Sunteți pe pagina 1din 12

RevIews

Bedside to bench: defining the


immunopathogenesis of psoriatic
arthritis
Arlene Bravo and Arthur Kavanaugh   *
Abstract | Psoriatic arthritis (PsA) is an immune-​mediated, systemic inflammatory disorder.
PsA can present with heterogeneous clinical features. Advances in understanding the
immunopathogenesis of PsA have helped to facilitate the development of agents targeting
specific components of the dysregulated inflammatory and immune responses relevant to PsA.
Interestingly , agents with distinct mechanisms of action have shown differential responses across
the various disease domains of PsA , counter to what might have been expected from basic
science investigations. Here, we review data utilizing various novel targeted therapies for PsA ,
focusing on biologic and targeted synthetic therapies. These data might support the idea of a
‘bedside to bench’ concept, whereby results from clinical trials of specific targeted therapies
inform our understanding of the immunopathogenesis of PsA. For example, TNF inhibition
confers substantial and comparable benefit for all domains of PsA , supporting the view that TNF
is a central pro-​inflammatory cytokine across diverse areas of disease involvement. On the other
hand, inhibition of IL-12–IL-23, as compared with inhibition of TNF, has greater efficacy for
psoriasis, comparable efficacy for peripheral arthritis, but was ineffective in studies of axial
spondyloarthritis. Data from studies of agents with distinct mechanisms of action will help to
further refine our understanding of the immunopathogenesis of PsA.

Psoriatic arthritis (PsA) is a chronic inflammatory and maintenance of PsA. For example, genetic studies
autoimmune disease that affects approximately 20% reveal that HLA-​B13, HLA-​B17, HLA-​B57 and HLA-​
of patients with psoriasis1. PsA is generally categorized Cw*0602 occur with increased frequency in patients
as being within the spondyloarthritis (SpA) group of with PsA compared with the general population, with
disease entities2. The clinical manifestations of PsA are HLA-​Cw*0602 being most closely related to psoriasis5,10.
diverse, and can include peripheral arthritis, axial arthri- HLA-​B27 alleles have been associated with symmet-
tis, enthesitis, dactylitis, skin and nail disease, as well as ric sacroiliitis and enthesitis, and HLA-​B08 is associated
other symptoms, including anterior uveitis and intesti- with dactylitis, asymmetric sacroiliitis, joint fusion and
nal inflammation1. Patients with PsA have considera- joint deformity in patients with PsA5,6. Furthermore,
ble disease-​related morbidity, including a substantially IL23R genetic polymorphisms have been identified to
increased risk of atherosclerotic cardiovascular disease, be potential risk factors, not only for PsA but also for
and several associated risk factors for heart disease such other forms of SpA11. Environmental factors that have
as metabolic syndrome, hypertension, and hyperlipidae- been suggested to potentially trigger psoriasis and PsA
mia3. PsA, which is equally prevalent among men and include physical trauma, a number of infections, obesity
women, can impact patients’ psychological and phys- and smoking6,12,13. Gut dysbiosis has also been suggested
ical well-​being, and is associated with quality-​of-life to be relevant to the pathogenesis of PsA, perhaps even
impairment4. having an aetiological role14.
Division of Rheumatology, Although all aspects of the immunopathogenesis of Immune-​mediated mechanisms in PsA result in
Allergy & Immunology, PsA have not yet been fully delineated, key work from the increased production of myriad pro-​inflammatory
University of California San
Diego, San Diego, CA, USA.
numerous groups around the world has helped to shed mediators. As identified by immunohistological and
light on many components that contribute to this sys- other analyses, these mediators might work in synergy
*e-​mail: akavanaugh@
ucsd.edu temic inflammatory autoimmune condition1,5–9. Various to help perpetuate the chronic inflammation charac-
https://doi.org/10.1038/ genetic, environmental and immunological factors teristic of the disease (Fig. 1; Table 1). Key pathological
s41584-019-0285-8 have been suggested to contribute to the development features of PsA are synovial membrane inflammation

Nature Reviews | Rheumatology


Reviews

Key points diseases and highlight the lessons learned about the
immunopathogenesis of these diseases.
• The development and introduction of novel targeted therapies has improved
outcomes for patients with autoimmune systemic inflammatory diseases, including Cytokine targets
psoriatic arthritis (PsA).
TNF inhibitors. TNF is a pro-​inflammatory cytokine
• Whereas some agents, such as TNF inhibitors, have been highly effective across many that is upregulated in both the skin and the synovium in
autoimmune diseases, and across domains of disease, other agents have had
patients with PsA; it is also upregulated in other chronic
disparate impacts on various diseases and domains.
inflammatory diseases, including RA, inflammatory
• With a ‘bedside to bench’ approach to systemic autoimmune diseases, data from bowel disease (IBD; including Crohn’s disease and ulcer-
clinical trials targeting various immune mediators could further our understanding
ative colitis), uveitis, psoriasis and AS5. Macrophages,
of the immunopathogenesis of PsA and other diseases.
T cells, fibroblast-​like synoviocytes, B cells and other
cells produce TNF, with activated macrophages pre-
and increased angiogenesis; these changes, which have sumably being the most relevant source in the inflamed
simi­larities across several types of SpA, including synovium (Fig. 2; Table 1). This cytokine can have an
ankylosing spondylitis (AS) and arthritis associated important role in the activation of circulating and resi-
with Crohn’s disease and ulcerative colitis, can facili- dent immune cells, thereby inducing production of other
tate immune cell migration from the peripheral blood cytokines as well as other mediators, such as chemokines,
into the inflamed joint5. The influx of immune cells, and the release of matrix metalloproteinases (MMPs)5.
such as dendritic cells, macrophages, innate lym- Targeted inhibition of TNF, beginning in the early
phoid cells (ILCs), mucosal-​associated invariant T 1990s and initially for the treatment of RA, proved that
(MAIT) cells, natural killer (NK) cells, mast cells and targeting a single specific molecule could be highly
others, can result in the further production of numerous effective therapy for chronic inflammatory diseases18.
pro-inflammatory mediators (Fig. 1; Table 1). These pro-​ This effect was not necessarily predictable beforehand,
inflammatory mediators can then interact with various as cytokines function in large networks, with individ-
synovial resident cells, including chondrocytes, osteo­ ual cytokines having pleiotropic effector functions
blasts, osteoclasts and fibroblasts5. These events can and several different cytokines having redundancies
help to propagate an autoinflammatory feedback loop, in their mechanisms. Within these large, complex
resulting in the manifestations and sequelae of disease5. cytokine networks, individual mediators are subject
However, although many mediators have been identified to cascades of competing stimulatory and inhibitory
in the PsA synovium and skin and other affected sites, activities. Nevertheless, the notable clinical efficacy of
the precise contribution of each of the individual medi- TNF inhibitors could be said to establish the central
ators remains incompletely understood. Indeed, emerg- role of TNF in the immunopathogenesis of diverse
ing results of trials utilizing specific targeted therapies inflammatory diseases.
suggest that some targets could be of greater relevance Substantial clinical evidence demonstrates that
than others in PsA. TNF inhibitors provide clinical benefit, improve qual-
The development of highly effective biologic DMA­RDs ity of life and reduce structural damage, first shown
in the 1990s helped to spawn further therapeutic in patients with RA and subsequently in patients with
advances in the treatment of inflammatory arthritides, PsA19–23. To date, five TNF inhibitors (adalimumab,
including PsA. These advances have ultimately ushered etanercept, certolizumab, golimumab and infliximab)
in what might be called a ‘bedside to bench’ approach. have been approved by the FDA as well as by regula-
Different from the traditional model whereby basic tory agencies around the world for the treatment of
research generates potential targets for therapy, with several chronic inflammatory diseases (Fig. 3). In PsA,
this new approach, data from human trials of drugs TNF inhibition was shown to be highly effective across
that specifically target varied mediators have provided essentially all of the disparate domains of disease,
insight into the molecular classification of different including peripheral arthritis, enthesitis, dactylitis, skin
chronic inflammatory diseases. For example, inhibi- and nail psoriasis, axial arthritis, IBD and uveitis24–28.
tion of IL-12–IL-23 and IL-17 have proven efficacy in Patients with PsA have an increased prevalence and
PsA (as well as in psoriasis), whereas inhibition of these risk of developing IBD, including Crohn’s disease
same targets in rheumatoid arthritis (RA) has not been and ulcerative colitis29–31. TNF inhibitors have sepa-
effective, even though superficially, RA and PsA have rately been proven effective in the treatment of IBD;
some clinical resemblance and immunohistological adalimumab, golimumab and infliximab were shown
studies could suggest a role for these cytokines in both to be safe and effective for inducing and maintaining
conditions. Furthermore, inhibition of IL-23 has shown clinical remission in patients with moderate-​to-severe
efficacy in psoriasis and PsA, but it has been ineffective ulcerative colitis32–34. Furthermore, a systematic review
for patients with axial SpA15–17. and meta-​analysis found that TNF inhibitors were
Taken together, the data from these and other clin- effective in inducing remission of Crohn’s disease and
ical trials support the notion that inflammatory dis- ulcerative colitis35.
eases such as PsA could perhaps be better defined by TNF inhibition has been effective, not only in treat-
a target-​based disease taxonomy rather than purely ing patients with PsA but also in axial SpA and AS, as
based on immunohistological and other basic studies. revealed in clinical trials36. In addition, juvenile idio-
In this Review, we examine the data from clinical tri- pathic arthritis (JIA) and even some autoinflammat­ory
als of targeted therapies in PsA and other autoimmune conditions, such as familial Mediterranean fever (FMF),

www.nature.com/nrrheum
Reviews

show responsiveness to TNF inhibition37,38. Together, of the inflammatory response in this particular dis-
these data suggest that some mechanisms of inflam- ease. The fact that a potentially pathogenic mediator is
mation are conserved across diverse autoimmune and present, even in abundance, in a given disease does not
rheumatic diseases. guarantee that its inhibition will be clinically successful.
However, although targeted inhibition of TNF is This observation highlights the complex nature of inter-
remarkably successful in the treatment of many chronic actions of myriad mediators of the immune response
systemic inflammatory diseases, this is not the case with that function in complex cascades. Results from clinical
multiple sclerosis (MS) (Fig. 3). This lack of efficacy was data spur further consideration of the functions of these
surprising when first observed, insofar as an argument pleiotropic mediators in specific diseases.
could be made from an immunohistochemical stand- Although targeting the TNF pathway is a mainstay of
point that TNF could be a viable therapeutic target in treating PsA, not all patients treated with TNF inhibitors
MS; to the contrary, clinical studies revealed that patients achieve remission, or the complete absence of disease
with MS treated with anti-​TNF agents had more exacer- activity across all domains. The substantial clinical effi-
bations of MS than patients treated with placebo39. It is cacy seen with TNF inhibitors essentially ‘raised the bar’
proposed that inactivation of TNF in the central nerv- with regard to the goals of therapy for PsA, which helped
ous system affects TNF receptor 2 (TNFR2)-mediated to drive additional research towards understanding the
signalling, which is thought to have a crucial role in contribution of mediators other than TNF to the patho-
remyelination40. Thus, clinical trial data helped to refine genesis of PsA and to the development of alternative
the understanding of the role of a specific component therapies targeting them.

Post-capillary
venule

Co-stimulatory/
adhesion molecules
Endothelial cells

T cell B cell
ECM
PMN molecules

Mast cell

ILC
Plasmacytoid and
myeloid dendritic cells MHC
molecule MAIT cell

NK cell
Resident tissue cells Macrophage

Neuropeptides
• Cytokines
• Chemokines
• Peptide mediators
• Lipid mediators

Synovial and skin Cartilage, bone Systemic


inflammation/ and intestinal manifestations
proliferation damage

Fig. 1 | Immunopathogenesis of Psa. Immune cells, such as T cells, dendritic cells, macrophages, innate lymphoid cells
(ILCs), mucosal-​associated invariant T (MAIT) cells, natural killer (NK) cells, mast cells and others, can produce numerous
pro-​inflammatory mediators. These pro-​inflammatory mediators can then interact with various tissue-​resident cells,
including fibroblast-​like synoviocytes, keratinocytes and intestinal epithelial cells. The propagation of further pro-​
inflammatory mediators results in the manifestations and sequelae of psoriatic arthritis (PsA), including synovial and
skin inflammation and proliferation, and damage to cartilage, bone and intestinal tissue. ECM, extracellular matrix;
PMN, polymorphonuclear neutrophils.

Nature Reviews | Rheumatology


Reviews

Table 1 | Immune mediators potentially relevant to the pathogenesis of Psa and associated conditions
Source mediator(s) Refs
Vasculature
Adhesion molecules on endothelial cells α4 integrin, α4/β7 integrin, LFA-1, LFA-3, ICAM, VCAM 118–121,123,124,

and leukocytes 132,133,151–153

Inflammatory cells
TH1 cells TNF, IFNγ, IL-2, CCL5 (RANTES) 6,154–156

TH2 cells IL-4, IL-5, IL-10, IL-13 155,156

Treg cells IL-10, IL-35, TGFβ 156,157

TH9 cells IL-9, IL-17 , IL-21 158

TH22 cells IL-22, TNF 159

TH17 cells IL-17A , IL-17F, IL-22, TNF 6,155,156

ILCs IL-17A , IL-22 160

NK cells IFNγ, TNF 161

MAIT cells IL-17A , IFNγ 42,162,163

Dendritic cells IL-23, IL-6, IL-12, IL-18, IL-21, IL-23, IL-33, TNF, IFNγ, TGFβ 164–166

Macrophages TNF, IL-18, Il-6, IL-12, IL-15, IL-19, IL-20, IL-23, IL-24, IL-26, IL-32, 167–169

IL-33, IL-38, OSM, GM–CSF, ROS, nitric oxide


Mast cells IL-17A , IL-17F 170

Neutrophils IL-1B, OSM, IL-6, IL-17 , IL-18, TNF, GM–CSF, MMPs, MIF 170

Intracellular
Signalling molecules in diverse cell types JAKs, PDE4 171,172

Synovial cells
Fibroblast-​like synoviocytes IL-1β, IL-6, IL-8, IL-15, IL-18, IL-19, IL-20, IL-26, IL-34, IL-38, MIF, 173,174

MMPs, GM–CSF, growth factors, RANKL


Chondrocytes MMPs, nitric oxide, ADAMTS 175–177

Osteoblasts RANKL 178

Osteoclasts Cathepsin K , MMPs 179–181

ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs; CCL5, CC-​chemokine ligand 5; GM–CSF, granulocyte–
macrophage colony-​stimulating factor ; ICAM, intercellular adhesion molecule; ILC, innate lymphoid cell; JAK , Janus kinase; LFA-1,
lymphocyte function associated antigen 1; MAIT cell, mucosal-​associated invariant T cell; MIF, macrophage migration inhibitory
factor ; MMP, matrix metalloproteinase; NK cell, natural killer cell; OSM, oncostatin M; PDE4, phosphodiesterase 4; PsA , psoriatic
arthritis; RANKL , receptor activator of nuclear factor κΒ ligand; ROS, reactive oxygen species; TGFβ, transforming growth factor β;
VCAM, vascular cell adhesion molecule.

IL-17 inhibitors. Human IL-17 includes several isoforms, in RA and IBD, therapeutic targeting of IL-17 has proven
such as IL-17A, IL-17F, IL-17E and IL-17C. The greatest ineffective or minimally effective in RA44,45. Notably,
amount of investigation to date has focused on the pro-​ although immunopathological studies suggest that IL-17
inflammatory cytokine IL-17A, and to a lesser extent could have a role in IBD, and hence possibly be a suitable
IL-17F; these two cytokines function as IL-17A and therapeutic target, IL-17 inhibition has been ineffective
IL-17F homodimers and as an IL-17A–IL-17F hetero­ or even caused worsening of disease in studies of IBD44,46.
dimer. Although many studies specify the isoforms, The reasons for this potentially paradoxical effect are
studies that mention only ‘IL-17’ often refer to IL-17A. not clear, but might relate to changes to the local micro­
IL-17A has been shown to be increased in synovial biome (especially the fungal constituents), alterations in
fluid of patients with PsA and in patients with other the composition of intestinal mucous, interference with
arthritides; it is also overexpressed in skin affected by gut epithelial integrity, or a combination of these factors,
psoriasis41. IL-17 can have various pathogenic roles, all related to IL-17 inhibition47.
including stimulation of osteoclastogenesis, chondro- Clinical studies reveal that treatment with the mono­
cyte activation, fibrinogenesis and angiogenesis, as well clonal antibodies specifically targeting IL-17A that
as stimulation of pro-​inflammatory cytokine and MMP are currently approved for PsA, secukinumab and
production42. T helper 17 (TH17) cells, γδ T cells, CD8+ ixekizumab, improved peripheral arthritis, inhibited
T cells and ILC-3 are probably the primary sources of the progression of peripheral joint radiographic dam-
IL-17 in PsA and other inflammatory diseases, although age, reduced the frequency of enthesitis and dactylitis,
other cells can produce IL-17A5,43 (Figs 1,2; Table 1). improved psoriasis disease activity, and improved the
Targeting the IL-17 cytokine axis has therapeutic ben- physical functioning and quality of life of patients with
efit in patients with PsA as well as with AS. However, PsA48–50. Interestingly, whereas the efficacy of IL-17
despite overexpression of IL-17 at sites of inflammation inhibition appears to be comparable to that of TNF

www.nature.com/nrrheum
Reviews

Cell signalling • Anakinra


• Canakinumab
• Rilonacept
Apremilast • Tofacitinib
• Baricitinib
• Filgotinib
• Upadacitinib IL-1
PDE4 • Tocilizumab
IL-6 • Clazakizumab
JAK Macrophage/ TNF
CD80/ monocyte
CD86
CD28
• Adalimumab
Cytokine
• Certolizumab
• Etanercept
• Infliximab
Rituximab APC T cell • Golimumab
CD20
Abatacept Ustekinumab
• Guselkumab
• Tildrakizumab
IL-12 IL-23 • Risankizumab
B cell • Mirikizumab

Leukocyte

TH1 cell TH17 cell


Natalizumab α4 α4β7
(anti-α4-chain)
IL-17F IL-17A
Vedolizumab
(anti-α4β7- • Secukinumab
complex) • Ixekizumab
• Bimekizumab • Bimekizumab
• Brodalumab • Brodalumab
Endothelium VCAM ICAM MadCAM-1

Fig. 2 | Summary of immune target interactions in Psa. Therapeutic targets relevant to psoriatic arthritis (PsA) include
various cytokines (for example, TNF, IL-12, IL-23 and IL-17), non-​cytokine pro-​inflammatory mediators (such as Janus
kinases (JAKs), phosphodiesterase 4 (PDE4) and adhesion molecules) and various immune cells. APC, antigen-​presenting
cell; ICAM, intercellular adhesion molecule; MadCAM-1, mucosal vascular addressin cell adhesion molecule 1;
PsA , psoriatic arthritis; VCAM, vascular cell adhesion molecule.

inhibition for peripheral arthritis in PsA, its efficacy in IL-12 is produced by monocytes and macrophages and
regard to psoriasis is generally greater than that of TNF51. stimulates TH1 cells, whereas IL-23 is produced mainly
As far as other domains of disease are concerned, IL-17 by myeloid dendritic cells and can regulate TH17 cells
inhibition has also proven effective in the treatment (Fig. 2; Table 1). In animal models of enthesitis, IL-23
of AS, and would presumably therefore be effective in has also been shown to stimulate the release of IL-22;
patients with PsA who have axial arthritis52. this release of IL-22 has been associated with keratino-
In addition to the two aforementioned anti-​IL-17A cyte proliferation and new bone formation58,59. IL-23
monoclonal antibodies, there are other agents that target can also upregulate the production of IL-17, which, as
IL-17 (Fig. 2). Brodalumab, a monoclonal antibody spe- noted earlier, has numerous roles in the pathogenesis
cific for IL-17 receptor A, is approved by the FDA for the of joint damage.
treatment of severe plaque psoriasis53. Bimekizumab, a Interestingly, prior to the greater understanding of
monoclonal antibody that neutralizes both IL-17A and psoriasis that has come to attention, particularly over the
IL-17F, has shown therapeutic potential in treating AS, past decade, such as increased appreciation of the role
psoriasis and PsA54–56. of IL-17 and IL-23, IL-12 was considered to be a viable
Whether differences in the fine specificity of inhibi- therapeutic target in psoriasis, a putative ‘TH1’ disease60.
tors of the various IL-17 isoforms will result in important Early success in targeting IL-12 came with the use of
differences in efficacy across distinct domains of disease, ustekinumab, a monoclonal antibody that targeted the
or in safety, remains to be delineated. p40 subunit common to IL-12 and IL-2361. Presently,
however, the notable efficacy of IL-12–IL-23 inhibi-
IL-12–IL-23 and IL-23 inhibitors. IL-12 and IL-23, tion in psoriasis – which is considerably greater than
cytokines within the IL-12 family (along with IL-27 that seen with TNF inhibition – is thought to probably
and IL-35), might contribute to pathogenesis in PsA57. derive mostly, if not entirely, from its effect on IL-23

Nature Reviews | Rheumatology


Reviews

Cytokine targets Non-cytokine targets

Chronic inflammatory IL-12/ CD80/


disease TNF IL-6R IL-1 IL-23 IL-17A IL-23 Integrin JAKs CD86 PDE4 CD20
Rheumatoid arthritis – – – – –
Autoinflammatory
disease/sJIA

Crohn’s disease – + +
Anti-α4, α4/β7

Ulcerative colitis + – + +
Anti-α4/β7

Psoriasis +
Anti-LFA1 (CD11a)
Psoriatic arthritis + + + –
Anti-LFA3
Ankylosing spondylitis/
axSpA – – – – + – –

Multiple sclerosis – +
Anti-α4

FDA-approved – Disease-aggravating effect


+ Preliminary data on clinical efficacy – Failed to meet primary endpoints
Insufficient data/not studied

Fig. 3 | Summary of cytokine and non-​cytokine targets in various chronic inflammatory diseases. This figure
illustrates the clinical efficacy of the inhibition of various cytokines and non-​cytokine targets with targeted therapies
that are FDA-​approved (dark green), therapies with preliminary data on clinical efficacy (light green), clinical data
resulting in disease-​aggravating effects (red), therapies for which clinical data failed to meet primary endpoints
(dark grey), or therapies with insufficient or no clinical data (white). axSpA, axial spondyloarthritis; IL-6R , IL-6 receptor;
JAK , Janus kinase; PDE4, phosphodiesterase 4; sJIA , systemic juvenile idiopathic arthritis.

rather than IL-1260. In PsA, inhibition of IL-12 and IL-23 inflamed sites in RA and AS, and could therefore be
has been effective for several disease domains, including potential targets of therapy, targeting the IL-12–IL-23
peripheral arthritis, enthesitis and psoriasis62. In addi- pathway has not been effective in treating either of
tion, inhibition of IL-12 and IL-23 has been effective these diseases17,76–78. In well-​conducted clinical trials,
in treating patients with IBD63,64. As well as improv- neither ustekinumab nor guselkumab was effective
ing the signs and symptoms of peripheral arthritis in in treating patients with RA78. Perhaps more surpris-
PsA, ustekinumab reduced radiographic progression ingly, given not only immunohistological data but also
of peripheral joint disease61,65. IL-12–IL-23 inhibition genetic studies showing an association between allelic
has also been suggested to be possibly superior to TNF polymorphisms in IL-23 and SpA, the IL-23 inhibitor
inhibition in achieving clearance of enthesitis in patients risankizumab failed to show efficacy in a trial of active
with PsA66. AS17. Also, studies assessing ustekinumab in AS as well
As with psoriasis, it has been suggested that it is as in non-​radiographic axial SpA were stopped owing
really inhibition of IL-23, rather than that of IL-12, that to inefficacy79.
is relevant to the efficacy of IL-12–IL-23 inhibition in The reasons underlying these surprising data are not
PsA. Currently, a number of IL-23p19-specific inhibi­ certain. It is possible that IL-23 alone is not a primary
tors are approved for the treatment of psoriasis and driver of AS immunopathogenesis; for example, the
others are in the latter phases of development, including contribution of IL-23 might be primarily through IL-17
guselkumab, risankizumab, mirikizumab, tildrakizumab upregulation, and it is possible that other sources of
and brazikumab67–70. Notably, when assessed in psoria- IL-17 are more relevant or compensatory in AS, obviat-
sis, these agents have been highly effective, with clinical ing the potential benefit of IL-23 inhibition. Of note, this
improvement greater than that seen with TNF inhibi- rationale would not apply in RA, as both IL-12–IL-23
tors68. These agents have also shown therapeutic promise and IL-17 inhibition were ineffective in this disease44,78
in treating peripheral arthritis in patients with PsA and These studies suggest that despite some immuno­
their development for PsA is proceeding16,71,72. As was histochemical similarities, there are differences in the
noted with IL-12–IL-23 inhibition, IL-23 inhibition has immunopathogenesis of RA, PsA and axial SpA. The
also been shown to be effective in IBD73–75 IL-12–IL-23 pathway might be a driving force for
Although immunohistological studies have sug- inducing inflammation in psoriasis as well as in many
gested that IL-12 and IL-23 can be found at actively domains of PsA, exclusive of axial disease.

www.nature.com/nrrheum
Reviews

IL-6 inhibitors. IL-6 is a pro-​inflammatory cytokine (an IL-1 receptor–Fc- fusion protein) have impressive
produced by T cells, macrophages and other cells such efficacy in patients with various autoinflammatory
as synovial fibroblasts, that can be associated with path- disorders and for sJIA95–98 (Fig. 3).
ogenic TH17 cell differentiation and IL-17 production5,80
(Fig. 2; Table 1). Studies have demonstrated elevated lev- Non-​cytokine targets
els of IL-6 in patients with PsA81. Activated TH17 cells Janus kinase inhibitors. Agents targeting intracellular
produce IL-6, which can then promote IL-12 and IL-23 tyrosine kinases that regulate inflammatory responses
expression, thereby stimulating TH1 and TH17 cell differ- via cytokine signalling pathways have added to the ther-
entiation and the promotion of inflammation82 (Table 1). apeutic armamentarium in rheumatic diseases (Figs 2,3).
Although these observations would suggest a possible There are four Janus kinase enzymes (JAKs): JAK1,
rationale for targeting IL-6 in PsA, in a clinical trial, JAK2, JAK3 and Tyk2. JAKs interact with signal trans-
treatment with the IL-6 inhibitor clazakizumab resulted ducers and activators of transcription (STAT) family
in quite small improvements in peripheral arthritis members to modulate gene transcription downstream
and minimal, if any, effect on psoriasis in patients with of various cytokines that lack intrinsic signalling capa-
active PsA83. Owing to this limited efficacy, the devel- bilities through their specific receptors99,100. JAKs have
opment of this agent for the treatment of PsA has been been targeted with small molecule JAK inhibitors (also
halted. Interestingly, despite immunopathophysiologi- known as jakinibs), which have been shown to have dif-
cal evidence that suggests that IL-6 might have a role ferential selectivity for the four JAK isoforms in vitro100.
in SpA, anti-​IL 6 therapies were also shown to be inef- Tofacitinib, an orally administered inhibitor of JAK3 and
fective in the treatment of AS84,85. These data stand in JAK1 and to a lesser extent JAK2, has been approved
distinction to the substantial efficacy of IL-6 inhibition for the treatment of RA, PsA, psoriasis and ulcerative
in patients with RA and various other inflammatory colitis101–106 (Fig. 3).
arthritides. Tocilizumab, a humanized anti-​human Several randomized trials have demonstrated that
IL-6 receptor antibody, is effective in the treatment of tofacitinib has efficacy in treating active PsA101,107. The
RA and systemic JIA (sJIA)86,87. IL-6 inhibition has also mechanisms underlying the efficacy of tofacitinib in PsA
been effective in some autoinflammatory conditions88. are not fully defined. Although the effects of tofacitinib
These data highlight that the identification of a puta- could be attributable in part to the blockade of JAK3,
tive target in inflamed tissues does not equate to clinical resulting in inhibition of IL-2, IL-4 and IL-15, a more
efficacy inhibiting that target in patients with the disease likely explanation might be that blockade of JAK1 and
under study. JAK2 inhibits signalling by IL-6, IFNγ, IL-12 and IL-23,
which, as noted, are key mediators in the pathogene-
IL-1 inhibitors. IL-1 levels are highly upregulated in the sis of PsA101,107. However, in discussing the mechanisms
synovium and skin of patients with PsA and psoriasis, of action of JAK inhibitors, it is relevant to note that
respectively, and studies have shown that the IL-1 gene ‘selectivity’ is relative, and the in vivo situation can dif-
family might have a role in the development of PsA89. An fer from in vitro observations108 Also, the JAKs mostly
approach to IL-1 blockade involves the use of its endo­ function as heterodimers and even heterotrimers (the
genous antagonist, IL-1 receptor antagonist (IL-1Ra), a exception being the JAK2 homodimer, which is down-
glycoprotein that competitively inhibits binding of IL-1α stream of haematopoietic growth factor receptors)100.
and IL-1β to the cell surface IL-1 receptor. Recombinant Therefore, speculation about the ultimate mechanism
IL-1ra, anakinra, has received regulatory approval from by which these agents are effective might be tenuous.
the FDA and other agencies for the treatment of RA90. In controlled trials, tofacitinib has had some efficacy
Despite this approval, anakinra is rarely used to treat in psoriasis, but the extent of response was less than that
RA owing to its substantially lower efficacy compared seen with agents with different mechanisms of action,
with TNF inhibitors91. This difference in efficacy was such as TNF inhibitors and especially inhibitors of
not necessarily expected beforehand, as not only is IL-1 IL-17 and IL-23102. Interestingly, in RA, JAK inhibitors
in as great abundance as TNF in the inflamed RA syn- appear to be as effective as TNF inhibitors109. Some of
ovium but also the myriad in vitro and in vivo activ- the observed clinical effect in certain domains of PsA
ities of these two pro-​inflammatory cytokines would compared with other domains or with other conditions,
appear to largely overlap92,93. The far greater clinical such as RA, might relate to dose. In clinical trials in PsA,
efficacy of TNF inhibition shows that data from clini­ lower doses of tofacitinib seemed as effective at con-
cal studies — in a ‘bedside to bench’ approach — can trolling peripheral arthritis as higher doses, similar to
inform the total understanding of complex autoimmune what was seen in RA, whereas in psoriasis, higher doses
inflammatory diseases. were more effective, although the extent of response was
A single-​centre, open-​label, pilot study revealed still less than that seen with other agents101,107,110. The
that anakinra has little benefit in PsA; thus, despite mechanistic basis of this dose differential is unknown.
data from immunopathological studies that show its JAK inhibition has also shown clinical efficacy in
presence in sites of inflammation in PsA, IL-1 does treating IBD. In patients with moderately to severely
not seem to be a central driver of PsA inflammation94. active ulcerative colitis, tofacitinib was more effective
In contrast to its minimal effect in PsA, IL-1 inhibi- in induction and maintenance therapy compared with
tion has shown benefit in autoinflammatory diseases; placebo, at higher doses than those approved for use
studies show that anakinra, canakinumab (a human in RA and PsA103. In one Crohn’s disease study, treat-
monoclonal antibody specific for IL-1β) and rilonacept ment with tofacitinib did not meet its primary efficacy

Nature Reviews | Rheumatology


Reviews

end point, but tofacitinib and other JAK inhibitors are is effective in moderately to severely active Crohn’s dis-
actively being studied in Crohn’s disease as well as ulcer- ease119. Natalizumab has also been shown to be an effec-
ative colitis111. JAK inhibition might also be effective for tive drug for the treatment of relapsing–remitting MS
AS: one phase II study revealed that tofacitinib demon- and reduced the risk of relapse during 2 years of treat-
strated greater clinical efficacy than placebo in reducing ment118. On the contrary, natalizumab was ineffective in
the signs and symptoms of AS112. In addition, filgotinib, the treatment of RA126, in another example of different­
a selective JAK1 inhibitor, has been shown effective with ial efficacy of targeted therapies across distinct diseases.
respect to clinical as well as imaging (MRI) outcomes in Vedolizumab, a humanized monoclonal antibody that
the treatment of AS and PsA113,114. Several JAK inhibi- binds α4β7 integrin expressed on leukocytes, blocks the
tors, including the JAK1 and JAK2 inhibitor baricitinib, interaction of α4β7 integrin with MadCAM-1 on intes-
which has been approved for use in RA, and the JAK1 tinal endothelial cells (Fig. 2); thus, vedolizumab selec-
inhibitors filgotinib and upadacitinib, are being studied tively blocks lymphocyte trafficking in intestinal tissues.
in PsA114–116. Hypothetically, JAK inhibitors with other Vedolizumab is approved by the FDA for the treatment
specificities, for example, those targeting Tyk2, might of ulcerative colitis and Crohn’s disease; however, it has
be advantageous in PsA and psoriasis; this concept is been suggested that use of vedolizumab might be associ-
presently under study117. ated with flares of arthritis in patients with IBD120,127–131.
Efalizumab, a monoclonal antibody against CD11a, one
Selective adhesion molecule inhibitors. Selective adhe- of the subunits of LFA-1, interferes with T-​cell activation
sion molecule inhibitors might help to prevent the migra- in the lymph nodes and migration of T cells to the skin,
tion of various leukocytes to distinct inflammatory sites. ultimately providing clinical benefit in patients with
Approaches to inhibiting adhesion molecules include moderate-​to-severe psoriasis123. Although efalizumab
agents targeting α4-integrin, α4/β7-integrin, lympho- was effective in the treatment of psoriasis, it was not
cyte function associated antigen 1 (LFA-1) and LFA-3. effective in treating PsA122. Alefacept, a fusion protein
Therapies directed at adhesion molecules have been of soluble LFA-3 with Fc fragments of IgG1, has shown
studied in a number of autoimmune diseases and have some clinical efficacy in psoriasis and PsA124,132,133.
shown clinical benefit in IBD, MS, psoriasis and PsA ICAM-1 and VCAM-1 are implicated in the propaga-
but only minimal effectiveness in RA118–125 (Fig. 3) . tion of inflammation and are upregulated in inflamma-
Natalizumab, a humanized IgG4 anti-​α4-integrin mono­ tory diseases, including Crohn’s disease and RA, as well
clonal antibody, inhibits the interaction between α4β7 as PsA134–136. Although adhesion molecule targeted thera-
integrin and mucosal addressin cell adhesion molecule-1 pies have been effective in IBD and in MS, results in RA,
(MadCAM-1) and between α4β1 integrin and vascular-​ for example, with a monoclonal antibody to ICAM-1,
cell adhesion molecule 1 (VCAM-1) (Fig. 2). Natalizumab have been much less successful125.
In summary, despite a putative rationale for their
Psoriasis targeting, and despite efficacy in diverse diseases such as
IBD and MS, specific adhesion molecule inhibitors have
not been proven effective in patients with peripheral
Exposome arthritis, including PsA (Fig. 3).

Genome Metabolome Proteome Microbiome Epigenome Phosphodiesterase 4 inhibitors. Phosphodiesterase-4


(PDE4) is an enzyme that stimulates cyclic AMP degra-
dation (Fig. 2), which promotes inflammatory responses
• PsA signs and symptoms by altering levels of various cytokines. Apremilast, an
• Activity in various domains
oral PDE4 inhibitor approved for use in PsA, has been
shown to be an effective and well-​tolerated therapy for
PsA (Fig. 3). Apremilast is effective in improving var-
Peripheral Axial Enthesitis Dactylitis Skin and IBD Uveitis ious signs and symptoms of patients with active PsA,
arthritis arthritis nails including benefits for psoriasis, enthesitis, dactylitis and
peripheral arthritis137,138. Although effective in PsA
Data from clinical trials of specific targeted therapies: and psoriasis, PDE4 inhibition did not show meaningful
Clinical impact on individual domains
efficacy in either AS or in RA139,140. Ongoing studies are
evaluating the efficacy and safety of apremilast in IBD141.
Integration of data to help define endotype
T-​cell activation inhibitors. It is well known that T cell
Fig. 4 | Bedside to bench: towards precision medicine in Psa. Individuals with activation plays a crucial role in the inflammatory
psoriasis have a unique measure of lifetime exposure that relates to their health, thus response, which provides the rationale for using T cell
defining their ‘exposome’, which can further influence their genome, metabolome, targeted therapies for the treatment of diseases such as
proteome, microbiome and epigenome. These domains can, in turn, affect the signs
RA and PsA. Abatacept is a fusion protein (CTLA4-Ig)
and symptoms of psoriatic arthritis (PsA), including various clinical domains. Data from
clinical trials of specific targeted therapies have had a substantial effect on treatment that modulates a co-​stimulatory signal necessary for
of these clinical domains for individuals with PsA. Integrating data from clinical trials T cell activation, by binding to the CD80–CD86 ligands
will help to characterize PsA endotypes, defined by the molecular mechanisms on the surface of antigen-​presenting cells142 (Fig. 2). As
underlying the clinical phenotype and/or disease features, which will in turn further evident from a Cochrane systematic review, abatacept
our understanding of the various omic domains. IBD, inflammatory bowel disease. is efficacious and safe in the treatment of RA (Fig. 3)143.

www.nature.com/nrrheum
Reviews

A phase III trial revealed that patients with PsA treated Box 1 | Future directions
with abatacept had improvements in peripheral arthri-
tis irrespective of prior exposure to TNF inhibitors, • Future clinical trials comparing agents with different
although effects on skin lesions were minimal, despite mechanisms of action (that is, TNF, IL-12–IL-23 and/or
strong suggestion of a potential immunopathogenic role IL-17 inhibition) in head-​to-head studies, supplemented
with translational research, could provide us with
for T cell activation in psoriasis144.
information about which cytokine pathway
predominates in the pathophysiology of psoriatic
CD20 inhibitors. B cell depletion therapy by targeting arthritis (PsA).
CD20, a B lymphocyte specific molecule, has proven • Future clinical trials evaluating multidomain measures,
beneficial in various autoimmune disorders. Treatment such as composite disease activity indices and minimal
with rituximab, an anti-​CD20 monoclonal antibody, has disease activity criteria, might inform us about which
been efficacious in treating patients with either RA or targeted therapy enables better disease control for PsA.
MS145–147 (Figs 2,3). In patients with AS, rituximab did not • The microbiome of the gut or skin has been suggested
seem effective in treating patients who had previously to be relevant to the pathogenesis of PsA and could
had an inadequate response to TNF inhibition; how- provide potential therapeutic targets.
ever, it was efficacious in treating TNF-​inhibitor-naive
patients148. An exploratory study revealed that rituximab
had minimal efficacy in PsA149. and other target-​based taxonomy of varied chronic
inflammatory diseases. The ‘bedside-​to-bench’ approach
From bedside to bench in PsA might also afford advances in precision medicine of PsA
As a result of years of intense basic and translational in the future (Fig. 4). Individuals with psoriasis have life-
research by many investigators worldwide, our under- time environmental exposures that affect the specific
standing of the immunopathogenesis of PsA and other nature of their immune response, thus defining their
chronic systemic inflammatory diseases has progressed ‘exposome’. Such exposures can influence genomic,
tremendously. One result of such study has been the metabolomic, proteomic, microbiomic, and epigenomic
identification of numerous components of the immune parameters. These varied ‘omic’ parameters can affect
and inflammatory responses that appear to be over­ the signs and symptoms seen in patients with PsA,
expressed or activated in these diseases. These dysregu­ including various clinical domains such as peripheral
lated constituents could then be considered potential arthritis, axial arthritis, dactylitis, skin and nail disease,
therapeutic targets. Along with progress in biotechnol- IBD and uveitis. Data from clinical trials of specific
ogy, these discoveries led to the development of pharma- targeted therapies have made a substantial impact on
ceutical agents specifically targeting many soluble and the treatment of these clinical domains for individuals
cell surface pro-​inflammatory mediators. with PsA. Data from clinical trials of specific targeted
Clinical results with these agents have been sur- therapies could help to further delineate the underlying
prising in some cases. Despite presumptive immuno- mechanisms of disease (Box 1). Further future trials that
histopathological and other evidence suggesting an directly compare agents with different mechanisms of
aetiopathogenic role for various mediators in specific action should provide additional information about the
distinct diseases, therapies targeting these mediators relative roles of different pathways, specifically as related
have been ineffective in some studies. For example, IL-6 to particular disease domains (Box 1). Integrating the
and IL-1 inhibition have not been nearly as effective in data from various clinical trials will aid in characterizing
PsA as has TNF inhibition83,150. PsA endotypes, defined by the molecular mechanisms
Data across the various domains of disease in PsA underlying the disease’s clinical phenotype and/or fea-
have also generated some unexpected results. For exam- tures, which will in turn further our understanding of
ple, IL-17 inhibition is effective in treating peripheral various omic parameters.
arthritis, enthesitis, dactylitis and axial arthritis in PsA
and is more effective than TNF inhibition for treating Conclusions
psoriasis; however, IL-17 inhibition was ineffective for, A number of pro-​inflammatory mediators have been
or worsened, IBD. IL-23 inhibition and dual IL-12– implicated in the immunopathogenesis of PsA on the
IL-23 inhibition also work across various PsA domains, basis of immunohistological studies and other lines of
including peripheral arthritis, enthesitis, dactylitis and evidence. However, some therapies that were ‘supposed
IBD, and are also highly effective for psoriasis; however, to work’ on the basis of these findings have failed in
they are ineffective for axial arthritis15,17,61,64,66–68. clinical trials. Still others affected some domains of dis-
Many other examples, as reviewed earlier, reveal clin- ease more or less than others. These findings support a
ical data that were unexpected given potentially promis- focus on the ‘bedside-​to-bench’ rather than the oppo-
ing data from prior immunological studies. One might site approach, as they reinforce the notion that valuable
liken the situation to trying to ascertain the cause of an information can be gleaned from analysing the clinical
ongoing fire: at initial glance, it might be difficult to dif- efficacy of targeted therapies in PsA and other auto­
ferentiate those who started the fire from those who are immune inflammatory diseases. With the development of
trying to put it out or those who are just bystanders. additional novel therapies, more will no doubt be learned
Thus, a ‘bedside-​to-bench’ approach with data from about these complex systemic inflammatory conditions.
clinical trials that targeted various immune mediators
can help to inform our understanding of the cytokine Published online xx xx xxxx

Nature Reviews | Rheumatology


Reviews

1. Ritchlin, C. T., Colbert, R. A. & Gladman, D. D. Psoriatic 25. Mease, P. J. Adalimumab: an anti-​TNF agent for the 47. Hall, A. O., Towne, J. E. & Plevy, S. E. Get the IL-17F
arthritis. N. Engl. J. Med. 376, 2095–2096 (2017). treatment of psoriatic arthritis. Expert Opin. Biol. Ther. outta here! Nat. Immunol. 19, 648–650 (2018).
2. Gladman, D. D., Antoni, C., Mease, P., Clegg, D. O. 5, 1491–1504 (2005). 48. Mease, P. J. et al. Secukinumab inhibition of
& Nash, P. Psoriatic arthritis: epidemiology, clinical 26. Kavanaugh, A. et al. Golimumab, a new human tumor interleukin-17A in patients with psoriatic arthritis.
features, course, and outcome. Ann. Rheum. Dis. 64, necrosis factor α antibody, administered every four N. Engl. J. Med. 373, 1329–1339 (2015).
ii14–ii17 (2005). Suppl 2. weeks as a subcutaneous injection in psoriatic arthritis: 49. McInnes, I. B. et al. Secukinumab, a human anti-​
3. Horreau, C. et al. Cardiovascular morbidity and twenty-​four-week efficacy and safety results of a interleukin-17A monoclonal antibody, in patients with
mortality in psoriasis and psoriatic arthritis: randomized, placebo-​controlled study. Arthritis Rheum. psoriatic arthritis (FUTURE 2): a randomised, double-​
a systematic literature review. J. Eur. Acad. Dermatol. 60, 976–986 (2009). blind, placebo-​controlled, phase 3 trial. Lancet 386,
Venereol. 27, Suppl 3,12–29 (2013). 27. Mease, P. J. et al. Effect of certolizumab pegol on signs 1137–1146 (2015).
4. Husni, M. E., Merola, J. F. & Davin, S. The psychosocial and symptoms in patients with psoriatic arthritis: 50. Mease, P. J. et al. Ixekizumab, an interleukin-17A
burden of psoriatic arthritis. Semin. Arthritis Rheum. 24-week results of a Phase 3 double-​blind randomised specific monoclonal antibody, for the treatment of
47, 351–360 (2017). placebo-​controlled study (RAPID-​PsA). Ann. Rheum. Dis. biologic-​naive patients with active psoriatic arthritis:
5. Veale, D. J. & Fearon, U. The pathogenesis of psoriatic 73, 48–55 (2014). results from the 24-week randomised, double-​blind,
arthritis. Lancet 391, 2273–2284 (2018). 28. Mease, P. J. et al. Etanercept treatment of psoriatic placebo-​controlled and active (adalimumab)-controlled
6. Barnas, J. L. & Ritchlin, C. T. Etiology and pathogenesis arthritis: safety, efficacy, and effect on disease period of the phase III trial SPIRIT-​P1. Ann. Rheum. Dis.
of psoriatic arthritis. Rheum. Dis. Clin. North Am. 41, progression. Arthritis Rheum. 50, 2264–2272 (2004). 76, 79–87 (2017).
643–663 (2015). 29. Li, W. Q., Han, J. L., Chan, A. T. & Qureshi, A. A. 51. Nash, P. et al. Ixekizumab for the treatment of
7. Cafaro, G. & McInnes, I. B. Psoriatic arthritis: tissue-​ Psoriasis, psoriatic arthritis and increased risk of patients with active psoriatic arthritis and an
directed inflammation? Clin. Rheumatol. 37, 859–868 incident Crohn’s disease in US women. Ann. Rheum. Dis. inadequate response to tumour necrosis factor
(2018). 72, 1200–1205 (2013). inhibitors: results from the 24-week randomised,
8. Boutet, M. A., Nerviani, A., Gallo Afflitto, G. & 30. Makredes, M., Robinson, D. Jr., Bala, M. double-​blind, placebo-​controlled period of the
Pitzalis, C. Role of the IL-23/IL-17 axis in psoriasis & Kimball, A. B. The burden of autoimmune disease: SPIRIT-​P2 phase 3 trial. Lancet 389, 2317–2327
and psoriatic arthritis: the clinical importance of its a comparison of prevalence ratios in patients with (2017).
divergence in skin and joints. Int. J. Mol. Sci. 19, E530 psoriatic arthritis and psoriasis. J. Am. Acad. Dermatol. 52. Baeten, D. et al. Anti-​interleukin-17A monoclonal
(2018). 61, 405–410 (2009). antibody secukinumab in treatment of ankylosing
9. Millar, N. L., Murrell, G. A. & McInnes, I. B. 31. Egeberg, A., Thyssen, J. P., Burisch, J. & Colombel, J. F. spondylitis: a randomised, double-​blind, placebo-
Inflammatory mechanisms in tendinopathy — towards Incidence and risk of inflammatory bowel disease in controlled trial. Lancet 382, 1705–1713 (2013).
translation. Nat. Rev. Rheumatol. 13, 110–122 (2017). patients with psoriasis-​a nationwide 20-year cohort 53. Mease, P. J. et al. Brodalumab, an anti-​IL17RA
10. Prinz, J. C. Human leukocyte antigen-​class I alleles study. J. Invest. Dermatol. 139, 316–323 (2019). monoclonal antibody, in psoriatic arthritis. N. Engl.
and the autoreactive T cell response in psoriasis 32. Rutgeerts, P. et al. Infliximab for induction and J. Med. 370, 2295–2306 (2014).
pathogenesis. Front. Immunol. 9, 954 (2018). maintenance therapy for ulcerative colitis. N. Engl. 54. Glatt, S. et al. Dual IL-17A and IL-17F neutralisation
11. Yago, T. et al. IL-23 and Th17 disease in inflammatory J. Med. 353, 2462–2476 (2005). by bimekizumab in psoriatic arthritis: evidence from
arthritis. J. Clin. Med. 6, pii: E81 https://doi.org/ 33. Sandborn, W. J. et al. Adalimumab induces and preclinical experiments and a randomised placebo-​
10.3390/jcm6090081 (2017). maintains clinical remission in patients with moderate-​ controlled clinical trial that IL-17F contributes to
12. Thorarensen, S. M. et al. Physical trauma recorded in to-severe ulcerative colitis. Gastroenterology 142, human chronic tissue inflammation. Ann. Rheum. Dis.
primary care is associated with the onset of psoriatic 257–265 e251-253 (2012). 77, 523–532 (2018).
arthritis among patients with psoriasis. Ann. Rheum. 34. Sandborn, W. J. et al. Subcutaneous golimumab 55. Glatt, S. et al. First-​in-human randomized study of
Dis. 76, 521–525 (2017). maintains clinical response in patients with moderate-​ bimekizumab, a humanized monoclonal antibody and
13. Duffin, K. C. et al. Association between IL13 to-severe ulcerative colitis. Gastroenterology 146, selective dual inhibitor of IL-17A and IL-17F, in mild
polymorphisms and psoriatic arthritis is modified 96–109 e101 (2014). psoriasis. Br. J. Clin. Pharmacol. 83, 991–1001
by smoking. J. Invest. Dermatol. 129, 2777–2783 35. Ford, A. C. et al. Efficacy of biological therapies in (2017).
(2009). inflammatory bowel disease: systematic review and 56. US National Library of Medicine. ClinicalTrials.gov
14. Scher, J. U., Littman, D. R. & Abramson, S. B. meta-​analysis. Am. J. Gastroenterol. 106, 644–659 https://clinicaltrials.gov/ct2/show/NCT02963506
Microbiome in inflammatory arthritis and human quiz 660 (2011). (2018).
rheumatic diseases. Arthritis Rheumatol. 68, 35–45 36. Callhoff, J., Sieper, J., Weiss, A., Zink, A. & Listing, J. 57. Vignali, D. A. & Kuchroo, V. K. IL-12 family cytokines:
(2016). Efficacy of TNFα blockers in patients with ankylosing immunological playmakers. Nat. Immunol. 13,
15. Gordon, K. B. et al. Efficacy of guselkumab in spondylitis and non-​radiographic axial spondyloarthritis: 722–728 (2012).
subpopulations of patients with moderate-​to-severe a meta-​analysis. Ann. Rheum. Dis. 74, 1241–1248 58. Sherlock, J. P. et al. IL-23 induces spondyloarthropathy
plaque psoriasis: a pooled analysis of the phase III (2015). by acting on ROR-​γt+ CD3+CD4-CD8- entheseal
VOYAGE 1 and VOYAGE 2 studies. Br. J. Dermatol. 37. Seyahi, E., Ozdogan, H., Celik, S., Ugurlu, S. & Yazici, H. resident T cells. Nat. Med. 18, 1069–1076 (2012).
178, 132–139 (2018). Treatment options in colchicine resistant familial 59. Fujita, H. Role of IL-22 in the pathogenesis of skin
16. Deodhar, A. et al. Efficacy and safety of guselkumab Mediterranean fever patients: thalidomide and diseases. Nihon Rinsho Meneki Gakkai Kaishi 35,
in patients with active psoriatic arthritis: a randomised, etanercept as adjunctive agents. Clin. Exp. Rheumatol. 168–175 (2012).
double-​blind, placebo-​controlled, phase 2 study. Lancet 24, S99–S103 (2006). 60. Hawkes, J. E., Chan, T. C. & Krueger, J. G. Psoriasis
391, 2213–2224 (2018). 38. Kearsley-​Fleet, L. et al. Effectiveness and safety of TNF pathogenesis and the development of novel targeted
17. Baeten, D. et al. Risankizumab, an IL-23 inhibitor, inhibitors in adults with juvenile idiopathic arthritis. immune therapies. J. Allergy Clin. Immunol. 140,
for ankylosing spondylitis: results of a randomised, RMD Open 2, e000273 (2016). 645–653 (2017).
double-​blind, placebo-​controlled, proof-​of-concept, 39. TNF neutralization in MS: results of a randomized, 61. Kavanaugh, A. et al. Ustekinumab, an anti-​IL-12/23
dose-​finding phase 2 study. Ann. Rheum. Dis. 77, placebo-​controlled multicenter study. The Lenercept p40 monoclonal antibody, inhibits radiographic
1295–1302 (2018). Multiple Sclerosis Study Group and the University of progression in patients with active psoriatic arthritis:
18. Chatzantoni, K. & Mouzaki, A. Anti-​TNF-α antibody British Columbia MS/MRI analysis group. Neurology results of an integrated analysis of radiographic data
therapies in autoimmune diseases. Curr. Top. Med. 53, 457–465 (1999). from the phase 3, multicentre, randomised, double-​
Chem. 6, 1707–1714 (2006). 40. McCoy, M. K. & Tansey, M. G. TNF signaling inhibition blind, placebo-​controlled PSUMMIT-1 and PSUMMIT-2
19. Maini, R. et al. Infliximab (chimeric anti-​tumour in the CNS: implications for normal brain function and trials. Ann. Rheum. Dis. 73, 1000–1006 (2014).
necrosis factor α monoclonal antibody) versus placebo neurodegenerative disease. J. Neuroinflammation 5, 62. Kavanaugh, A. et al. Efficacy and safety of ustekinumab
in rheumatoid arthritis patients receiving concomitant 45 (2008). in psoriatic arthritis patients with peripheral arthritis
methotrexate: a randomised phase III trial. ATTRACT 41. Kirkham, B. W., Kavanaugh, A. & Reich, K. Interleukin- and physician-​reported spondylitis: post-​hoc analyses
Study Group. Lancet 354, 1932–1939 (1999). 17A: a unique pathway in immune-​mediated diseases: from two phase III, multicentre, double-​blind, placebo-​
20. Weinblatt, M. E. et al. Adalimumab, a fully human psoriasis, psoriatic arthritis and rheumatoid arthritis. controlled studies (PSUMMIT-1/PSUMMIT-2).
anti-​tumor necrosis factor α monoclonal antibody, Immunology 141, 133–142 (2014). Ann. Rheum. Dis. 75, 1984–1988 (2016).
for the treatment of rheumatoid arthritis in patients 42. Menon, B. et al. Interleukin-17+CD8+ T cells are 63. Sandborn, W. J. et al. Ustekinumab induction and
taking concomitant methotrexate: the ARMADA trial. enriched in the joints of patients with psoriatic maintenance therapy in refractory Crohn’s disease.
Arthritis Rheum. 48, 35–45 (2003). arthritis and correlate with disease activity and joint N. Engl. J. Med. 367, 1519–1528 (2012).
21. Keystone, E. C. et al. Golimumab, a human antibody to damage progression. Arthritis Rheumatol. 66, 64. Feagan, B. G. et al. Ustekinumab as induction and
tumour necrosis factor α given by monthly subcutaneous 1272–1281 (2014). maintenance therapy for Crohn’s disease. N. Engl.
injections, in active rheumatoid arthritis despite 43. Lin, A. M. et al. Mast cells and neutrophils release J. Med. 375, 1946–1960 (2016).
methotrexate therapy: the GO-​FORWARD Study. IL-17 through extracellular trap formation in psoriasis. 65. McInnes, I. B. et al. Efficacy and safety of ustekinumab
Ann. Rheum. Dis. 68, 789–796 (2009). J. Immunol. 187, 490–500 (2011). in patients with active psoriatic arthritis: 1 year results
22. Moreland, L. W. et al. Etanercept therapy in 44. Genovese, M. C. et al. Efficacy and safety of of the phase 3, multicentre, double-​blind, placebo-​
rheumatoid arthritis. A randomized, controlled trial. secukinumab in patients with rheumatoid arthritis: controlled PSUMMIT 1 trial. Lancet 382, 780–789
Ann. Intern. Med. 130, 478–486 (1999). a phase II, dose-​finding, double-​blind, randomised, (2013).
23. Keystone, E. et al. Certolizumab pegol plus placebo controlled study. Ann. Rheum. Dis. 72, 66. Araujo, E. G. et al. Effects of ustekinumab versus tumor
methotrexate is significantly more effective than 863–869 (2013). necrosis factor inhibition on enthesitis: results from the
placebo plus methotrexate in active rheumatoid 45. Fujino, S. et al. Increased expression of interleukin 17 Enthesial Clearance in Psoriatic Arthritis (ECLIPSA)
arthritis: findings of a fifty-​two-week, phase III, in inflammatory bowel disease. Gut 52, 65–70 study. Semin. Arthritis Rheum. 48, 632–637 (2019).
multicenter, randomized, double-​blind, placebo-​ (2003). 67. Ibler, E. & Gordon, K. B. IL-23 inhibitors for moderate-​
controlled, parallel-​group study. Arthritis Rheum. 58, 46. Hueber, W. et al. Secukinumab, a human anti-​IL-17A to-severe psoriasis. Semin. Cutan. Med. Surg. 37,
3319–3329 (2008). monoclonal antibody, for moderate to severe Crohn’s 158–162 (2018).
24. Antoni, C. et al. Infliximab improves signs and disease: unexpected results of a randomised, double-​ 68. Fotiadou, C., Lazaridou, E., Sotiriou, E. & Ioannides, D.
symptoms of psoriatic arthritis: results of the IMPACT blind placebo-​controlled trial. Gut 61, 1693–1700 Targeting IL-23 in psoriasis: current perspectives.
2 trial. Ann. Rheum. Dis. 64, 1150–1157 (2005). (2012). Psoriasis 8, 1–5 (2018).

www.nature.com/nrrheum
Reviews

69. US National Library of Medicine. ClinicalTrials.gov 92. Dayer, J. M. & Bresnihan, B. Targeting interleukin-1 in 116. US National Library of Medicine. ClinicalTrials.gov
https://clinicaltrials.gov/ct2/show/NCT01094093 the treatment of rheumatoid arthritis. Arthritis Rheum. https://clinicaltrials.gov/ct2/show/NCT03104374
(2017). 46, 574–578 (2002). (2019).
70. Reich, K. et al. Efficacy and safety of mirikizumab 93. Dayer, J. M. Interleukin 1 or tumor necrosis factor-​ 117. Papp, K. et al. Phase 2 trial of selective tyrosine kinase
(LY3074828) in the treatment of moderate-​to-severe alpha: which is the real target in rheumatoid arthritis? 2 inhibition in psoriasis. N. Engl. J. Med. 379,
plaque psoriasis: results from a randomized phase II J. Rheumatol. Suppl. 65, 10–15 (2002). 1313–1321 (2018).
study. Br. J. Dermatol. 181, 88–95 (2019). 94. Gibbs, A. et al. Anakinra (Kineret) in psoriasis and 118. Pucci, E. et al. Natalizumab for relapsing remitting
71. Mease P. J. et al. Efficacy and safety of risankizumab, psoriatic arthritis: a single-​center, open-​label, pilot multiple sclerosis. Cochrane Database Syst. Rev. https://
a selective IL-23p19 inhibitor, in patients with active study. Arthritis Res. Ther. 7, 68 (2005). doi.org/10.1002/14651858.CD007621.pub2 (2011).
psoriatic arthritis over 24 weeks: results from a 95. Gul, A. et al. Efficacy and safety of canakinumab 119. Guagnozzi, D. & Caprilli, R. Natalizumab in the
phase 2 trial [OP0307]. Ann. Rheum. Dis. 77, in adolescents and adults with colchicine-​resistant treatment of Crohn’s disease. Biologics 2, 275–284
200–201 (2018). familial Mediterranean fever. Arthritis Res. Ther. 17, (2008).
72. Langley, R. G. et al. FRI0445 Tildrakizumab treatment 243 (2015). 120. Feagan, B. G. et al. Vedolizumab as induction and
improved measures of psoriatic arthritis in adults with 96. De Benedetti, F. et al. Canakinumab for the treatment maintenance therapy for ulcerative colitis. N. Engl.
chronic plaque psoriasis. Ann. Rheum. Dis. 75, of autoinflammatory recurrent fever syndromes. J. Med. 369, 699–710 (2013).
596–597, (2016). N. Engl. J. Med. 378, 1908–1919 (2018). 121. Sandborn, W. J. et al. Vedolizumab as induction and
73. Feagan, B. G. et al. Risankizumab in patients with 97. Hashkes, P. J. et al. Rilonacept for colchicine-​resistant maintenance therapy for Crohn’s disease. N. Engl.
moderate to severe Crohn’s disease: an open-​label or -intolerant familial Mediterranean fever: J. Med. 369, 711–721 (2013).
extension study. Lancet Gastroenterol. Hepatol. 3, a randomized trial. Ann. Intern. Med. 157, 533–541 122. Papp, K. A., Caro, I., Leung, H. M., Garovoy, M. &
671–680 (2018). (2012). Mease, P. J. Efalizumab for the treatment of psoriatic
74. Sands, B. E. et al. Efficacy and safety of MEDI2070, 98. Ben-​Zvi, I. et al. Anakinra for colchicine-​resistant arthritis. J. Cutan. Med. Surg. 11, 57–66 (2007).
an antibody against interleukin 23, in patients with familial Mediterranean fever: a randomized, double-​ 123. Boehncke, W. H. Efalizumab in the treatment of
moderate to severe Crohn’s disease: a phase 2a study. blind, placebo-​controlled trial. Arthritis Rheumatol. psoriasis. Biologics 1, 301–309 (2007).
Gastroenterology 153, 77–86.e6 (2017). 69, 854–862 (2017). 124. Mease, P. J. & Reich, K. Alefacept in Psoriatic Arthritis
75. Sandborn, W. J. et al. Efficacy and safety of anti-​ 99. O’Shea, J. J. et al. The JAK-​STAT pathway: impact Study Group. Alefacept with methotrexate for
interleukin-23 therapy with mirikizumab (LY3074828) on human disease and therapeutic intervention. treatment of psoriatic arthritis: open-​label extension
in patients with moderate-​to-severe ulcerative colitis in Annu. Rev. Med. 66, 311–328 (2015). of a randomized, double-​blind, placebo-​controlled
a phase 2 study [abstract 882]. Gastroenterology 154 100. Winthrop, K. L. The emerging safety profile of JAK study. J. Am. Acad. Dermatol. 60, 402–411 (2009).
(Suppl.), S-1360–S-1361 (2018). inhibitors in rheumatic disease. Nat. Rev. Rheumatol. 125. Kavanaugh, A. F. et al. A phase I/II open label study of
76. Kim, W. et al. The role of IL-12 in inflammatory activity 13, 320 (2017). the safety and efficacy of an anti-​ICAM-1 (intercellular
of patients with rheumatoid arthritis (RA). Clin. Exp. 101. Mease, P. et al. Tofacitinib or adalimumab versus adhesion molecule-1; CD54) monoclonal antibody
Immunol. 119, 175–181 (2000). placebo for psoriatic arthritis. N. Engl. J. Med. 377, in early rheumatoid arthritis. J. Rheumatol. 23,
77. Zaky, D. S. & El-​Nahrery, E. M. Role of interleukin-23 1537–1550 (2017). 1338–1344 (1996).
as a biomarker in rheumatoid arthritis patients and its 102. Kuo, C. M., Tung, T. H., Wang, S. H. & Chi, C. C. 126. US National Library of Medicine. ClinicalTrials.gov
correlation with disease activity. Int. Immunopharmacol. Efficacy and safety of tofacitinib for moderate-​ https://clinicaltrials.gov/ct2/show/NCT00083759
31, 105–108 (2016). to-severe plaque psoriasis: a systematic review and (2016)
78. Smolen, J. S. et al. A randomised phase II study meta-​analysis of randomized controlled trials. J. Eur. 127. Mosli, M. H. & Feagan, B. G. Vedolizumab for Crohn’s
evaluating the efficacy and safety of subcutaneously Acad. Dermatol. Venereol. 32, 355–362 (2018). disease. Expert Opin. Biol. Ther. 13, 455–463 (2013).
administered ustekinumab and guselkumab in 103. Sandborn, W. J. et al. Tofacitinib as induction and 128. Varkas, G. et al. An induction or flare of arthritis
patients with active rheumatoid arthritis despite maintenance therapy for ulcerative colitis. N. Engl. and/or sacroiliitis by vedolizumab in inflammatory
treatment with methotrexate. Ann. Rheum. Dis. 76, J. Med. 376, 1723–1736 (2017). bowel disease: a case series. Ann. Rheum. Dis. 76,
831–839 (2017). 104. van Vollenhoven, R. F. et al. Tofacitinib or adalimumab 878–881 (2017).
79. US National Library of Medicine. ClinicalTrials.gov versus placebo in rheumatoid arthritis. N. Engl. J. Med. 129. Garcia-​Vicuna, R. & Brown, M. A. Vedolizumab for
https://clinicaltrials.gov/ct2/show/NCT02407223 367, 508–519 (2012). inflammatory bowel disease: a two-​edge sword in the
(2019). 105. van der Heijde, D. et al. Tofacitinib (CP-690,550) gut-​joint/enthesis axis. Rheumatology 58, 937–939
80. Merola, J. F., Espinoza, L. R. & Fleischmann, R. in patients with rheumatoid arthritis receiving (2019).
Distinguishing rheumatoid arthritis from psoriatic methotrexate: twelve-​month data from a twenty-​four- 130. Paccou, J. et al. OP0029 Clinical effect of vedolizumab
arthritis. RMD Open 4, e000656 (2018). month phase III randomized radiographic study. on articular manifestations in patients with
81. Przepiera-​Bedzak, H., Fischer, K. & Brzosko, M. Serum Arthritis Rheum. 65, 559–570 (2013). spondyloarthritis associated with inflammatory bowel
IL-6 and IL-23 levels and their correlation with 106. Fleischmann, R. et al. Placebo-​controlled trial of disease. Ann. Rheum. Dis. 77, 64–65 (2018).
angiogenic cytokines and disease activity in ankylosing tofacitinib monotherapy in rheumatoid arthritis. 131. Orlando, A. et al. Clinical benefit of vedolizumab
spondylitis, psoriatic arthritis, and SAPHO syndrome. N. Engl. J. Med. 367, 495–507 (2012). on articular manifestations in patients with active
Mediators Inflamm. 2015, 785705 (2015). 107. Gladman, D. et al. Tofacitinib for psoriatic arthritis spondyloarthritis associated with inflammatory bowel
82. Tesmer, L. A., Lundy, S. K., Sarkar, S. & Fox, D. A. in patients with an inadequate response to TNF disease. Ann. Rheum. Dis. 76, e31 (2017).
Th17 cells in human disease. Immunol. Rev. 223, Inhibitors. N. Engl. J. Med. 377, 1525–1536 (2017). 132. Mease, P. J., Gladman, D. D. & Keystone, E. C.
87–113 (2008). 108. Tanaka, Y., Maeshima, K. & Yamaoka, K. In vitro and Alefacept in Psoriatic Arthritis Study Group. Alefacept
83. Mease, P. J. et al. The efficacy and safety of in vivo analysis of a JAK inhibitor in rheumatoid in combination with methotrexate for the treatment
clazakizumab, an anti-​interleukin-6 monoclonal arthritis. Ann. Rheum. Dis. 71, i70–i74 (2012). of psoriatic arthritis: results of a randomized, double-​
antibody, in a phase IIb study of adults with active 109. Fleischmann, R. et al. Phase IIb dose-​ranging study blind, placebo-​controlled study. Arthritis Rheum. 54,
psoriatic arthritis. Arthritis Rheumatol. 68, 2163–2173 of the oral JAK inhibitor tofacitinib (CP-690,550) or 1638–1645 (2006).
(2016). adalimumab monotherapy versus placebo in patients 133. Ellis, C. N. & Krueger, G. G. Alefacept Clinical Study
84. Sieper, J., Porter-​Brown, B., Thompson, L., Harari, O. with active rheumatoid arthritis with an inadequate Group. Treatment of chronic plaque psoriasis by
& Dougados, M. Assessment of short-​term response to disease-​modifying antirheumatic drugs. selective targeting of memory effector T lymphocytes.
symptomatic efficacy of tocilizumab in ankylosing Arthritis Rheum. 64, 617–629 (2012). N. Engl. J. Med. 345, 248–255 (2001).
spondylitis: results of randomised, placebo-​controlled 110. Hutmacher, M. M. et al. Evaluating dosage optimality 134. Malizia, G. et al. Expression of leukocyte adhesion
trials. Ann. Rheum. Dis. 73, 95–100 (2014). for tofacitinib, an oral Janus kinase inhibitor, in molecules by mucosal mononuclear phagocytes in
85. Sieper, J. et al. Sarilumab for the treatment of plaque psoriasis, and the influence of body weight. inflammatory bowel disease. Gastroenterology 100,
ankylosing spondylitis: results of a Phase II, CPT Pharmacomet. Syst. Pharmacol. 6, 322–330 150–159 (1991).
randomised, double-​blind, placebo-​controlled study (2017). 135. Davies, M. E., Sharma, H. & Pigott, R. ICAM-1
(ALIGN). Ann. Rheum. Dis. 74, 1051–1057 (2015). 111. Panes, J. et al. Tofacitinib for induction and maintenance expression on chondrocytes in rheumatoid arthritis:
86. Smolen, J. S. et al. Effect of interleukin-6 receptor therapy of Crohn’s disease: results of two phase IIb induction by synovial cytokines. Mediators Inflamm. 1,
inhibition with tocilizumab in patients with rheumatoid randomised placebo-​controlled trials. Gut 66, 71–74 (1992).
arthritis (OPTION study): a double-​blind, placebo-​ 1049–1059 (2017). 136. Wang, L., Ding, Y., Guo, X. & Zhao, Q. Role and
controlled, randomised trial. Lancet 371, 987–997 112. van der Heijde, D. et al. Tofacitinib in patients with mechanism of vascular cell adhesion molecule-1 in the
(2008). ankylosing spondylitis: a phase II, 16-week, randomised, development of rheumatoid arthritis. Exp. Ther. Med.
87. De Benedetti, F. et al. Randomized trial of tocilizumab placebo-​controlled, dose-​ranging study. Ann. Rheum. 10, 1229–1233 (2015).
in systemic juvenile idiopathic arthritis. N. Engl. J. Med. Dis. 76, 1340–1347 (2017). 137. Gladman, D. D. et al. Therapeutic benefit of
367, 2385–2395 (2012). 113. van der Heijde, D. et al. Efficacy and safety of filgotinib, apremilast on enthesitis and dactylitis in patients with
88. Fujikawa, K. et al. Interleukin-6 targeting therapy in a selective Janus kinase 1 inhibitor, in patients with psoriatic arthritis: a pooled analysis of the PALACE
familial Mediterranean fever. Clin. Exp. Rheumatol. active ankylosing spondylitis (TORTUGA): results from 1-3 studies. RMD Open 4, e000669 (2018).
31, 150–151 (2013). a randomised, placebo-​controlled, phase 2 trial. 138. Kavanaugh, A. et al. Longterm (52-week) results of
89. Ravindran, J. S. et al. Interleukin 1α, interleukin 1β Lancet 392, 2378–2387 (2018). a phase III randomized, controlled trial of apremilast
and interleukin 1 receptor gene polymorphisms in 114. Mease, P. et al. Efficacy and safety of filgotinib, a in patients with psoriatic arthritis. J. Rheumatol. 42,
psoriatic arthritis. Rheumatology 43, 22–26 (2004). selective Janus kinase 1 inhibitor, in patients with 479–488 (2015).
90. Mertens, M. & Singh, J. A. Anakinra for rheumatoid active psoriatic arthritis (EQUATOR): results from 139. Genovese, M. C. et al. Apremilast in patients with
arthritis. Cochrane Database Syst. Rev., CD005121, a randomised, placebo-​controlled, phase 2 trial. active rheumatoid arthritis: a phase II, multicenter,
https://doi.org/10.1002/14651858.CD005121.pub3 Lancet 392, 2367–2377 (2018). randomized, double-​blind, placebo-​controlled,
(2009). 115. Smolen, J. S. et al. Patient-​reported outcomes from a parallel-​group study. Arthritis Rheumatol. 67,
91. Singh, J. A. et al. A network meta-​analysis of randomised phase III study of baricitinib in patients 1703–1710 (2015).
randomized controlled trials of biologics for rheumatoid with rheumatoid arthritis and an inadequate response 140. US National Library of Medicine. ClinicalTrials.gov
arthritis: a Cochrane overview. CMAJ 181, 787–796 to biological agents (RA-​BEACON). Ann. Rheum. Dis. https://clinicaltrials.gov/ct2/show/NCT01583374
(2009). 76, 694–700 (2017). (2019).

Nature Reviews | Rheumatology


Reviews

141. US National Library of Medicine. ClinicalTrials.gov and clinical features. Arthritis Res. Ther. 14, R93 171. Veale, D. J. et al. The rationale for Janus kinase
https://clinicaltrials.gov/ct2/show/NCT02289417 (2012). inhibitors for the treatment of spondyloarthritis.
(2019) 156. Zhu, J., Yamane, H. & Paul, W. E. Differentiation of Rheumatology 58, 197–205 (2019).
142. Moreland, L., Bate, G. & Kirkpatrick, P. Abatacept. effector CD4 T cell populations*. Annu. Rev. Immunol. 172. Li, H., Zuo, J. & Tang, W. Phosphodiesterase-4
Nat. Rev. Drug Discov. 5, 185–186 (2006). 28, 445–489 (2010). inhibitors for the treatment of inflammatory diseases.
143. Maxwell, L. J. & Singh, J. A. Abatacept for rheumatoid 157. Szentpetery, A. et al. Abatacept reduces synovial Front. Pharmacol. 9, 1048 (2018).
arthritis: a Cochrane systematic review. J. Rheumatol. regulatory T-​cell expression in patients with psoriatic 173. Ospelt, C. Synovial fibroblasts in 2017. RMD Open 3,
37, 234–245 (2010). arthritis. Arthritis Res. Ther. 19, 158 (2017). e000471 (2017).
144. Mease, P. J. et al. Efficacy and safety of abatacept, 158. Ciccia, F. et al. Interleukin-9 and T helper type 9 cells 174. Espinoza, L. R. et al. Fibroblast function in psoriatic
a T-cell modulator, in a randomised, double-​blind, in rheumatic diseases. Clin. Exp. Immunol. 185, arthritis. II. Increased expression of beta platelet
placebo-​controlled, phase III study in psoriatic 125–132 (2016). derived growth factor receptors and increased
arthritis. Ann. Rheum. Dis. 76, 1550–1558 (2017). 159. Mitra, A., Raychaudhuri, S. K. & Raychaudhuri, S. P. production of growth factor and cytokines. J. Rheumatol.
145. Edwards, J. C. et al. Efficacy of B-​cell-targeted therapy Functional role of IL-22 in psoriatic arthritis. Arthritis 21, 1507–1511 (1994).
with rituximab in patients with rheumatoid arthritis. Res. Ther. 14, R65 (2012). 175. Goldring, M. B. & Berenbaum, F. The regulation of
N. Engl. J. Med. 350, 2572–2581 (2004). 160. Soare, A. et al. Cutting edge: Homeostasis of innate chondrocyte function by proinflammatory mediators:
146. Cohen, S. B. et al. Rituximab for rheumatoid arthritis lymphoid cells is imbalanced in psoriatic arthritis. prostaglandins and nitric oxide. Clin. Orthop.
refractory to anti-​tumor necrosis factor therapy: results J. Immunol. 200, 1249–1254 (2018). Relat. Res., S37-S46, https://doi.org/10.1097/
of a multicenter, randomized, double-​blind, placebo-​ 161. Dunphy, S. & Gardiner, C. M. NK cells and psoriasis. 01.blo.0000144484.69656.e4 (2004).
controlled, phase III trial evaluating primary efficacy J. Biomed. Biotechnol. 2011, 248317 (2011). 176. Burrage, P. S., Mix, K. S. & Brinckerhoff, C. E. Matrix
and safety at twenty-​four weeks. Arthritis Rheum. 54, 162. Chiba, A. et al. Mucosal-​associated invariant T cells metalloproteinases: role in arthritis. Front. Biosci. 11,
2793–2806 (2006). promote inflammation and exacerbate disease in 529–543 (2006).
147. Memon, A. B. et al. Long-​term safety of rituximab murine models of arthritis. Arthritis Rheum. 64, 177. Lin, E. A. & Liu, C. J. The role of ADAMTSs in arthritis.
induced peripheral B-​cell depletion in autoimmune 153–161 (2012). Protein Cell 1, 33–47 (2010).
neurological diseases. PLoS One 13, e0190425 (2018). 163. Teunissen, M. B. M. et al. The IL-17A-​producing CD8+ 178. Rahimi, H. & Ritchlin, C. T. Altered bone biology in
148. Song, I. H. et al. Different response to rituximab in T-​cell population in psoriatic lesional skin comprises psoriatic arthritis. Curr. Rheumatol. Rep. 14, 349–357
tumor necrosis factor blocker-​naive patients with active mucosa-​associated invariant T cells and conventional (2012).
ankylosing spondylitis and in patients in whom tumor T cells. J. Invest. Dermatol. 134, 2898–2907 (2014). 179. Mensah, K. A., Schwarz, E. M. & Ritchlin, C. T. Altered
necrosis factor blockers have failed: a twenty-​four-week 164. Jongbloed, S. L. et al. Enumeration and phenotypical bone remodeling in psoriatic arthritis. Curr. Rheumatol.
clinical trial. Arthritis Rheum. 62, 1290–1297 (2010). analysis of distinct dendritic cell subsets in psoriatic Rep. 10, 311–317 (2008).
149. Jimenez-​Boj, E. et al. Rituximab in psoriatic arthritis: arthritis and rheumatoid arthritis. Arthritis Res. Ther. 180. Coury, F., Peyruchaud, O. & Machuca-​Gayet, I.
an exploratory evaluation. Ann. Rheum. Dis. 71, 8, R15 (2006). Osteoimmunology of bone loss in inflammatory
1868–1871 (2012). 165. Lande, R. et al. Characterization and recruitment of rheumatic diseases. Front. Immunol. 10, 679 (2019).
150. Jung, N. et al. An open-​label pilot study of the efficacy plasmacytoid dendritic cells in synovial fluid and tissue 181. Delaisse, J. M. et al. Matrix metalloproteinases
and safety of anakinra in patients with psoriatic of patients with chronic inflammatory arthritis. (MMP) and cathepsin K contribute differently to
arthritis refractory to or intolerant of methotrexate J. Immunol. 173, 2815–2824 (2004). osteoclastic activities. Microsc. Res. Tech. 61,
(MTX). Clin. Rheumatol. 29, 1169–1173 (2010). 166. Wenink, M. H. et al. Impaired dendritic cell 504–513 (2003).
151. Veale, D., Rogers, S. & Fitzgerald, O. proinflammatory cytokine production in psoriatic
Immunolocalization of adhesion molecules in psoriatic arthritis. Arthritis Rheum. 63, 3313–3322 (2011). Author contributions
arthritis, psoriatic and normal skin. Br. J. Dermatol. 167. Danning, C. L. et al. Macrophage-​derived cytokine and Both authors contributed equally to all aspects of the
132, 32–38 (1995). nuclear factor κB p65 expression in synovial membrane article.
152. Riccieri, V. et al. Adhesion molecule expression in the and skin of patients with psoriatic arthritis. Arthritis
synovial membrane of psoriatic arthritis. Ann. Rheum. Rheum. 43, 1244–1256 (2000). Competing interests
Dis. 61, 569–570 (2002). 168. Vandooren, B. et al. Absence of a classically activated The authors declare no competing interests.
153. Riccieri, V. et al. [Immunohistochemical analysis of macrophage cytokine signature in peripheral
the expression of main adhesion molecules and tumor spondylarthritis, including psoriatic arthritis. Arthritis Peer review information
necrosis factors in the synovial membrane of psoriatic Rheum. 60, 966–975 (2009). Nature Reviews Rheumatology thanks S. D’Angelo and the
arthritis]. Reumatismo 55, 164–170 (2003). 169. Arango Duque, G. & Descoteaux, A. Macrophage other, anonymous, reviewer(s) for their contribution to
154. Diani, M., Altomare, G. & Reali, E. T cell responses in cytokines: involvement in immunity and infectious the peer review of this work.
psoriasis and psoriatic arthritis. Autoimmun. Rev. 14, diseases. Front. Immunol. 5, 491 (2014).
286–292 (2015). 170. Noordenbos, T. et al. Interleukin-17-positive mast cells Publisher’s note
155. Celis, R. et al. Synovial cytokine expression in psoriatic contribute to synovial inflammation in spondylarthritis. Springer Nature remains neutral with regard to jurisdictional
arthritis and associations with lymphoid neogenesis Arthritis Rheum. 64, 99–109 (2012). claims in published maps and institutional affiliations.

www.nature.com/nrrheum

S-ar putea să vă placă și