuhr AS, Bay-Jensen AC, Karsdal MA, Lories RJ, De Vlam K. CRP and a
[177] Springer Nature Switzerland AG. AdisInsight. Available at: https://adisinsight.sp biomarker of type i collagen degradation, C1M, can differentiate anti-
ringer.com/; 2020 (Accessed: 8th April 2020). inflammatory treatment response in ankylosing spondylitis. Biomark Med 2016;
[178] Geven EJW, et al. S100A8/A9, a potent serum and molecular imaging biomarker 10:197–208.
for synovial inflammation and joint destruction in seronegative experimental [207] Bay-Jensen AC, et al. Ankylosing spondylitis is characterized by an increased
arthritis. Arthritis Res Ther 2016;18:247. turnover of several different metalloproteinase-derived collagen species: a cross-
[179] Rudwaleit M, et al. The early disease stage in axial spondylarthritis: results from sectional study. Rheumatol Int 2012;32:3565–72.
the German spondyloarthritis inception cohort. Arthritis Rheum 2009;60:717–27. [208] Bay-Jensen AC, et al. Enzyme-linked immunosorbent assay (ELISAs) for
[180] Dougados M, et al. Clinical relevance of C-reactive protein in axial involvement of metalloproteinase derived type II collagen neoepitope, CIIM-Increased serum
ankylosing spondylitis. J Rheumatol 1999;26:971–4. CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem 2011;44:
[181] Maksymowych WP. Biomarkers for diagnosis of axial spondyloarthritis, disease 423–9.
activity, prognosis, and prediction of response to therapy. Front Immunol 2019; [209] Vassiliadis E, et al. Measurement of co3-610, a potential liver biomarker derived
10:305. from matrix metalloproteinase-9 degradation of collagen type III, in a rat model
[182] Eyre S, Orozco G, Worthington J. The genetics revolution in rheumatology: large of reversible carbon-tetrachloride-induced fibrosis. Biomark Insights 2011;6:
scale genomic arrays and genetic mapping. Nat Rev Rheumatol 2017;13:421–32. 49–58.
[183] Hueber W, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for [210] Sand JM, et al. MMP mediated degradation of type IV collagen alpha 1 and alpha
moderate to severe Crohn’s disease: unexpected results of a randomised, double- 3 chains reflects basement membrane remodeling in experimental and clinical
blindplacebo- controlled trial. Gut 2012;61:1693–700. fibrosis - Validation of two novel biomarker assays. PLoS One 2013;e84934:8.
[184] Prajzlerová K, Grobelná K, Pavelka K, Šenolt L, Filková M. An update on [211] Bay-Jensen AC, et al. Circulating protein fragments of cartilage and connective
biomarkers in axial spondyloarthritis. Autoimmun Rev 2016;15:501–9. tissue degradation are diagnostic and prognostic markers of rheumatoid arthritis
[185] Poddubnyy D, et al. Rates and predictors of radiographic sacroiliitis progression and ankylosing spondylitis. PLoS One 2013;8:e54504.
over 2 years in patients with axial spondyloarthritis. Ann Rheum Dis 2011;70: [212] Hušáková M, et al. Metabolites of type I, II, III, and IV collagen may serve as
1369–74. markers of disease activity in axial spondyloarthritis. Sci Rep 2019;9:11218.
[186] Poddubnyy D, et al. High disease activity according to the Ankylosing Spondylitis [213] Gudmann NS, et al. Type I and III collagen turnover is increased in axial
Disease Activity Score is associated with accelerated radiographic spinal spondyloarthritis and psoriatic arthritis. Associations with disease activity and
progression in patients with early axial spondyloarthritis: results from the German diagnostic capacity. Clin Exp Rheumatol 2017;35:653–9.
SPondyloarthritis Inception Cohort. Ann Rheum Dis 2016;75:2114–8. [214] Gudmann NS, et al. Chondrocyte activity is increased in psoriatic arthritis and
[187] Visvanathan S, et al. Inflammatory biomarkers, disease activity and spinal disease axial spondyloarthritis. Arthritis Res Ther 2016;18:141.
measures in patients with ankylosing spondylitis after treatment with infliximab. [215] Klingberg E, Nurkkala M, Carlsten H, Forsblad-D’Elia H. Biomarkers of bone
Ann Rheum Dis 2008;67:511–7. metabolism in ankylosing spondylitis in relation to osteoproliferation and
[188] Pedersen SJ, et al. Radiographic progression is associated with resolution of osteoporosis. J Rheumatol 2014;41:1349–56.
systemic inflammation in patients with axial spondylarthritis treated with tumor [216] Appel H, et al. Altered skeletal expression of sclerostin and its link to radiographic
necrosis factor α inhibitors: a study of radiographic progression, inflammation on progression in ankylosing spondylitis. Arthritis Rheum 2009;60:3257–62.
magnetic resonance imaging, and c. Arthritis Rheum 2011;63:3789–800. [217] Saad CGS, et al. Low sclerostin levels: a predictive marker of persistent
[189] De Vries MK, et al. Erythrocyte sedimentation rate, C-reactive protein level, and inflammation in ankylosing spondylitis during anti-tumor necrosis factor
serum amyloid a protein for patient selection and monitoring of anti-tumor therapy? Arthritis Res Ther 2012;14:R216.
necrosis factor treatment in ankylosing spondylitis. Arthritis Care Res 2009;61: [218] Kwon SR, et al. Dickkopf-1 level is lower in patients with ankylosing spondylitis
1484–90. than in healthy people and is not influenced by anti-tumor necrosis factor
[190] Cresswell P. Assembly, transport, and function of MHC Class II molecules. Annu therapy. Rheumatol Int 2012;32:2523–7.
Rev Immunol 1994;12:259–93. [219] Shi J, Ying H, Du J, Shen B. Serum sclerostin levels in patients with ankylosing
[191] Leng L, et al. MIF signal transduction initiated by binding to CD74. J Exp Med spondylitis and rheumatoid arthritis: a systematic review and meta-analysis.
2003;197:1467–76. Biomed Res Int 2017;2017:1–7.
[192] Baerlecken NT, et al. Autoantibodies against CD74 in spondyloarthritis. Ann [220] Kim SR, et al. Visfatin promotes angiogenesis by activation of extracellular signal-
Rheum Dis 2014;73:1211–4. regulated kinase 1/2. Biochem Biophys Res Commun 2007;357:150–6.
[193] Baraliakos X, Baerlecken N, Witte T, Heldmann F, Braun J. High prevalence of [221] Syrbe U, et al. Serum adipokine levels in patients with ankylosing spondylitis and
anti-CD74 antibodies specific for the HLA class II-associated invariant chain their relationship to clinical parameters and radiographic spinal progression.
peptide (CLIP) in patients with axial spondyloarthritis. Ann Rheum Dis 2014;73: Arthritis Rheumatol 2015;67:678–85.
1079–82. [222] Hartl A, et al. Serum levels of leptin and high molecular weight adiponectin are
[194] De Winter JJ, et al. Anti-CD74 antibodies have no diagnostic value in early axial inversely associated with radiographic spinal progression in patients with
spondyloarthritis: data from the spondyloarthritis caught early (SPACE) cohort. ankylosing spondylitis: Results from the ENRADAS trial. Arthritis Res Ther 2017;
Arthritis Res Ther 2018;20:38. 19:140.
[195] Liu Y, Liao X, Shi G. Autoantibodies in spondyloarthritis, focusing on anti-CD74 [223] Park MC, Lee SW, Choi ST, Park YB, Lee SK. Serum leptin levels correlate with
antibodies. Front Immunol 2019;10:5. interleukin-6 levels and disease activity in patients with ankylosing spondylitis.
[196] Schett G. Bone formation versus bone: Resorption in ankylosing spondylitis. Adv Scand J Rheumatol 2007;36:101–6.
Exp Med Biol 2009;649:114–21. [224] Kozaci LD, Sari I, Alacacioglu A, Akar S, Akkoc N. Evaluation of inflammation and
[197] Zolnierowicz S. Type 2A protein phosphatase, the complex regulator of numerous oxidative stress in ankylosing spondylitis: a role for macrophage migration
signaling pathways. Biochem Pharmacol 2000;60:1225–35. inhibitory factor. Mod Rheumatol 2010;20:34–9.
[198] Kim YG, et al. Role of protein phosphatase magnesium-dependent 1a and anti- [225] Ranganathan V, et al. Macrophage migration inhibitory factor induces
protein phosphatase magnesium-dependent 1A autoantibodies in ankylosing inflammation and predicts spinal progression in ankylosing spondylitis. Arthritis
spondylitis. Arthritis Rheumatol 2014;10:2793–803. Rheumatol 2017;69:1796–806.
[199] Fang F, Pan J, Xu L, Li G, Wang J. Identification of potential transcriptomic [226] Sieper J, Appel H, Braun J, Rudwaleit M. Critical appraisal of assessment of
markers in developing ankylosing spondylitis: a meta-analysis of gene expression structural damage in ankylosing spondylitis: implications for treatment outcomes.
profiles. Biomed Res Int 2015;2015(826316). Arthritis Rheum 2008;58:649–56.
[200] Perez-Sanchez C, et al. Circulating microRNAs as potential biomarkers of disease
activity and structural damage in ankylosing spondylitis patients. Hum Mol Genet
2018;27:875–90.

16

You might also like