ArticlePDF AvailableLiterature Review

Systemic effects of IL-6 blockade in rheumatoid arthritis beyond the joints

Authors:

Abstract and Figures

Interleukin (IL)-6 is produced locally in response to an inflammatory stimulus, and is able to induce systemic manifestations at distance from the site of inflammation. Its unique signaling mechanism, including classical and trans-signaling pathways, leads to a major expansion in the number of cell types responding to IL-6. This pleiotropic cytokine is a key factor in the pathogenesis of rheumatoid arthritis (RA) and is involved in many extra-articular manifestations that accompany the disease. Thus, IL-6 blockade is associated with various biological effects beyond the joints. In this review, the systemic effects of IL-6 in RA comorbidities and the consequences of its blockade will be discussed, including anemia of chronic disease, cardiovascular risks, bone and muscle functions, and neuro-psychological manifestations.
Content may be subject to copyright.
Cytokine 149 (2022) 155742
Available online 20 October 2021
1043-4666/© 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Systemic effects of IL-6 blockade in rheumatoid arthritis beyond the joints
Matthias Jarlborg
a
,
b
, Cem Gabay
a
,
*
a
Division of Rheumatology, University Hospital of Geneva, and Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland
b
VIB-UGent Center for Inammation Research and Ghent University, Ghent, Belgium
ARTICLE INFO
Keywords:
Interleukin-6
IL-6 blockade
Rheumatoid arthritis
Systemic effects
ABSTRACT
Interleukin (IL)-6 is produced locally in response to an inammatory stimulus, and is able to induce systemic
manifestations at distance from the site of inammation. Its unique signaling mechanism, including classical and
trans-signaling pathways, leads to a major expansion in the number of cell types responding to IL-6. This
pleiotropic cytokine is a key factor in the pathogenesis of rheumatoid arthritis (RA) and is involved in many
extra-articular manifestations that accompany the disease. Thus, IL-6 blockade is associated with various bio-
logical effects beyond the joints. In this review, the systemic effects of IL-6 in RA comorbidities and the con-
sequences of its blockade will be discussed, including anemia of chronic disease, cardiovascular risks, bone and
muscle functions, and neuro-psychological manifestations.
1. Introduction
Interleukin (IL-) 6 is a pleiotropic pro-inammatory cytokine which
plays an important role in rheumatoid arthritis (RA) and its comorbid-
ities. As a chief stimulator of the production of most acute-phase pro-
teins, IL-6 is involved in acute inammation [1]. IL-6 is also a key player
in immune regulation, including the transition between acute and
chronic inammation [2]. Since the discovery of its role as a T-cell factor
for B cell differentiation [3] and its identication in 1986 [4], IL-6
targeting has considerably improved the management of numerous in-
ammatory rheumatic diseases, and in particular RA [5].
IL-6 expression is up-regulated by various transcription factors,
including NF-κB which is activated by other proinammatory cytokines
(such as IL-1β, TNF
α
and IL-17) [6], and by Toll-like receptors (TLRs)-
mediated signals [7]. In response to cellular stress signals, most leuco-
cytes and stromal cells are able to produce IL-6. The pleiotropic effects of
this cytokine can be explained by its peculiar signaling pathway,
engaging an heterodimer receptor formed by 1) its specic receptor, IL-
6R, expressed in a restricted manner and 2) the co-receptor, the glyco-
protein gp130 that is ubiquitously expressed in different cell types and
also used by other members of the IL-6 family of cytokines [8]. The
receptor IL-6R exists indeed in both transmembrane (IL-6R) and soluble
(sIL-6R) forms [9,10]. The classic signaling involves binding to
membrane-bound IL-6R. This receptor is only present in some cell types
such as hepatocytes, neutrophils, monocytes, and some lymphocytes.
The binding to IL-6R is followed by its dimerization with gp130 which,
in turn, allows signal transduction. Most importantly, the co-receptor
gp130 can also be activated by binding to soluble IL-6-sIL-6R com-
plexes (a process called trans-signalization). This signaling pathway leads
to a major extension in the number of cell types that can respond to IL-6.
Indeed, when circulating IL-6 levels exceed circulating sgp130 levels, IL-
6 can bind to sIL-6R and acts systemically like a hormone. Both classic
and trans-signaling pathways lead to signal transduction through the
Janus kinase (JAK) signal transducer and activator of transcription 3
(STAT3) pathway, as well as the mitogen-activated protein kinase
(MAPK) cascade [11,12].
IL-6 has numerous metabolic and homeostatic effects distant from
the site of inammation. The use of anti-IL-6R drugs in rheumatoid
arthritis (RA) for more than a decade [13] has led to a better under-
standing of the broad spectrum of action of this cytokine. RA is a
frequent autoimmune disease, characterized by a chronic and progres-
sive inammation of various organs, mostly the joints, and immune
dysregulation. Many cytokines, including IL-6, are involved in the
pathogenesis and maintenance of the disease [14]. Recently, the new
concept of signature cytokine hub considers IL-6 together with TNF-
α
as
critical cytokines node in RA [15]. In addition, RA is associated with an
increased risk of developing comorbid conditions in which IL-6 is also
directly involved [16]. These comorbidities include cardiovascular dis-
eases, infections, osteoporosis, depression, neoplasia, anemia (among
others), and are associated with higher morbidity and mortality [17]. In
this narrative review, we will discuss the key roles of IL-6 in most RA
comorbidities, namely anemia of chronic disease, cardiovascular risks,
* Corresponding author.
E-mail address: cem.gabay@unige.ch (C. Gabay).
Contents lists available at ScienceDirect
Cytokine
journal homepage: www.elsevier.com/locate/cytokine
https://doi.org/10.1016/j.cyto.2021.155742
Received 5 September 2021; Received in revised form 13 September 2021; Accepted 5 October 2021
Cytokine 149 (2022) 155742
2
bone and muscle dysfunctions, and neuro-psychological manifestations.
First, an overview of the effects of IL-6 in systemic inammation and in
systemic manifestations associated with RA will be exposed. The sub-
sequent consequences of its blockade beyond the joints will be then
discussed, whether benecial or detrimental.
2. IL-6 and systemic inammation
IL-6 plays a major role in systemic inammation and immunity. It is
rapidly synthesized and released by myeloid cells in response to various
danger signals and cytokines such as IL-1 and TNF-
α
. From the local site
of inammation, IL-6 is able to reach key organs through the blood-
stream and induces systemic responses. This process is referred to as the
acute-phase response [18]. With other cytokines, such as TNF-
α
, IL-1
and IFN-γ, IL-6 acts synergistically to induce inammatory systemic
responses [19]. However, IL-6, also known as hepatocyte-stimulary factor,
seems to have stronger and broader effects, especially on hepatocytes
[20].
Acute-phase response is accompanied by changes in plasma con-
centrations of multiple proteins [1], known as the acute-phase proteins,
the circulating levels of which change by at least 25% during inam-
mation. More specically, the levels of positive acute-phase proteins
increase, while plasma concentrations of negative acute-phase proteins
decrease (eg. albumin, transthyretin, bronectin) during the acute-
phase response. Hepatocytes express both IL-6R and gp130 allowing a
prompt response to IL-6 through the classical signaling pathway [21].
Positive acute-phase proteins include C-reactive protein (CRP), com-
plement factors and mannose-binding lectin, serum amyloid A (SAA),
brinogen, ferritin, and hepcidin (among others). Their increase in the
bloodstream facilitates the elimination of pathogens and damaged cells.
Commonly used as a biomarker of inammation, CRP has also biologic
effects: it participates to complement activation, and functions as a
pattern recognition molecule able to opsonize pathogens or damaged
cells [22]. In addition to promote inammation, acute-phase proteins
such as brinogen and haptoglobin - are involved in coagulation and
healing. Mice decient for IL-6 exhibit severely impaired acute phase
protein production, and are unable to develop optimal responses to
trauma and certain types of pathogens [23]. Moreover, IL-6 and acute
phase response are also associated with changes in many physiological
and behavioral processes (such as fever, pain; hematopoietic and
metabolic perturbations; anorexia and somnolence). Fig. 1 summarizes
some of the numerous systemic effects of IL-6.
As mentioned in the introduction, IL-6 is not only involved in acute
inammation, but also prepares the transition to chronic and adaptive
responses. The rst steps of inammation are characterized by the inux
of neutrophils to the site of injury or infection. Their accumulation in-
duces the release of high amounts of IL-6 but also of sIL-6R by shedding
of its membrane-bound form [24,25]. The complex IL-6-sIL-6R-gp130 is
then able to stimulate stromal cells, mostly endothelial and smooth
muscle cells, but also synoviocytes [26]. Notably, trans-signaling pathway
of IL-6 is involved in endothelial activation and production monocyte
chemoattractant protein (MCP)-1 favoring transition from neutrophil to
monocyte recruitment [27]. In other terms, IL-6 trans-signaling promotes
secondary accumulation of monocytes to the site of inammation, which
is the hallmark of chronic inammation [2]. IL-6 is also involved in the
healing process by promoting differentiation of bone marrow-derived
monocytes towards macrophages and pro-resolving M2-like responses
[28,29]. Furthermore, IL-6 has anti-inammatory properties. Notably, it
stimulates the production of IL-1 and TNF-
α
antagonists such as IL-1Ra
and soluble p55 respectively [30].
Trans-signaling pathway has a pivotal role on lymphocytes and
adaptive immunity. IL-6 is involved in the differentiation of T helper
(Th) CD4+cells. Depending on the local microenvironment and
together with TGF-β and IL-23, IL-6 promotes Th17 polarization and
inhibits regulatory T cell (Treg) differentiation, favoring pro-
inammatory responses and mucosal immune defense [31,32]. IL-6
trans-signaling has also been shown to induce a rapid activation of
effector functions in cytotoxic CD8+T cells [33]. In addition, IL-6
Fig. 1. This gure summarizes the pleiotropic effects of IL-6 on various organs and tissues. The complexes formed by IL-6, its soluble receptor (sIL-6R) and by
glycoprotein (gp) 130 are able to induce various systemic effects distant from the initial site of inammation. Abbreviation: CRP (C-reactive protein), DAMPs
(damage-associated molecular patterns), HPA (hypothalamicpituitary-adrenal axis), IFN-γ (interferon gamma), PAMPs (pathogen-associated molecular patterns),
Tfh (follicular helper T cells), Th17 (T-helper 17 cell), TNF-
α
(Tumor necrosis factor-alpha). This gure is made with illustrations obtained from https://smart.servier.
com/.
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
3
contributes to B cells differentiation and activation. In association with
IL-2 and IL-10, IL-6 induces the maturation of plasmablasts into early
plasma cells [34]. Originally described as a B cell stimulatory factor, IL-6
promotes the production of immunoglobulins in vitro [35]. In transgenic
mice, overexpression of IL-6 induces plasmocytosis with hyper-
gammaglobulinemia [36]. Together with IL-21 and IL-23 [37,38], IL-6 is
indeed necessary for the differentiation of follicular helper T cells (Tfh)
which, in turn, can activate B cells in germinal centers [39,40,41]. Some
recent data suggest that gp130/STAT3 signaling is involved in the
development of tertiary lymphoid structures [42]. These ectopic
lymphoid tissues are particularly important in the pathogenesis of
autoimmune diseases and autoantibodies production [43].
Hence, IL-6 orchestrates the immune response from its rst innate
steps to the late adaptive stages, including its resolution. Dysregulation
and persistent IL-6 production leads to a chronic inammation state as
seen in many inammatory and autoimmune diseases, as well as in
cancers. IL-6-producing cardiac myxoma illustrates the clinical conse-
quences of its overexpression with development of fever, increased
acute-phase proteins, lymph node enlargement and autoimmune fea-
tures including polyarthritis and autoantibody production [44]. High
levels of IL-6 are also observed in the rheumatoid synovium [45], in
hyperplastic lymph nodes from Castlemans disease [46], or in multiple
myeloma [47]. These ndings led to the development of treatments
targeting IL-6. The rst clinical trials were conducted in patients with
multiple myeloma, with mitigated results [48]. Later, a humanized anti-
IL-6R antibody (tocilizumab) was successfully used to treat chronic in-
ammatory symptoms in Castlemans disease [49]. Since the late 2000
s, IL-6R inhibitors, including tocilizumab and sarilumab have been
licensed for the management of RA refractory to conventional synthetic
disease modifying anti-rheumatic drugs (DMARDs). The use of IL-6R
inhibitors was associated with improvement of composite scores of
disease activity as well as with inhibition of structural damage [5053].
Both tocilizumab and sarilumab were superior to adalimumab, a TNF
antagonist, in the control of inammatory manifestations when used as
monotherapy [5457]. More recently, tocilizumab was proven efca-
cious in the treatment of other inammatory diseases, including sys-
temic juvenile idiopathic arthritis, adult onset-Stills disease, and giant
cell arteritis [5860]. In addition to their benecial effects in the control
of inammatory manifestations, IL-6 inhibitors are characterized by
their ability to suppress acute-phase proteins. A rapid and sustained
normalization of CRP levels is indeed observed upon IL-6R inhibition
[61,53,62]. This is associated with clinical improvement and the
reduction of many biomarkers of disease severity [6365]. Tocilizumab
suppresses fever and CRP in RA patients undergoing surgery [66]. Thus,
by masking CRP elevation, IL-6 blockade rises major concerns regarding
interpretation of inammatory markers dependent on the acute-phase
response [67]. Besides improvements of systemic and joint inamma-
tion, IL-6 blockade is also associated with signicant benets on RA
comorbidities. In the following sections, we will discuss the role of IL-6
in RA comorbidities and the consequences of its blockade beyond the
joints.
3. Systemic benets of IL-6 blockade in rheumatoid arthritis
3.1. Hematological effects of IL-6
The changes in blood cell count accompanying inammation are
driven by cytokines, including IL-6 [68]. Anemia is the most frequent
extra-articular manifestation associated with RA. It affects 16% of newly
diagnosed RA patients in a British cohort [69] and has a lifetime prev-
alence of up to 60% according to older publications [70]. In addition to
its contribution to fatigue, anemia is an independent predictor of disease
severity and radiographic progression in RA [71]. Moreover, low he-
moglobin levels are associated with higher Health Assessment Ques-
tionnaire (HAQ) scores, indicative of physical disability [72]. Anemia of
chronic diseases (ACD, or inammatory anemia) is the main
presentation of anemia that occurs together with RA [73]. ACD is
typically normochromic, normocytic, and is due to iron sequestration.
IL-6 stimulates hepatocytes to produce proteins involved in iron meta-
bolism, mainly hepcidin [74], and ferritin [75]. Hepcidin, a hormone-
like peptide, inhibits intestinal iron absorption by binding to the ferro-
portin channel and promoting its internalization and degradation
[76,77]. Similarly, hepcidin inhibits the release of iron from reticulo-
endothelial cells. Serum hepcidin levels correlate with RA disease ac-
tivity, and are associated to RA comorbidities such as coronary
atherosclerosis and osteoporosis [78,79]. The increased ferritin levels
also participate to iron sequestration. Altogether, lower transferrin
saturation and lower serum iron concentrations result in ACD [80]. The
decrease in iron availability seems to be a general defense mechanism
against many pathogens [81]. Both anemia and increased brinogen
levels contribute to the elevation of erythrocyte sedimentation rate
(ESR) observed during inammation [82].
IL-6R inhibition counteracts the effects of IL-6 on iron metabolism
leading to greater iron availability. Tocilizumab induces a rapid and
sustained reduction of hepcidin levels in patients with Castleman disease
[83]. This has also been observed in RA, with improvement of anemia
[84]. In a cohort of RA patients treated with either tocilizumab or
iniximab (a TNF-inhibitor, TNFi), IL-6 blockade was more efcient to
reduce hepcidin levels and to improve anemia [85]. Hashimoto et al.
have demonstrated that tocilizumab is an independent factor associated
with the increase of hemoglobin levels [86]. In a post-hoc analysis of the
MONARCH study, sarilumab resulted in larger increase in hemoglobin
levels at week 12 and 24 compared to adalimumab. This effect was
associated with a larger decrease of hepcidin levels in the sarilumab
group. Correlations between hepcidin levels and disease activity
(including patient-reported outcomes) have been reported in RA, and
hepcidin appears to be a predictor of treatment efcacy of both sar-
ilumab and adalimumab at 24 weeks [65]. Thus, these studies suggest
that IL-6R inhibitors are treatments of choice for RA patients with ACD.
Thrombocytosis is involved in the promotion of hemostasis but also
in inammation, host defense and healing [87]. Inammatory throm-
bocytosis is also a direct consequence of IL-6 signaling. IL-6 stimulates
megakaryocytopoïeses through enhancement of thrombopoietin (TPO)
expression by hepatocytes [88,89]. Moreover, IL-6 has direct effects on
megakaryocytes differentiation in vitro [90]. In a mouse model, high IL-6
production resulted in thrombocytosis, and IL-6 blockade was followed
by platelet count normalization [91]. In RA, thrombocytosis correlates
with IL-6 levels and disease activity [92,93]. Of note, platelet count
decreases to a greater extent in RA patients treated with tocilizumab
compared to adalimumab [54]. In this clinical trial, low grade throm-
bocytopenia occurred also more frequently in the tocilizumab group
compared to adalimumab (9.3% vs 3.1%). However, no
thrombocytopenia-related complications (severe thrombocytopenia or
bleeding) were reported. Likewise, high platelet count together with low
hemoglobin levels seem to be good predictors for IL-6R inhibitor efcacy
[94].
Neutrophils are closely involved in IL-6 biology since they express IL-
6R and can secrete its soluble form in large amount, allowing trans-
signaling [24]. Data on the direct effects of IL-6 on neutrophils remain
however conicting. Clinical studies demonstrated that IL-6 blockade
induces a transient but signicant neutropenia [95]. Wright et al.
showed that tocilizumab does not induce an increase of neutrophil
apoptosis, neither affects neutrophil function [96]. Thus, one explana-
tion could be that IL-6R inhibitors increase neutrophil margination or
alter neutrophil trafcking [97]. Fortunately, there is no evidence for a
higher risk of serious infection secondary to tocilizumab induced-
neutropenia [98].
3.2. Effects of IL-6 on cardiovascular risks
RA is associated with excess of mortality compared to general pop-
ulation. The standardized mortality ratio (SMR) is estimated between
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
4
1.27 and 2.26, depending on populations studied [99,100]. Presence of
extra-articular manifestations constitutes the strongest predictor of
mortality [99]. Despite the signicant decline in mortality in general
population, the death rate in the paired cohort of RA patients appeared
to stay relatively at [101]. A more recent study in a large Korean cohort
still conrms the excess of mortality in RA, with a SMR of 1.65 (95% CI
1.441.87). Cardiovascular disease (CVD) accounted for 14.2% of the
causes of death in this study [102]. It is estimate that up to 50% of
premature deaths in RA are attributable to CVD [103].
RA confers a signicantly higher risk of coronary heart disease
(CHD) and congestive heart failure compared to general population
[104106]. RA patients have a 1.5 to 2-fold increased risk of CV events,
similar to the magnitude of risk in type 2 diabetes mellitus (DM) [107].
Recently, a large retrospective study showed a similar decline in 10-year
risk of acute myocardial infarction in both RA and non-RA populations,
indicating that the excess risk of CV events still persists [105].
Four major factors contribute to CV risk in RA. 1) The usual CV risks
factors (notably smoking, dyslipidemia, hypertension, obesity and DM)
coexist with a higher prevalence in RA [108,109]. 2) The use of some
treatments such as glucocorticoids and non-steroidal anti-inammatory
drugs, is associated with an increased CVD risk [110,111]. 3) The
presence of anti-citrullinated peptide antibodies (ACPAs) and rheuma-
toid factors (RFs) appeared to be independent risk factors for CVD and
CV mortality [112114]. 4) Finally, elevated RA disease activity with
persistent inammation represents an independent and major risk factor
for CVD [115,116].
In this context, IL-6 plays a role on several levels. Systemic inam-
mation and the acute phase response assessed by CRP levels and ESR
are indeed associated with atherosclerosis. There are also clear links
between IL-6 and CVD [117,118]. A meta-analysis showed that one
standard deviation increase in IL-6 levels is associated with an adjusted
relative risk of 1.25 (1.191.32) of non-fatal myocardial infarction or
CHD death [119]. Using Mendelian randomization analysis, a study
showed that single nucleotide polymorphisms of IL6R gene leading to a
decrease in CRP levels were associated with a decreased odd of CHD
events in the general population [120]. Indeed, IL-6 directly inuences
endothelium homeostasis and promotes the progression of atheroma.
Elevated serum IL-6 levels were associated with higher coronary artery
calcications [121]. While favoring endothelial activation and mono-
cyte recruitment to the atheroma, IL-6 together with angiotensin II in-
uence atheroma stability [122124].
The increased CV risk in RA is paradoxically associated with a
decrease of total cholesterol levels [125]. This phenomenon called lipid
paradox is probably due to the effects of IL-6 on lipid metabolism
[126]. IL-6 is involved in adipose tissue metabolism [127]. Notably,
high IL-6 levels are associated with an increased expression of lipopro-
tein(a) (Lp(a)), a known risk factor for premature atherosclerosis
[128130]. In RA, Lp(a) concentrations are elevated, and associated
with disease activity and inammatory biomarkers [131]. Resident
macrophages and adipocytes are important sources of IL-6 in adipose
tissue of obese patients, and contribute to insulin resistance [132,133].
A recent cohort study of RA patients showed that IL-6 was independently
associated with the incidence of type 2 DM [134].
In mice, IL-6 treatment induced an increase of very-low-density li-
poprotein receptor in various tissues, and decreased circulating total
cholesterol and triglyceride levels. In the same mice, anti-IL6R antibody
reversed these lipid level changes [135]. In RA patients, low-density
lipoprotein cholesterol (LDL-C) and high-density lipoprotein choles-
terol (HDL-C) increased more with tocilizumab compared to adalimu-
mab. However, known lipid and lipid-associated CV-risk biomarkers
(including HDL-SAA, secretory phospholipase A2 IIA and Lp(a))
decreased more with tocilizumab than with adalimumab [136]. Another
study, comparing levels of biomarkers associated with CVD in RA pa-
tients whose disease was insufciently controlled by TNF inhibitors,
showed a similar effect of different biological treatments on CV risk-
associated biomarkers. However, tocilizumab was associated with a
better improvement of Lp(a) level and leptin/adiponectin ratio [137].
Regarding insulin resistance, an observational study demonstrated a
signicant reduction of glycosylated hemoglobin (HbA1c) in RA pa-
tients treated with tocilizumab and TNFi. This decrease was numerically
higher for tocilizumab compared to TNFi [138]. Sarilumab was also
associated with decreased HbA1c levels in a post hoc analysis regroup-
ing three placebo-controlled phase III studies [139].
The overall effects of IL-6 blockade on CV outcomes appear favor-
able. A meta-analysis concluded that tocilizumab had a CV safety prole
similar to that of other biological agents, despite the increase in
cholesterol levels [140]. A retrospective post-hoc study in tocilizumab-
treated RA patients showed that the risk of major cardiac events was
not related to changes of lipid levels over the rst 24 months of therapy,
but rather to the control of disease activity [141]. Long-term exposition
to tocilizumab was associated with a decreased incidence of serious
infections and serious cardiac dysfunctions [142]. The development of
acute myocardial infarction in RA patients using tocilizumab tends to be
low, even compared to patients with TNFi and abatacept [143]. Long
term safety of sarilumab showed comparable results [144].
Moreover, IL-6 blockade seems to have some benecial effects on
cardiovascular system. A pilot study showed an improvement of
vascular endothelial function (assessed by brachial artery ow-mediated
vasodilatation and carotid to femoral pulse wave velocity) in RA patients
after 3 and 6 months of tocilizumab treatment [145]. In RA patients
without cardiac symptoms, tocilizumab improved cardiac ejection
fraction assessed by cardiac magnetic resonance [146]. Apart from
rheumatologic conditions, a large cohort of patients with heart failure
showed that circulating levels of IL-6 were signicantly increased in
more than half of patients with heart failure, and were associated with
poorer outcome [147]. These data support the use of IL-6 blockade in RA
patients with CV disease, and more generally, suggest that IL-6 inhibi-
tion could offer interesting perspectives in individuals with CV disease.
3.3. Inuence of IL-6 on neuroendocrine system
Patient-reported outcomes (PROs) include symptoms related to pain,
function and global assessment of disease activity, overlapping with the
concept of health-related quality of life [148]. RA is frequently associ-
ated with symptoms such as pain, fatigue and mood disorders that all
have a signicant impact on the quality of life [149151]. Thus, PROs
remain as relevant for disease activity assessment as traditional out-
comes based on less subjective variables [152155].
Fatigue and sleep disturbance are very common in RA, with a
prevalence 4 to 8-times higher than in the general population [156]. The
causal factors of fatigue are multifactorial and appear to be related more
to pain and impaired function than to inammation itself [157,158].
Pain accompanying RA is also multifactorial, involving local inam-
mation and joint damage, but also peripheral and central nociception
[159,160]. Same observations can be made for depression and mood
disorders that readily accompany RA [161]. Furthermore, fatigue, pain
and mood are inuencing each other, leading to mutual amplication of
these symptoms [162,163].
Neurohormonal perturbations are partly responsible for these
symptoms in RA, and are also driven by inammatory processes. It is
now well established that IL-6 is associated with neuroendocrine effects
[164,165]. Interestingly, increased IL-6 levels have been observed in
major depression [166,167]. In animal model, IL-6 knockout mice
exhibit abnormal behaviors characterized by a resistance to stress [168].
Neural cells are indeed able to respond to IL-6 through trans-signaling
[169171]. Cytokines, including IL-6, can activate the hypothal-
amicpituitary-adrenal (HPA) axis, resulting in increased levels of the
stress hormone, cortisol [172,173]. However, patients with RA have
decreased circulating cortisol concentrations, despite elevated IL-6
levels [174]. This may be due to hyporesponsive adrenal glands or to
an altered HPA response to IL-6 [175]. In human and rat, there are some
evidences that IL-6 and its circadian pattern of secretion are involved in
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
5
sleep regulation [176]. In addition, cerebral structures such as the hy-
pothalamus, are able to produce IL-6 and increase its plasma concen-
trations in response to stress [177]. Thus, interactions between IL-6 and
neuroendocrine responses are occurring in both directions. For instance,
it has been shown that the emotional state during a noxious stimulus
inuences IL-6 levels [178]. IL-6 is also involved in nociceptive path-
ways, and seems to play a role in hyperalgesia. Neurons of the dorsal
root ganglions express gp130 and exposure to IL-6 increases their
responsiveness to painful stimuli [179]. Electrophysiological experi-
ments in rats demonstrated that IL-6 is able to induce sensitization of C-
bers to mechanical stimulation [180]. Other experiments on rats also
deciphered the role of IL-6 in central nociception through spinal cord
neurons [181]. Thus, IL-6 is involved in both central (spinal and supra-
supraspinal) and peripherical levels of nociception.
For these reasons, many studies have investigated the effects of IL-6R
inhibitors on PROs. The RADIATE randomized controlled study was
designed to investigate the effects of tocilizumab versus placebo on
PROs in RA refractory to TNFi. Tocilizumab was clearly superior to
placebo already after 2 weeks on several PROs, notably on pain
(measured with a visual analogue scale), disability (assessed with the
health assessment questionary, HAQ) and fatigue (evaluated by the
Functional Assessment of Chronic Illness Therapy, FACIT, score) [182].
The adjunction of sarilumab to methotrexate also improved PROs in
patients with inadequate response to methotrexate [183] or TNFi [184].
In the head-to-head MONARCH trial, sarilumab monotherapy was
associated with a better improvement of PROs compared to adalimumab
monotherapy [57]. The superiority of tocilizumab was also demon-
strated for the composite PROs 7-domain score, RA Impact of Disease
(RAID score) [185]. Regarding psychosocial symptoms, the randomized
OPTION trial demonstrated a signicant improvement of the mental
component summary (MCS) score in the tocilizumab group compared to
placebo [186]. The superiority of tocilizumab compared to adalimumab
on same score was also observed in the ADACTA trial [54]. A cohort
study showed improvement of scores reecting depression (Hamilton
Depression Score) and anxiety (Hamilton Anxiety Score) after tocilizu-
mab introduction [187]. An observational study conrmed that tocili-
zumab use was associated with decreased depressive symptoms in RA
patients compared to other biological agents [188]. However, a recent
study, conducted in patients with planned allogenic hematopoietic stem
cell transplantation, showed that a single dose of tocilizumab was not
able to improve depressive symptoms. On contrary, it resulted in a sig-
nicant worsening of multiple PROs [189]. This illustrates our still
partial knowledge of the effects of IL-6 on the neuroendocrine system.
Nevertheless, the vast majority of studies showed that IL-6 blockade is
associated with improvement on pain, fatigue and mood disorders that
accompany RA.
3.4. Role of IL-6 on muscle and bone homeostasis
IL-6 has also been described as a myokine that participates in the
crosstalk between skeletal muscle and bone during exercise. IL-6 is
indeed released upon muscle contraction with a marked increase in
bloodstream during physical exercise [190]. It is hypothesized that the
rapid release of IL-6 acts in a paracrine, autocrine and endocrine way to
induce the metabolic adaptations in response to exercise (such as the
induction of lipolyze/glycogenolysis and catabolism, activation of HPA
axis, and bone turnover stimulation) [191]. Although IL-6 is known to be
associated with insulin resistance, a study demonstrated that acute IL-6
treatment enhances glucose uptake by skeletal muscles [192]. On bone,
IL-6 promotes osteocalcin bioactivation which further participates to the
physiological adaptations to exercise [193]. It has been suggested that
IL-6 release during exercise promotes also anti-inammatory effects
through induction of IL-10 and IL-1Ra [194]. However, IL-6 over-
production in inammatory diseases such as RA has detrimental effects
on both bone and muscle. Interestingly, transgenic mice overexpressing
IL-6 present an amyotrophic phenotype that is reversed by IL-6R
antibody treatment [195]. It is believed that age-related skeletal muscle
wasting is in part due to the increase of plasmatic IL-6 levels during
aging [196,197].
The effects of IL-6 on bone remodeling have been better studied.
Chronic overexpression of IL-6 in transgenic mice induces important
alterations in cortical and trabecular microarchitecture, and precludes
the normal development of bone in prepubertal mice [198]. The same
group showed that mice expressing high levels of IL-6 during the post-
birth period developed growth impairment with a reduced level of
insulin-like growth factor-I [199,200]. This nding enlightens mecha-
nism of stunted growth that can be seen in systemic juvenile rheumatoid
arthritis. Human trochanteric bone reverse transcription-polymerase
chain reaction (RT-PCR) analysis showed increased expression of IL-6
and RANK mRNA in patients with femoral neck fragility-fracture
compared to healthy bone obtained from cadavers [201]. IL-6 and its
soluble receptor are able to induce the expression of receptor activator
of nuclear factor-κB ligand (RANKL) by broblast-like synoviocytes,
leading to osteoclast activation and bone resorption [26]. IL-6R trans-
signaling also appears to be involved in postmenopausal osteoporosis
[202].
Musculoskeletal manifestations of RA are not limited to the joints.
The disease commonly affects muscle and bone compartments, notably
osteoporosis and sarcopenia [203,204]. Patients with RA have a 2- fold
increased risk of osteoporotic fractures compared to a matched control-
population [205]. The origin is multifactorial, including disease dura-
tion and activity, and the use of some treatments such as glucocorticoids,
opioids and selective-serotonin reuptake-inhibitors [206,207]. Despite
marked improvement in RA management, the risk of vertebral fracture
remains high in RA [208]. Osteoporosis is indeed still frequent, with a
prevalence around 30% in a RA cohort [209]. However, analysis from
the German National Database conrmed a decrease of osteoporosis
prevalence in RA compared to the previous decade, indicating a bene-
cial effect of the use of more recent RA treatments [210]. Regarding
muscle mass, a recent meta-analysis reported a 31% prevalence of sar-
copenia in RA (i.e. 3 times higher than in the general population) [211].
In this study, disease activity was a predictive factor for sarcopenia, as
well as the use of glucocorticoids [212]. In addition, sarcopenia is also
associated to vertebral fractures in RA, having a synergistic effect with
osteoporosis [213].
To date, there is no evidence that IL-6 blockade protects against
fragility fractures. A recent cohort study did not nd any difference in
the risk of fractures between the different biologics used in RA [214].
However, in the MONARCH trial, procollagen type 1 N-terminal pro-
peptide (P1NP), a marker of bone formation, increased more in the
sarilumab group compared to adalimumab. Similarly, patients treated
with sarilumab had a greater reduction of RANKL compared to those
treated with adalimumab [65]. In a prospective study, tocilizumab was
associated with a decrease of C-terminal cross-linking telopeptide of
type I collagen (CTX), another biomarker of bone resorption, and with
increased bone mineral density after 2 years in ACPA-positive RA pa-
tients [215]. Less is known about the effect of IL-6 blockade on skeletal
muscle. The use of tocilizumab in RA is associated with weight gain
[216]. A recent study showed that this observation was partly related to
muscle gain (with a reduction of sarcopenia prevalence) [217]. Overall,
there are indirect evidences that IL-6 blockade is associated with
improved muscle and bone function in RA.
4. Systemic adverse effects of IL-6 blockade in rheumatoid
arthritis
Considering IL-6 broad spectrum of activities, IL-6 blockade is
obviously associated with a variety of adverse effects. As for other bio-
logic (bDMARDs) or conventional synthetic (cs)DMARDs, the major
safety concern of IL-6 inhibitors is the occurrence of serious infections
due to their immunosuppressive effects [218,219]. The role of IL-6 in
host defense against infections has been reviewed elsewhere [220]. In
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
6
addition, RA confers an increased risk of serious infections, which is
associated with higher mortality rate compared to the general popula-
tion [221,16,17]. This increased risk appears to be related to disease
activity [222]. Biological DMARDs are also associated with an increased
risk compared to csDMARDs [223].
A large US multi-database cohort showed no difference in the risk of
serious infections between RA patients treated with tocilizumab
compared to other biologics or tofacitinib, a Janus kinase inhibitor. But,
when used as a rst line therapy, tocilizumab was associated with an
increased risk for serious bacterial infections, including skin and soft
tissue infections as well as diverticulitis compared to TNFi and abatacept
[224]. A British prospective observational study also demonstrated an
increased risk of tocilizumab compared to etanercept (hazard ratio (HR)
of 1.22, with a 95% condence interval (CI) between 1.02 and 1.47)
[225]. The better safety prole of TNFi compared to tocilizumab was not
conrmed in a Japanese cohort study adjusting for covariates [226].
Sarilumab had the same safety prole as tocilizumab regardless of the
route of administration [227,228]. After a follow-up of 5 years, safety
prole of sarilumab in RA refractory to TNFi remained unchanged. The
most frequent adverse events were neutropenia with an incidence of
15.3 cases per 100 patients-years (PY) [229]. The incidence of serious
infection was 3.9 per 100 PY, comparable to other bDMARDs. In a cu-
mulative safety analysis, most frequent serious infections were pneu-
monia (with an incidence rate of 1 per 100 PY), followed by
gastroenteritis and urinary tract infection [230]. In this study, tocilizu-
mab was also associated with a slight increased risk of opportunistic
infections (0.23 per 100 PY), with no case reported in the control group.
These infections included mycobacterial (with 8 cases of Mycobacterium
Tuberculosis), and fungal (Candidiasis, Pneumocystis Jiroveci, and Cryp-
tococcus) infections.
Hepatic and gastrointestinal adverse events have been associated
with IL-6 blockade. Indeed, IL-6 pathways are involved in liver ho-
meostasis and regeneration [231,232]. Mice decient for IL-6 are more
prone to develop alcoholic and non-alcoholic fatty liver disease [233].
Thus, IL-6 inhibitors have been associated with signicant transaminase
elevations (with an estimated incidence of 1.3/100 PY), particularly
when associated to other hepatotoxic drugs [230,234]. Results from an
observational study showed that the increase of transaminase levels is
more frequent among patients treated with tocilizumab than with TNFi
[235]. However, these hepatic perturbations are reversible, and do not
appear to be associated with long-term hepatic lesions [236,237]. In
particular, no increase in malignant hepatic neoplasm was observed in
patients treated with tocilizumab compared to other biologics [238].
Some case reports and post-approval studies identied also pancreatitis
as a potential adverse effect of IL-6 blockade, possibly due to increased
levels of triglycerides [239]. More importantly, RA patients receiving
tocilizumab have a 2- fold higher risk of lower intestinal perforation
compared to those receiving a TNFi [240]. An increased risk of intestinal
perforation was conrmed with an adjusted HR of 4.5 (95% CI
2.019.99) [241]. One explanation could be the protective effect of IL-6
on intestinal mucosa. Indeed, experimental data have shown that IL-6
inhibits enterocytes cell death, and is involved in epithelial regenera-
tion [242,243]. The fact that IL-6 blockade precludes the interpretation
of acute phase biomarkers is also a potential factor that could delay
recognition of serious infections or diverticulitis. Table 1 summarizes
some of the main systemic side effects of IL-6 blockade. In general, real
world registries tend to show that IL-6R inhibitors induce a rapid and
long-term improvement of RA with a good safety prole
[235,244,245,142]. A recent meta-analysis including 88 randomized
controlled trials found that tocilizumab was associated with a better
Table 1
This table summarizes the effects of IL-6 on most RA comorbidities. The subsequent systemic effects associated to its blockade, whether benecial or detrimental, are
also presented. Abbreviation: ANC (absolute neutrophil count), bDMARD (Biologic Disease-modifying Antirheumatic Drug), CRP (C-reactive protein), ESR (eryth-
rocyte sedimentation rate), GI (gastro-intestinal), LDL (low-density lipoprotein), TB (Tuberculosis).
RA comorbidities IL-6 effects Consequences of IL-6 blockade Management of IL-6 blockade sides effects
Infections Key regulator cytokine of both innate and
adaptative immunity. Participates to the
defense against bacterial and fungal pathogen
[220]
Benecial: Good safety prole compared to other
bDMARD.
Long-term use is associated with a decreased
incidence of serious infections [142].
Detrimental: Suppression of fever and CRP [66,67].
Increase risk of serious bacterial infections, skin and
soft tissue infections, and diverticulitis. Rare
opportunistic infections (TB, fungi) [229,230].
Caution with CRP and ESR interpretation.
Treatment interruption during active infection
[252]. Perioperative interruption (>3 weeks
before surgery) [253]. Avoid glucocorticoid co-
medication.
TB screening prior treatment initiation. Avoid IL-6
blockade if latent TB [254].
Cardiovascular
diseases
Involved in endothelial activation and
homeostasis; acceleration of atheroma [119-
124].
Participates to lipid and glucide metabolism
[125-134].
Benecial: Good safety prole [140]. Reduction of
glycosylated hemoglobin (HbA1c) [138,139].
Improvement of endothelial and cardiac function
[146,147].
Detrimental: Increase of total cholesterol, LDL-
cholesterol and triglyceride levels
Assessment of lipid prole at therapy initiation,
and regular monitoring thereafter. Consider
posology reduction [252], or concomitant use of
statin [255].
Hematological Stimulates hepatocytes to produce proteins
involved in iron metabolism [74,75].
Stimulates thrombopoietin expression
[88,89].
Interacts with neutrophil trafcking [97].
Benecial: Reduction of hepcidin levels, leading to
improvement of anemia of chronic disease
[64,84,85].
Detrimental: Neutropenia (most often transient)
[95].
Thrombocytopenia [54].
Neutrophils and thrombocytes monitoring:
treatment interruption if ANC 0.51 ×10
9
/l.
Discontinuation if ANC <0.5 ×10
9
/l. Avoid
initiation in patients with ANC <2 ×10
9
/l [98]
Hepato-
gastrointestinal
Involved in liver homeostasis and regeneration
[231-234].
Protective effects on intestinal mucosa
[242,243].
Detrimental: Elevated transaminases (reversible,
and most often moderate) [235-237].
Increase risk of GI perforation [241].
Avoid concomitant use of other hepatotoxic
drugs. Regular monitoring with dose adjustment if
persistent transaminases elevation [237].
Caution in patients at risk for GI perforation (older
age, comorbidities such as diabetes and
diverticulitis, use of high dose glucocorticoids)
[240].
Fatigue, pain and
mood disorders
Activation and alteration of the
hypothalamicpituitary-adrenal (HPA) axis
[172-177].
Central and peripherical pain control [178-
181].
Benecial: Improvement of patient reported
outcome [182-187]. Decreased depressive
symptoms in RA patients compared to other
biological agents [188].
Osteoporosis and
sarcopenia
Osteoclast activation and bone resorption
[26]. Chronic elevation of IL-6 promotes
muscle wasting [195-197].
Benecial: Improvement of biomarkers of bone
resorption [214,215]. Potential muscle gain
[216,217].
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
7
improvement of disease activity with same safety prole compared to
other bDMARDs and Janus kinase inhibitors in RA patients with inad-
equate response to at least one DMARD [246].
5. Conclusions
Originally named B-cell stimulatory factor-2 but also hepatocyte-
stimulary factor or interferon-β2, IL-6 is characterized by a wide spectrum
of action mediated by its two signaling pathways. Having both pro- and
anti-inammatory properties, IL-6 is a key regulator of inammation
and immunity. Its involvement in the pathogenesis of RA comorbidities
illustrates its pleiotropic effects. IL-6 is indeed associated with multiple
biological activities distant from the site of inammation. Likewise, IL-6
blockade has consequences far beyond the joints. The recent data on RA
suggest benecial effects of IL-6R inhibitors on many comorbidities.
Overall, IL-6 inhibition is associated with improvement of several sys-
temic manifestations, including anemia of chronic disease, CV events,
bone and muscle functions, and neuro-psychological manifestations as
assessed by PROs (mainly fatigue, mood disorders and pain). Impor-
tantly some of these systemic effects are not necessarily dependent on
the clinical efcacy of IL-6 inhibition on joint inammation.
Furthermore, IL-6R inhibitors and new antibodies directly targeting
IL-6 have increasing therapeutic applications beyond inammatory
rheumatic diseases [247]. For instance, sirukumab, an IL-6 neutralizing
antibody, is tested in patients with major depressive disorder in a phase
II study. Recently, a prospective study showed improvement in peri-
odontal status in patients with RA-associated periodontal disease [248].
Specic trans-signaling inhibition showed promising effect of olamkicept
in patients with active inammatory bowel disease in a phase 2 study
[249]. Finally, as IL-6R inhibition was already approved for treatment of
cytokine release syndrome [250], several retrospective studies and
randomized trials were conducted to test IL-6 inhibition in severe acute
respiratory syndrome associated to Covid-19 [251].
IL-6 biology, notably trans-signaling pathway and its systemic effects,
remains only partially elucidated yet. The therapeutic potential of IL-6
blockade goes far beyond joints, and is probably still in early stages.
Declaration of Competing Interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
References
[1] C. Gabay, I. Kushner, Acute-phase proteins and other systemic responses to
inammation, N. Engl. J. Med. 340 (6) (1999) 448454, https://doi.org/
10.1056/NEJM199902113400607.
[2] C. Gabay, Interleukin-6 and chronic inammation, Arthritis Res. Ther. 8 (Suppl 2)
(2006) S3, https://doi.org/10.1186/ar1917.
[3] T. Kishimoto, T. Hirano, T. Kuritani, Y. Yamamura, P. Ralph, R.A. Good,
Induction of IgG production in human B lymphoblastoid cell lines with normal
human T cells, Nature 271 (5647) (1978) 756758, https://doi.org/10.1038/
271756a0.
[4] T. Hirano, et al., Complementary DNA for a novel human interleukin (BSF-2) that
induces B lymphocytes to produce immunoglobulin, Nature 324 (6092) (1986)
7376, https://doi.org/10.1038/324073a0.
[5] L.H. Calabrese, S. Rose-John, IL-6 biology: implications for clinical targeting in
rheumatic disease, Nat. Rev. Rheumatol. 10 (12) (2014) 720727, https://doi.
org/10.1038/nrrheum.2014.127.
[6] S.-Y. Hwang, et al., IL-17 induces production of IL-6 and IL-8 in rheumatoid
arthritis synovial broblasts via NF-kappaB- and PI3-kinase/Akt-dependent
pathways, Arthritis Res. Ther. 6 (2) (2004) R120R128, https://doi.org/
10.1186/ar1038.
[7] M. Narazaki, T. Kishimoto, The two-faced cytokine IL-6 in host defense and
diseases, Int. J. Mol. Sci. 19 (11) (2018) 3528, https://doi.org/10.3390/
ijms19113528.
[8] C. Garbers, S. Aparicio-Siegmund, S. Rose-John, The IL-6/gp130/STAT3 signaling
axis: recent advances towards specic inhibition, Curr. Opin. Immunol. 34 (2015)
7582, https://doi.org/10.1016/j.coi.2015.02.008.
[9] S. Rose-John, Interleukin-6 biology is coordinated by membrane bound and
soluble receptors, Acta Biochim. Pol. 50 (3) (2003) 603611, doi: 035003603.
[10] J. Wolf, S. Rose-John, C. Garbers, Interleukin-6 and its receptors: a highly
regulated and dynamic system, Cytokine 70 (1) (2014) 1120, https://doi.org/
10.1016/j.cyto.2014.05.024.
[11] S. Kang, T. Tanaka, M. Narazaki, T. Kishimoto, Targeting interleukin-6 signaling
in clinic, Immunity 50 (4) (2019) 10071023, https://doi.org/10.1016/j.
immuni.2019.03.026.
[12] P.C. Heinrich, I. Behrmann, S. Haan, H.M. Hermanns, G. Müller-Newen,
F. Schaper, Principles of interleukin (IL)-6-type cytokine signalling and its
regulation, Biochem. J. 374 (Pt 1) (2003) 120, https://doi.org/10.1042/
BJ20030407.
[13] E.H. Choy, F. De Benedetti, T. Takeuchi, M. Hashizume, M.R. John, T. Kishimoto,
Translating IL-6 biology into effective treatments, Nat. Rev. Rheumatol. 16 (6)
(2020) 335345, https://doi.org/10.1038/s41584-020-0419-z.
[14] M. Noack, P. Miossec, Selected cytokine pathways in rheumatoid arthritis, Semin.
Immunopathol. 39 (4) (2017) 365383, https://doi.org/10.1007/s00281-017-
0619-z.
[15] Georg Schett, Iain B. McInnes, Markus F. Neurath, Reframing Immune-Mediated
Inammatory Diseases through Signature Cytokine Hubs, N. Engl. J. Med. (2021),
https://doi.org/10.1056/NEJMra1909094.
[16] M. Dougados, et al., Prevalence of comorbidities in rheumatoid arthritis and
evaluation of their monitoring: results of an international, cross-sectional study
(COMORA), Ann. Rheum. Dis. 73 (1) (2014) 6268, https://doi.org/10.1136/
annrheumdis-2013-204223.
[17] F. Wolfe, et al., The mortality of rheumatoid arthritis, Arthritis Rheum. 37 (4)
(1994) 481494, https://doi.org/10.1002/art.1780370408.
[18] J. Gauldie, C. Richards, H. Baumann, IL6 and the acute phase reaction, Res.
Immunol. 143 (7) (1992) 755759, https://doi.org/10.1016/0923-2494(92)
80018-g.
[19] I. Kushner, Regulation of the acute phase response by cytokines, Perspect. Biol.
Med. 36 (4) (1993) 611622, https://doi.org/10.1353/pbm.1993.0004.
[20] P.C. Heinrich, J.V. Castell, T. Andus, Interleukin-6 and the acute phase response,
Biochem. J. 265 (3) (1990) 621636, https://doi.org/10.1042/bj2650621.
[21] J. Gauldie, C. Richards, D. Harnish, P. Lansdorp, H. Baumann, Interferon beta 2/
B-cell stimulatory factor type 2 shares identity with monocyte-derived
hepatocyte-stimulating factor and regulates the major acute phase protein
response in liver cells, Proc. Natl. Acad. Sci. U. S. A. 84 (20) (1987) 72517255,
https://doi.org/10.1073/pnas.84.20.7251.
[22] S. Black, I. Kushner, D. Samols, C-reactive protein, J. Biol. Chem. 279 (47) (2004)
4848748490, https://doi.org/10.1074/jbc.R400025200.
[23] M. Kopf, et al., Impaired immune and acute-phase responses in interleukin-6-
decient mice, Nature 368 (6469) (1994) 339342, https://doi.org/10.1038/
368339a0.
[24] S.M. Hurst, et al., Il-6 and its soluble receptor orchestrate a temporal switch in the
pattern of leukocyte recruitment seen during acute inammation, Immunity 14
(6) (2001) 705714, https://doi.org/10.1016/s1074-7613(01)00151-0.
[25] P. Baran, R. Nitz, J. Gr¨
otzinger, J. Scheller, C. Garbers, Minimal interleukin 6 (IL-
6) receptor stalk composition for IL-6 receptor shedding and IL-6 classic signaling,
J. Biol. Chem. 288 (21) (2013) 1475614768, https://doi.org/10.1074/jbc.
M113.466169.
[26] M. Hashizume, N. Hayakawa, M. Mihara, IL-6 trans-signalling directly induces
RANKL on broblast-like synovial cells and is involved in RANKL induction by
TNF-alpha and IL-17, Rheumatol. Oxf. Engl. 47 (11) (2008) 16351640, https://
doi.org/10.1093/rheumatology/ken363.
[27] V. Marin, et al., The IL-6-soluble IL-6Ralpha autocrine loop of endothelial
activation as an intermediate between acute and chronic inammation: an
experimental model involving thrombin, J. Immunol. Baltim. Md 1950 167 (6)
(2001) 34353442, https://doi.org/10.4049/jimmunol.167.6.3435.
[28] P. Chomarat, J. Banchereau, J. Davoust, A.K. Palucka, IL-6 switches the
differentiation of monocytes from dendritic cells to macrophages, Nat. Immunol.
1 (6) (2000) 510514, https://doi.org/10.1038/82763.
[29] X.-L. Fu, et al., Interleukin 6 induces M2 macrophage differentiation by STAT3
activation that correlates with gastric cancer progression, Cancer Immunol.
Immunother. CII 66 (12) (2017) 15971608, https://doi.org/10.1007/s00262-
017-2052-5.
[30] H. Tilg, E. Trehu, M.B. Atkins, C.A. Dinarello, J.W. Mier, Interleukin-6 (IL-6) as an
anti-inammatory cytokine: induction of circulating IL-1 receptor antagonist and
soluble tumor necrosis factor receptor p55, Blood 83 (1) (1994) 113118.
[31] M. Ciofani, et al., A validated regulatory network for Th17 cell specication, Cell
151 (2) (2012) 289303, https://doi.org/10.1016/j.cell.2012.09.016.
[32] A. Kimura, T. Kishimoto, IL-6: regulator of Treg/Th17 balance, Eur. J. Immunol.
40 (7) (2010) 18301835, https://doi.org/10.1002/eji.201040391.
[33] J.P. B¨
ottcher, et al., IL-6 trans-signaling-dependent rapid development of
cytotoxic CD8+T cell function, Cell Rep. 8 (5) (2014) 13181327, https://doi.
org/10.1016/j.celrep.2014.07.008.
[34] G. Jego, R. Bataille, C. Pellat-Deceunynck, Interleukin-6 is a growth factor for
nonmalignant human plasmablasts, Blood 97 (6) (2001) 18171822, https://doi.
org/10.1182/blood.v97.6.1817.
[35] A. Muraguchi, et al., The essential role of B cell stimulatory factor 2 (BSF-2/IL-6)
for the terminal differentiation of B cells, J. Exp. Med. 167 (2) (1988) 332344,
https://doi.org/10.1084/jem.167.2.332.
[36] S. Suematsu, et al., IgG1 plasmacytosis in interleukin 6 transgenic mice, Proc.
Natl. Acad. Sci. U. S. A. 86 (19) (1989) 75477551, https://doi.org/10.1073/
pnas.86.19.7547.
[37] S.A. Diehl, H. Schmidlin, M. Nagasawa, B. Blom, H. Spits, IL-6 triggers IL-21
production by human CD4+T cells to drive STAT3-dependent plasma cell
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
8
differentiation in B cells, Immunol. Cell Biol. 90 (8) (2012) 802811, https://doi.
org/10.1038/icb.2012.17.
[38] L. Zhou, et al., IL-6 programs T(H)-17 cell differentiation by promoting sequential
engagement of the IL-21 and IL-23 pathways, Nat. Immunol. 8 (9) (2007)
967974, https://doi.org/10.1038/ni1488.
[39] N. Schmitt, et al., Human dendritic cells induce the differentiation of interleukin-
21-producing T follicular helper-like cells through interleukin-12, Immunity 31
(1) (2009) 158169, https://doi.org/10.1016/j.immuni.2009.04.016.
[40] G.M. Jogdand, S. Mohanty, S. Devadas, Regulators of Tfh cell differentiation,
Front. Immunol. 7 (2016) 520, https://doi.org/10.3389/mmu.2016.00520.
[41] O. Dienz, et al., The induction of antibody production by IL-6 is indirectly
mediated by IL-21 produced by CD4+T cells, J. Exp. Med. 206 (1) (2009) 6978,
https://doi.org/10.1084/jem.20081571.
[42] D.G. Hill, et al., Hyperactive gp130/STAT3-driven gastric tumourigenesis
promotes submucosal tertiary lymphoid structure development, Int. J. Cancer
143 (1) (2018) 167178, https://doi.org/10.1002/ijc.31298.
[43] E. Corsiero, A. Nerviani, M. Bombardieri, C. Pitzalis, Ectopic lymphoid structures:
powerhouse of autoimmunity, Front. Immunol. 7 (2016) 430, https://doi.org/
10.3389/mmu.2016.00430.
[44] L. Pinede, P. Duhaut, R. Loire, Clinical presentation of left atrial cardiac myxoma.
A series of 112 consecutive cases, Medicine (Baltimore) 80 (3) (2001) 159172,
https://doi.org/10.1097/00005792-200105000-00002.
[45] U. Sack, R.W. Kinne, T. Marx, P. Heppt, S. Bender, F. Emmrich, Interleukin-6 in
synovial uid is closely associated with chronic synovitis in rheumatoid arthritis,
Rheumatol. Int. 13 (2) (1993) 4551, https://doi.org/10.1007/BF00307733.
[46] A. Dispenzieri, D.C. Fajgenbaum, Overview of Castleman disease, Blood 135 (16)
(2020) 13531364, https://doi.org/10.1182/blood.2019000931.
[47] S. Gerlo, G. Haegeman, W. Vanden Berghe, Transcriptional regulation of
autocrine IL-6 expression in multiple myeloma cells, Cell. Signal. 20 (8) (2008)
14891496, https://doi.org/10.1016/j.cellsig.2008.04.004.
[48] Z.Y. Lu, H. Brailly, J. Wijdenes, R. Bataille, J.F. Rossi, B. Klein, Measurement of
whole body interleukin-6 (IL-6) production: prediction of the efcacy of anti-IL-6
treatments, Blood 86 (8) (1995) 31233131.
[49] N. Nishimoto, et al., Humanized anti-interleukin-6 receptor antibody treatment of
multicentric Castleman disease, Blood 106 (8) (2005) 26272632, https://doi.
org/10.1182/blood-2004-12-4602.
[50] N. Nishimoto, et al., Treatment of rheumatoid arthritis with humanized anti-
interleukin-6 receptor antibody: a multicenter, double-blind, placebo-controlled
trial, Arthritis Rheum. 50 (6) (2004) 17611769, https://doi.org/10.1002/
art.20303.
[51] R.N. Maini, et al., Double-blind randomized controlled clinical trial of the
interleukin-6 receptor antagonist, tocilizumab, in European patients with
rheumatoid arthritis who had an incomplete response to methotrexate, Arthritis
Rheum. 54 (9) (2006) 28172829, https://doi.org/10.1002/art.22033.
[52] N. Nishimoto, et al., Study of active controlled monotherapy used for rheumatoid
arthritis, an IL-6 inhibitor (SAMURAI): evidence of clinical and radiographic
benet from an x ray reader-blinded randomised controlled trial of tocilizumab,
Ann. Rheum. Dis. 66 (9) (2007) 11621167, https://doi.org/10.1136/
ard.2006.068064.
[53] M.C. Genovese, et al., Sarilumab plus methotrexate in patients with active
rheumatoid arthritis and inadequate response to methotrexate: results of a phase
III study, Arthritis Rheumatol. Hoboken NJ 67 (6) (2015) 14241437, https://
doi.org/10.1002/art.39093.
[54] C. Gabay, et al., Tocilizumab monotherapy versus adalimumab monotherapy for
treatment of rheumatoid arthritis (ADACTA): a randomised, double-blind,
controlled phase 4 trial, Lancet Lond. Engl. 381 (9877) (2013) 15411550,
https://doi.org/10.1016/S0140-6736(13)60250-0.
[55] S. Finzel, et al., Comparison of the effects of tocilizumab monotherapy and
adalimumab in combination with methotrexate on bone erosion repair in
rheumatoid arthritis, Ann. Rheum. Dis. 78 (9) (2019) 11861191, https://doi.
org/10.1136/annrheumdis-2018-214894.
[56] G.R. Burmester, et al., Efcacy and safety of sarilumab monotherapy versus
adalimumab monotherapy for the treatment of patients with active rheumatoid
arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial,
Ann. Rheum. Dis. 76 (5) (2017) 840847, https://doi.org/10.1136/annrheumdis-
2016-210310.
[57] V. Strand, et al., Patient-reported outcomes from a randomized phase III trial of
sarilumab monotherapy versus adalimumab monotherapy in patients with
rheumatoid arthritis, Arthritis Res. Ther. 20 (1) (2018) 129, https://doi.org/
10.1186/s13075-018-1614-z.
[58] S. Yokota, et al., Efcacy and safety of tocilizumab in patients with systemic-onset
juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled,
withdrawal phase III trial, Lancet Lond. Engl. 371 (9617) (2008) 9981006,
https://doi.org/10.1016/S0140-6736(08)60454-7.
[59] Y. Kaneko, et al., Tocilizumab in patients with adult-onset stills disease
refractory to glucocorticoid treatment: a randomised, double-blind, placebo-
controlled phase III trial, Ann. Rheum. Dis. 77 (12) (2018) 17201729, https://
doi.org/10.1136/annrheumdis-2018-213920.
[60] J.H. Stone, et al., Trial of Tocilizumab in giant-cell arteritis, N. Engl. J. Med. 377
(4) (2017) 317328, https://doi.org/10.1056/NEJMoa1613849.
[61] M.C. Genovese, et al., Interleukin-6 receptor inhibition with tocilizumab reduces
disease activity in rheumatoid arthritis with inadequate response to disease-
modifying antirheumatic drugs: the tocilizumab in combination with traditional
disease-modifying antirheumatic drug therapy study, Arthritis Rheum. 58 (10)
(2008) 29682980, https://doi.org/10.1002/art.23940.
[62] R. Fleischmann, et al., Sarilumab and nonbiologic disease-modifying
antirheumatic drugs in patients with active rheumatoid arthritis and inadequate
response or intolerance to tumor necrosis factor inhibitors: SARILUMAB PLUS
csDMARDs IN RA PATIENTS WHO WERE TREATED UNSUCCESSFULLY WITH
ANTI-TNF, Arthritis Rheumatol. 69 (2) (2017) 277290, https://doi.org/
10.1002/art.39944.
[63] P. Juhl, C.S. Thudium, N.S. Gudmann, M.A. Karsdal, A.-C. Bay-Jensen, A.
S. Siebuhr, IL-6 receptor inhibition modulates type III collagen and C-reactive
protein degradation in rheumatoid arthritis patients with an inadequate response
to anti-tumour necrosis factor therapy: analysis of connective tissue turnover in
the tocilizumab RADIATE study, Clin. Exp. Rheumatol. 36 (4) (2018) 568574.
[64] A. Boyapati, J. Msihid, S. Fiore, J. van Adelsberg, N.M.H. Graham, J.D. Hamilton,
Sarilumab plus methotrexate suppresses circulating biomarkers of bone
resorption and synovial damage in patients with rheumatoid arthritis and
inadequate response to methotrexate: a biomarker study of MOBILITY, Arthritis
Res. Ther. 18 (1) (2016) 225, https://doi.org/10.1186/s13075-016-1132-9.
[65] C. Gabay, et al., Sarilumab and adalimumab differential effects on bone
remodelling and cardiovascular risk biomarkers, and predictions of treatment
outcomes, Arthritis Res. Ther. 22 (1) (2020) 70, https://doi.org/10.1186/
s13075-020-02163-6.
[66] M. Hirao, et al., Laboratory and febrile features after joint surgery in patients with
rheumatoid arthritis treated with tocilizumab, Ann. Rheum. Dis. 68 (5) (2009)
654657, https://doi.org/10.1136/ard.2008.090068.
[67] M. Berman, et al., The effect of tocilizumab on inammatory markers in patients
hospitalized with serious infections. Case series and review of literature, Life
Basel Switz. 11 (3) (2021), https://doi.org/10.3390/life11030258.
[68] J.E. Trey, I. Kushner, The acute phase response and the hematopoietic system: the
role of cytokines, Crit. Rev. Oncol. Hematol. 21 (13) (1995) 118, https://doi.
org/10.1016/1040-8428(94)00141-3.
[69] E. Nikiphorou, et al., Haematological abnormalities in new-onset rheumatoid
arthritis and risk of common infections: a population-based study, Rheumatol.
Oxf. Engl. 59 (5) (2020) 9971005, https://doi.org/10.1093/rheumatology/
kez344.
[70] A. Wilson, H.-T. Yu, L.T. Goodnough, A.R. Nissenson, Prevalence and outcomes of
anemia in rheumatoid arthritis: a systematic review of the literature, Am. J. Med.
116 (Suppl 7A) (2004) 50S57S, https://doi.org/10.1016/j.
amjmed.2003.12.012.
[71] B. M¨
oller, A. Scherer, F. F¨
orger, P.M. Villiger, A. Finckh, Swiss Clinical Quality
Management Program for Rheumatic Diseases, Anaemia may add information to
standardised disease activity assessment to predict radiographic damage in
rheumatoid arthritis: a prospective cohort study, Ann. Rheum. Dis. 73 (4) (2014)
691696, doi: 10.1136/annrheumdis-2012-202709.
[72] C. Han, et al., Association of anemia and physical disability among patients with
rheumatoid arthritis, J. Rheumatol. 34 (11) (2007) 21772182.
[73] H.R. Peeters, et al., Course and characteristics of anaemia in patients with
rheumatoid arthritis of recent onset, Ann. Rheum. Dis. 55 (3) (1996) 162168,
https://doi.org/10.1136/ard.55.3.162.
[74] E. Nemeth, et al., IL-6 mediates hypoferremia of inammation by inducing the
synthesis of the iron regulatory hormone hepcidin, J. Clin. Invest. 113 (9) (2004)
12711276, https://doi.org/10.1172/JCI20945.
[75] E.T. Ali, A.S. Jabbar, A.N. Mohammed, A comparative study of interleukin 6,
inammatory markers, ferritin, and hematological prole in rheumatoid arthritis
patients with anemia of chronic disease and iron deciency anemia, Anemia 2019
(2019) 3457347, https://doi.org/10.1155/2019/3457347.
[76] N.C. Andrews, Anemia of inammation: the cytokine-hepcidin link, J. Clin.
Invest. 113 (9) (2004) 12511253, https://doi.org/10.1172/JCI21441.
[77] E. Nemeth, et al., Hepcidin regulates cellular iron efux by binding to ferroportin
and inducing its internalization, Science 306 (5704) (2004) 20902093, https://
doi.org/10.1126/science.1104742.
[78] M.A. Abdel-Khalek, A.M. El-Barbary, S.-A.-M. Essa, A.S. Ghobashi, Serum
hepcidin: a direct link between anemia of inammation and coronary artery
atherosclerosis in patients with rheumatoid arthritis, J. Rheumatol. 38 (10)
(2011) 21532159, https://doi.org/10.3899/jrheum.110339.
[79] H. Sato, et al., Serum hepcidin level, iron metabolism and osteoporosis in patients
with rheumatoid arthritis, Sci. Rep. 10 (1) (2020) 9882, https://doi.org/
10.1038/s41598-020-66945-3.
[80] C. Masson, Rheumatoid anemia, Joint Bone Spine 78 (2) (2011) 131137,
https://doi.org/10.1016/j.jbspin.2010.05.017.
[81] K. Michels, E. Nemeth, T. Ganz, B. Mehrad, Hepcidin and host defense against
infectious diseases, PLoS Pathog. 11 (8) (2015), e1004998, https://doi.org/
10.1371/journal.ppat.1004998.
[82] H.C. Sox, Diagnostic decision: the erythrocyte sedimentation rate: guidelines for
rational use, Ann. Intern. Med. 104 (4) (1986) 515, https://doi.org/10.7326/
0003-4819-104-4-515.
[83] S.-N.-J. Song, N. Tomosugi, H. Kawabata, T. Ishikawa, T. Nishikawa, K. Yoshizaki,
Down-regulation of hepcidin resulting from long-term treatment with an anti-IL-6
receptor antibody (tocilizumab) improves anemia of inammation in multicentric
Castleman disease, Blood 116 (18) (2010) 36273634, https://doi.org/10.1182/
blood-2010-03-271791.
[84] J.D. Isaacs, O. Harari, U. Kobold, J.S. Lee, C. Bernasconi, Effect of tocilizumab on
haematological markers implicates interleukin-6 signalling in the anaemia of
rheumatoid arthritis, Arthritis Res. Ther. 15 (6) (2013) R204, https://doi.org/
10.1186/ar4397.
[85] S.-N.-J. Song, et al., Comparative evaluation of the effects of treatment with
tocilizumab and TNF-
α
inhibitors on serum hepcidin, anemia response and
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
9
disease activity in rheumatoid arthritis patients, Arthritis Res. Ther. 15 (5) (2013)
R141, https://doi.org/10.1186/ar4323.
[86] M. Hashimoto, et al., Increase of hemoglobin levels by anti-IL-6 receptor antibody
(tocilizumab) in rheumatoid arthritis, PloS One 9 (5) (2014), e98202, https://doi.
org/10.1371/journal.pone.0098202.
[87] A. Margraf, A. Zarbock, Platelets in inammation and resolution, J. Immunol.
Baltim. Md 1950 203 (9) (2019) 23572367, https://doi.org/10.4049/
jimmunol.1900899.
[88] C.W. Hollen, J. Henthorn, J.A. Koziol, S.A. Burstein, Serum interleukin-6 levels in
patients with thrombocytosis, Leuk. Lymphoma 8 (3) (1992) 235241, https://
doi.org/10.3109/10428199209054910.
[89] A. Kaser, et al., Interleukin-6 stimulates thrombopoiesis through thrombopoietin:
role in inammatory thrombocytosis, Blood 98 (9) (2001) 27202725, https://
doi.org/10.1182/blood.v98.9.2720.
[90] T. Ishibashi, H. Kimura, T. Uchida, S. Kariyone, P. Friese, S.A. Burstein, Human
interleukin 6 is a direct promoter of maturation of megakaryocytes in vitro, Proc.
Natl. Acad. Sci. U. S. A. 86 (15) (1989) 59535957, https://doi.org/10.1073/
pnas.86.15.5953.
[91] L. Zhang, et al., Thrombocytosis as a response to high interleukin-6 levels in
cGMP-dependent protein kinase I mutant mice, Arterioscler. Thromb. Vasc. Biol.
33 (8) (2013) 18201828, https://doi.org/10.1161/ATVBAHA.113.301507.
[92] I. Ertenli, I.C. Haznedaro˘
glu, S. Kiraz, I. Celik, M. Calgüneri, S. Kirazhi, Cytokines
affecting megakaryocytopoiesis in rheumatoid arthritis with thrombocytosis,
Rheumatol. Int. 16 (1) (1996) 58, https://doi.org/10.1007/BF01419947.
[93] I. Ertenli, S. Kiraz, M.A. Oztürk, I.c. Haznedaro˘
glu, I. Celik, M. Calgüneri,
Pathologic thrombopoiesis of rheumatoid arthritis, Rheumatol. Int. 23 (2) (2003)
4960, https://doi.org/10.1007/s00296-003-0289-0.
[94] J. Nakagawa, et al., A novel scoring system based on common laboratory tests
predicts the efcacy of TNF-inhibitor and IL-6 targeted therapy in patients with
rheumatoid arthritis: a retrospective, multicenter observational study, Arthritis
Res. Ther. 19 (1) (2017) 185, https://doi.org/10.1186/s13075-017-1387-9.
[95] I. Nakamura, Y. Omata, M. Naito, K. Ito, Blockade of interleukin 6 signaling
induces marked neutropenia in patients with rheumatoid arthritis, J. Rheumatol.
36 (2) (2009) 459460, https://doi.org/10.3899/jrheum.080930.
[96] H.L. Wright, A.L. Cross, S.W. Edwards, R.J. Moots, Effects of IL-6 and IL-6
blockade on neutrophil function in vitro and in vivo, Rheumatol. Oxf. Engl. 53 (7)
(2014) 13211331, https://doi.org/10.1093/rheumatology/keu035.
[97] L.S.C. Lok, et al., Effects of tocilizumab on neutrophil function and kinetics, Eur.
J. Clin. Invest. 47 (10) (2017) 736745, https://doi.org/10.1111/eci.12799.
[98] R.J. Moots, et al., Effect of tocilizumab on neutrophils in adult patients with
rheumatoid arthritis: pooled analysis of data from phase 3 and 4 clinical trials,
Rheumatol. Oxf. Engl. 56 (4) (2017) 541549, https://doi.org/10.1093/
rheumatology/kew370.
[99] S.E. Gabriel, et al., Survival in rheumatoid arthritis: a population-based analysis
of trends over 40 years, Arthritis Rheum. 48 (1) (2003) 5458, https://doi.org/
10.1002/art.10705.
[100] F. Wolfe, et al., The mortality of rheumatoid arthritis, Arthritis Rheum. 37 (4)
(1994) 481494, https://doi.org/10.1002/art.1780370408.
[101] A. Gonzalez, et al., The widening mortality gap between rheumatoid arthritis
patients and the general population, Arthritis Rheum. 56 (11) (2007) 35833587,
https://doi.org/10.1002/art.22979.
[102] Y.-K. Lee, et al., Excess mortality persists in patients with rheumatoid arthritis,
Int. J. Rheum. Dis. 24 (3) (2021) 364372, https://doi.org/10.1111/1756-
185X.14058.
[103] D.P.M. Symmons, S.E. Gabriel, Epidemiology of CVD in rheumatic disease, with a
focus on RA and SLE, Nat. Rev. Rheumatol. 7 (7) (2011) 399408, https://doi.
org/10.1038/nrrheum.2011.75.
[104] H. Maradit-Kremers, et al., Increased unrecognized coronary heart disease and
sudden deaths in rheumatoid arthritis: a population-based cohort study, Arthritis
Rheum. 52 (2) (2005) 402411, https://doi.org/10.1002/art.20853.
[105] P.J. Nicola, et al., The risk of congestive heart failure in rheumatoid arthritis: a
population-based study over 46 years, Arthritis Rheum. 52 (2) (2005) 412420,
https://doi.org/10.1002/art.20855.
[106] O. Schieir, C. Tosevski, R.H. Glazier, S. Hogg-Johnson, E.M. Badley, Incident
myocardial infarction associated with major types of arthritis in the general
population: a systematic review and meta-analysis, Ann. Rheum. Dis. 76 (8)
(2017) 13961404, https://doi.org/10.1136/annrheumdis-2016-210275.
[107] M.J.L. Peters, et al., Does rheumatoid arthritis equal diabetes mellitus as an
independent risk factor for cardiovascular disease? A prospective study, Arthritis
Rheum. 61 (11) (2009) 15711579, https://doi.org/10.1002/art.24836.
[108] I. del Rinc´
on, et al., Systemic inammation and cardiovascular risk factors predict
rapid progression of atherosclerosis in rheumatoid arthritis, Ann. Rheum. Dis. 74
(6) (2015) 11181123, https://doi.org/10.1136/annrheumdis-2013-205058.
[109] K. Lauper, C. Gabay, Cardiovascular risk in patients with rheumatoid arthritis,
Semin. Immunopathol. 39 (4) (2017) 447459, https://doi.org/10.1007/s00281-
017-0632-2.
[110] I. del Rinc´
on, D.F. Battafarano, J.F. Restrepo, J.M. Erikson, A. Escalante,
Glucocorticoid dose thresholds associated with all-cause and cardiovascular
mortality in rheumatoid arthritis, Arthritis Rheumatol. Hoboken NJ 66 (2) (2014)
264272, https://doi.org/10.1002/art.38210.
[111] J. Braun, X. Baraliakos, T. Westhoff, Nonsteroidal anti-inammatory drugs and
cardiovascular risk - a matter of indication, Semin. Arthritis Rheum. 50 (2) (2020)
285288, https://doi.org/10.1016/j.semarthrit.2019.07.012.
[112] F.J. L´
opez-Longo, et al., Association between anti-cyclic citrullinated peptide
antibodies and ischemic heart disease in patients with rheumatoid arthritis,
Arthritis Rheum. 61 (4) (2009) 419424, https://doi.org/10.1002/art.24390.
[113] N.J. Goodson, N.J. Wiles, M. Lunt, E.M. Barrett, A.J. Silman, D.P.M. Symmons,
Mortality in early inammatory polyarthritis: cardiovascular mortality is
increased in seropositive patients, Arthritis Rheum. 46 (8) (2002) 20102019,
https://doi.org/10.1002/art.10419.
[114] G. Hjeltnes, I. Hollan, Ø. Førre, A. Wiik, K. Mikkelsen, S. Agewall, Anti-CCP and
RF IgM: predictors of impaired endothelial function in rheumatoid arthritis
patients, Scand. J. Rheumatol. 40 (6) (2011) 422427, https://doi.org/10.3109/
03009742.2011.585350.
[115] K. Lauper, D.S. Courvoisier, P. Chevallier, A. Finckh, C. Gabay, Incidence and
prevalence of major adverse cardiovascular events in rheumatoid arthritis,
psoriatic arthritis, and axial spondyloarthritis, Arthritis Care Res. 70 (12) (2018)
17561763, https://doi.org/10.1002/acr.23567.
[116] S. Skeoch, I.N. Bruce, Atherosclerosis in rheumatoid arthritis: is it all about
inammation? Nat. Rev. Rheumatol. 11 (7) (2015) 390400, https://doi.org/
10.1038/nrrheum.2015.40.
[117] J. Danesh, et al., Long-term interleukin-6 levels and subsequent risk of coronary
heart disease: two new prospective studies and a systematic review, PLoS Med. 5
(4) (2008), e78, https://doi.org/10.1371/journal.pmed.0050078.
[118] A. Saremi, et al., Association between IL-6 and the extent of coronary
atherosclerosis in the veterans affairs diabetes trial (VADT), Atherosclerosis 203
(2) (2009) 610614, https://doi.org/10.1016/j.atherosclerosis.2008.07.031.
[119] S. Kaptoge, et al., Inammatory cytokines and risk of coronary heart disease: new
prospective study and updated meta-analysis, Eur. Heart J. 35 (9) (2014)
578589, https://doi.org/10.1093/eurheartj/eht367.
[120] Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium
et al., The interleukin-6 receptor as a target for prevention of coronary heart
disease: a mendelian randomisation analysis, Lancet Lond. Engl. 379 (9822)
(2012) 12141224, doi: 10.1016/S0140-6736(12)60110-X.
[121] Y.H. Rho, et al., Inammatory mediators and premature coronary atherosclerosis
in rheumatoid arthritis, Arthritis Rheum. 61 (11) (2009) 15801585, https://doi.
org/10.1002/art.25009.
[122] S.A. Huber, P. Sakkinen, D. Conze, N. Hardin, R. Tracy, Interleukin-6 exacerbates
early atherosclerosis in mice, Arterioscler. Thromb. Vasc. Biol. 19 (10) (1999)
23642367, https://doi.org/10.1161/01.atv.19.10.2364.
[123] B. Schieffer, et al., Expression of angiotensin II and interleukin 6 in human
coronary atherosclerotic plaques: potential implications for inammation and
plaque instability, Circulation 101 (12) (2000) 13721378, https://doi.org/
10.1161/01.cir.101.12.1372.
[124] P. Libby, P.M. Ridker, A. Maseri, Inammation and atherosclerosis, Circulation
105 (9) (2002) 11351143, https://doi.org/10.1161/hc0902.104353.
[125] E. Choy, N. Sattar, Interpreting lipid levels in the context of high-grade
inammatory states with a focus on rheumatoid arthritis: a challenge to
conventional cardiovascular risk actions, Ann. Rheum. Dis. 68 (4) (2009)
460469, https://doi.org/10.1136/ard.2008.101964.
[126] M.M. van Gameren, et al., Effects of recombinant human interleukin-6 in cancer
patients: a phase I-II study, Blood 84 (5) (1994) 14341441.
[127] M.E. Trujillo, S. Sullivan, I. Harten, S.H. Schneider, A.S. Greenberg, S.K. Fried,
Interleukin-6 regulates human adipose tissue lipid metabolism and leptin
production in vitro, J. Clin. Endocrinol. Metab. 89 (11) (2004) 55775582,
https://doi.org/10.1210/jc.2004-0603.
[128] N. Müller, et al., IL-6 blockade by monoclonal antibodies inhibits apolipoprotein
(a) expression and lipoprotein (a) synthesis in humans, J. Lipid Res. 56 (5) (2015)
10341042, https://doi.org/10.1194/jlr.P052209.
[129] H.K. Berthold, M. Laudes, W. Krone, I. Gouni-Berthold, Association between the
interleukin-6 promoter polymorphism -174G/C and serum lipoprotein(a)
concentrations in humans, PloS One 6 (9) (2011), e24719, https://doi.org/
10.1371/journal.pone.0024719.
[130] R. Clarke, et al., Genetic variants associated with Lp(a) lipoprotein level and
coronary disease, N. Engl. J. Med. 361 (26) (2009) 25182528, https://doi.org/
10.1056/NEJMoa0902604.
[131] J. Wang, B. Hu, L. Kong, H. Cai, C. Zhang, Native, oxidized lipoprotein(a) and
lipoprotein(a) immune complex in patients with active and inactive rheumatoid
arthritis: plasma concentrations and relationship to inammation, Clin. Chim.
Acta Int. J. Clin. Chem. 390 (12) (2008) 6771, https://doi.org/10.1016/j.
cca.2007.12.015.
[132] A.D. Pradhan, J.E. Manson, N. Rifai, J.E. Buring, P.M. Ridker, C-reactive protein,
interleukin 6, and risk of developing type 2 diabetes mellitus, JAMA 286 (3)
(2001) 327334, https://doi.org/10.1001/jama.286.3.327.
[133] J. Spranger, et al., Inammatory cytokines and the risk to develop type 2 diabetes:
results of the prospective population-based European Prospective Investigation
into Cancer and Nutrition (EPIC)-Potsdam Study, Diabetes 52 (3) (2003)
812817, https://doi.org/10.2337/diabetes.52.3.812.
[134] J.F. Baker, et al., Disease activity, cytokines, chemokines and the risk of incident
diabetes in rheumatoid arthritis, Ann. Rheum. Dis. (2021), https://doi.org/
10.1136/annrheumdis-2020-219140.
[135] M. Hashizume, H. Yoshida, N. Koike, M. Suzuki, M. Mihara, Overproduced
interleukin 6 decreases blood lipid levels via upregulation of very-low-density
lipoprotein receptor, Ann. Rheum. Dis. 69 (4) (2010) 741746, https://doi.org/
10.1136/ard.2008.104844.
[136] C. Gabay, et al., Comparison of lipid and lipid-associated cardiovascular risk
marker changes after treatment with tocilizumab or adalimumab in patients with
rheumatoid arthritis, Ann. Rheum. Dis. 75 (10) (2016) 18061812, https://doi.
org/10.1136/annrheumdis-2015-207872.
[137] A. Virone, et al., Comparative effect of tumour necrosis factor inhibitors versus
other biological agents on cardiovascular risk-associated biomarkers in patients
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
10
with rheumatoid arthritis, RMD Open 5 (2) (2019), e000897, https://doi.org/
10.1136/rmdopen-2019-000897.
[138] Y. Otsuka, et al., Effects of tumor necrosis factor inhibitors and tocilizumab on the
glycosylated hemoglobin levels in patients with rheumatoid arthritis; an
observational study, PloS One 13 (4) (2018), e0196368, https://doi.org/
10.1371/journal.pone.0196368.
[139] M.C. Genovese, et al., Interleukin-6 receptor blockade or TNF
α
inhibition for
reducing glycaemia in patients with RA and diabetes: post hoc analyses of three
randomised, controlled trials, Arthritis Res. Ther. 22 (1) (2020) 206, https://doi.
org/10.1186/s13075-020-02229-5.
[140] B. Castagn´
e, M. Viprey, J. Martin, A.-M. Schott, M. Cucherat, M. Soubrier,
Cardiovascular safety of tocilizumab: A systematic review and network meta-
analysis, PloS One 14 (8) (2019), e0220178, https://doi.org/10.1371/journal.
pone.0220178.
[141] V.U. Rao, et al., An evaluation of risk factors for major adverse cardiovascular
events during tocilizumab therapy, Arthritis Rheumatol. Hoboken NJ 67 (2)
(2015) 372380, https://doi.org/10.1002/art.38920.
[142] K. Yamamoto, et al., Longterm safety of tocilizumab: results from 3 years of
followup postmarketing surveillance of 5573 patients with rheumatoid arthritis in
Japan, J. Rheumatol. 42 (8) (2015) 13681375, https://doi.org/10.3899/
jrheum.141210.
[143] J. Zhang, et al., Comparative effects of biologics on cardiovascular risk among
older patients with rheumatoid arthritis, Ann. Rheum. Dis. 75 (10) (2016)
18131818, https://doi.org/10.1136/annrheumdis-2015-207870.
[144] R. Fleischmann, et al., Long-term safety of sarilumab in rheumatoid arthritis: an
integrated analysis with up to 7 yearsfollow-up, Rheumatol. Oxf. Engl. 59 (2)
(2020) 292302, https://doi.org/10.1093/rheumatology/kez265.
[145] A.D. Protogerou, E. Zampeli, K. Fragiadaki, K. Stamatelopoulos, C. Papamichael,
P.P. Skakis, A pilot study of endothelial dysfunction and aortic stiffness after
interleukin-6 receptor inhibition in rheumatoid arthritis, Atherosclerosis 219 (2)
(2011) 734736, https://doi.org/10.1016/j.atherosclerosis.2011.09.015.
[146] H. Kobayashi, Y. Kobayashi, J.T. Giles, K. Yoneyama, Y. Nakajima, M. Takei,
Tocilizumab treatment increases left ventricular ejection fraction and decreases
left ventricular mass index in patients with rheumatoid arthritis without cardiac
symptoms: assessed using 3.0 tesla cardiac magnetic resonance imaging,
J. Rheumatol. 41 (10) (2014) 19161921, https://doi.org/10.3899/
jrheum.131540.
[147] G. Markousis-Mavrogenis, et al., The clinical signicance of interleukin-6 in heart
failure: results from the BIOSTAT-CHF study, Eur. J. Heart Fail. 21 (8) (2019)
965973, https://doi.org/10.1002/ejhf.1482.
[148] L.H.D. van Tuyl, K. Michaud, Patient-reported outcomes in rheumatoid arthritis,
Rheum. Dis. Clin. North Am. 42 (2) (2016) 219237, https://doi.org/10.1016/j.
rdc.2016.01.010.
[149] P. Vergne-Salle, et al., The burden of pain in rheumatoid arthritis: Impact of
disease activity and psychological factors, Eur. J. Pain Lond. Engl. 24 (10) (2020)
19791989, https://doi.org/10.1002/ejp.1651.
[150] E.J.F. Santos, C. Duarte, J.A.P. da Silva, R.J.O. Ferreira, The impact of fatigue in
rheumatoid arthritis and the challenges of its assessment, Rheumatol. Oxf. Engl.
58 (Suppl 5) (2019) v3v9, https://doi.org/10.1093/rheumatology/kez351.
[151] M.S. Chimenti, et al., The burden of depressive disorders in musculoskeletal
diseases: is there an association between mood and inammation? Ann. Gen.
Psychiatry 20 (1) (2021) 1, https://doi.org/10.1186/s12991-020-00322-2.
[152] P. Minnock, J. Kirwan, B. Bresnihan, Fatigue is a reliable, sensitive and unique
outcome measure in rheumatoid arthritis, Rheumatol. Oxf. Engl. 48 (12) (2009)
15331536, https://doi.org/10.1093/rheumatology/kep287.
[153] T. Pincus, C. Chung, O.G. Segurado, I. Amara, G.G. Koch, An index of patient
reported outcomes (PRO-Index) discriminates effectively between active and
control treatment in 4 clinical trials of adalimumab in rheumatoid arthritis,
J. Rheumatol. 33 (11) (2006) 21462152.
[154] S. Hewlett, et al., Outcomes generated by patients with rheumatoid arthritis: how
important are they? Musculoskeletal Care 3 (3) (2005) 131142, https://doi.org/
10.1002/msc.3.
[155] L. Gossec, et al., Finalisation and validation of the rheumatoid arthritis impact of
disease score, a patient-derived composite measure of impact of rheumatoid
arthritis: a EULAR initiative, Ann. Rheum. Dis. 70 (6) (2011) 935942, https://
doi.org/10.1136/ard.2010.142901.
[156] K.L. Druce, N. Basu, Predictors of fatigue in rheumatoid arthritis, Rheumatol. Oxf.
Engl. 58 (Suppl 5) (2019) v29v34, https://doi.org/10.1093/rheumatology/
kez346.
[157] D. van Hoogmoed, J. Fransen, G. Bleijenberg, P. van Riel, Physical and
psychosocial correlates of severe fatigue in rheumatoid arthritis, Rheumatol. Oxf.
Engl. 49 (7) (2010) 12941302, https://doi.org/10.1093/rheumatology/keq043.
[158] L.C. Pollard, E.H. Choy, J. Gonzalez, B. Khoshaba, D.L. Scott, Fatigue in
rheumatoid arthritis reects pain, not disease activity, Rheumatol. Oxf. Engl. 45
(7) (2006) 885889, https://doi.org/10.1093/rheumatology/kel021.
[159] D.A. Walsh, D.F. McWilliams, Mechanisms, impact and management of pain in
rheumatoid arthritis, Nat. Rev. Rheumatol. 10 (10) (2014) 581592, https://doi.
org/10.1038/nrrheum.2014.64.
[160] S.D. Boyden, I.N. Hossain, A. Wohlfahrt, Y.C. Lee, Non-inammatory causes of
pain in patients with rheumatoid arthritis, Curr. Rheumatol. Rep. 18 (6) (2016)
30, https://doi.org/10.1007/s11926-016-0581-0.
[161] L. Nerurkar, S. Siebert, I.B. McInnes, J. Cavanagh, Rheumatoid arthritis and
depression: an inammatory perspective, Lancet Psychiatry 6 (2) (2019)
164173, https://doi.org/10.1016/S2215-0366(18)30255-4.
[162] M.R. Irwin, et al., Sleep loss exacerbates fatigue, depression, and pain in
rheumatoid arthritis, Sleep 35 (4) (2012) 537543, https://doi.org/10.5665/
sleep.1742.
[163] J.A. Sturgeon, P.H. Finan, A.J. Zautra, Affective disturbance in rheumatoid
arthritis: psychological and disease-related pathways, Nat. Rev. Rheumatol. 12
(9) (2016) 532542, https://doi.org/10.1038/nrrheum.2016.112.
[164] E.H.S. Choy, L.H. Calabrese, Neuroendocrine and neurophysiological effects of
interleukin 6 in rheumatoid arthritis, Rheumatol. Oxf. Engl. 57 (11) (2018)
18851895, https://doi.org/10.1093/rheumatology/kex391.
[165] F. Atzeni, V. Nucera, I.F. Masala, P. Sarzi-Puttini, G. Bonitta, Il-6 Involvement in
pain, fatigue and mood disorders in rheumatoid arthritis and the effects of Il-6
inhibitor sarilumab, Pharmacol. Res. 149 (2019), 104402, https://doi.org/
10.1016/j.phrs.2019.104402.
[166] A. Ng, et al., IL-1β, IL-6, TNF-
α
and CRP in elderly patients with depression or
Alzheimers disease: systematic review and meta-analysis, Sci. Rep. 8 (1) (2018)
12050, https://doi.org/10.1038/s41598-018-30487-6.
[167] Y. Dowlati, et al., A meta-analysis of cytokines in major depression, Biol.
Psychiatry 67 (5) (2010) 446457, https://doi.org/10.1016/j.
biopsych.2009.09.033.
[168] S. Chourbaji, et al., IL-6 knockout mice exhibit resistance to stress-induced
development of depression-like behaviors, Neurobiol. Dis. 23 (3) (2006) 587594,
https://doi.org/10.1016/j.nbd.2006.05.001.
[169] P. M¨
arz, et al., Sympathetic neurons can produce and respond to interleukin 6,
Proc. Natl. Acad. Sci. U. S. A. 95 (6) (1998) 32513256, https://doi.org/10.1073/
pnas.95.6.3251.
[170] P. M¨
arz, T. Herget, E. Lang, U. Otten, S. Rose-John, Activation of gp130 by IL-6/
soluble IL-6 receptor induces neuronal differentiation, Eur. J. Neurosci. 9 (12)
(1997) 27652773, https://doi.org/10.1111/j.1460-9568.1997.tb01705.x.
[171] M. Thier, P. M¨
arz, U. Otten, J. Weis, S. Rose-John, Interleukin-6 (IL-6) and its
soluble receptor support survival of sensory neurons, J. Neurosci. Res. 55 (4)
(1999) 411422, https://doi.org/10.1002/(SICI)1097-4547(19990215)55:
4<411::AID-JNR2>3.0.CO;2-D.
[172] M. Girotti, J.J. Donegan, D.A. Morilak, Inuence of hypothalamic IL-6/gp130
receptor signaling on the HPA axis response to chronic stress,
Psychoneuroendocrinology 38 (7) (2013) 11581169, https://doi.org/10.1016/j.
psyneuen.2012.11.004.
[173] G. Mastorakos, G.P. Chrousos, J.S. Weber, Recombinant interleukin-6 activates
the hypothalamic-pituitary-adrenal axis in humans, J. Clin. Endocrinol. Metab. 77
(6) (1993) 16901694, https://doi.org/10.1210/jcem.77.6.8263159.
[174] A.M.M. Eijsbouts, F.H.J. van den Hoogen, R.F.J.M. Laan, A.R.M.M. Hermus, C.G.
J. Sweep, L.B.A. van de Putte, Hypothalamic-pituitary-adrenal axis activity in
patients with rheumatoid arthritis, Clin. Exp. Rheumatol. 23 (5) (2005) 658664.
[175] R. Imrich, J. Rovenský, Hypothalamic-pituitary-adrenal axis in rheumatoid
arthritis, Rheum. Dis. Clin. North Am. 36 (4) (2010) 721727, https://doi.org/
10.1016/j.rdc.2010.09.003.
[176] A.N. Vgontzas, E.O. Bixler, H.-M. Lin, P. Prolo, G. Trakada, G.P. Chrousos, IL-6
and its circadian secretion in humans, Neuroimmunomodulation 12 (3) (2005)
131140, https://doi.org/10.1159/000084844.
[177] R. Jankord, R. Zhang, J.N. Flak, M.B. Solomon, J. Albertz, J.P. Herman, Stress
activation of IL-6 neurons in the hypothalamus, Am. J. Physiol. Regul. Integr.
Comp. Physiol. 299 (1) (2010) R343R351, https://doi.org/10.1152/
ajpregu.00131.2010.
[178] R.R. Edwards, T. Kroni, J.A. Haythornthwaite, M.T. Smith, L. McGuire, G.
G. Page, Association of catastrophizing with interleukin-6 responses to acute pain,
Pain 140 (1) (2008) 135144, https://doi.org/10.1016/j.pain.2008.07.024.
[179] G.S. von Banchet, M. Kiehl, H.-G. Schaible, Acute and long-term effects of IL-6 on
cultured dorsal root ganglion neurones from adult rat, J. Neurochem. 94 (1)
(2005) 238248, https://doi.org/10.1111/j.1471-4159.2005.03185.x.
[180] D. Brenn, F. Richter, H.-G. Schaible, Sensitization of unmyelinated sensory bers
of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an
inammatory mechanism of joint pain, Arthritis Rheum. 56 (1) (2007) 351359,
https://doi.org/10.1002/art.22282.
[181] E. Vazquez, J. Kahlenbach, G. Segond von Banchet, C. K¨
onig, H.-G. Schaible,
A. Ebersberger, Spinal interleukin-6 is an amplier of arthritic pain in the rat,
Arthritis Rheum. 64 (7) (2012) 22332242, https://doi.org/10.1002/art.34384.
[182] V. Strand, G.R. Burmester, S. Ogale, J. Devenport, A. John, P. Emery,
Improvements in health-related quality of life after treatment with tocilizumab in
patients with rheumatoid arthritis refractory to tumour necrosis factor inhibitors:
results from the 24-week randomized controlled RADIATE study, Rheumatol. Oxf.
Engl. 51 (10) (2012) 18601869, https://doi.org/10.1093/rheumatology/
kes131.
[183] V. Strand, et al., Sarilumab plus methotrexate improves patient-reported
outcomes in patients with active rheumatoid arthritis and inadequate responses to
methotrexate: results of a phase III trial, Arthritis Res. Ther. 18 (2016) 198,
https://doi.org/10.1186/s13075-016-1096-9.
[184] V. Strand, et al., Sarilumab improves patient-reported outcomes in rheumatoid
arthritis patients with inadequate response/intolerance to tumour necrosis factor
inhibitors, RMD Open 3 (1) (2017), e000416, https://doi.org/10.1136/rmdopen-
2016-000416.
[185] L. Gossec, et al., Effects of Sarilumab on Rheumatoid Arthritis as Reported by
Patients Using the Rheumatoid Arthritis Impact of Disease Scale, J. Rheumatol. 46
(10) (2019) 12591267, https://doi.org/10.3899/jrheum.180904.
[186] J.S. Smolen, et al., Effect of interleukin-6 receptor inhibition with tocilizumab in
patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-
controlled, randomised trial, Lancet Lond. Engl. 371 (9617) (2008) 987997,
https://doi.org/10.1016/S0140-6736(08)60453-5.
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
11
[187] S. Tiosano, et al., The impact of tocilizumab on anxiety and depression in patients
with rheumatoid arthritis, Eur. J. Clin. Invest. 50 (9) (2020), e13268, https://doi.
org/10.1111/eci.13268.
[188] M. Figueiredo-Braga, et al., Inuence of biological therapeutics, cytokines, and
disease activity on depression in rheumatoid arthritis, J. Immunol. Res. 2018
(2018) 5954897, https://doi.org/10.1155/2018/5954897.
[189] J.M. Knight, et al., The IL-6 antagonist tocilizumab is associated with worse
depression and related symptoms in the medically ill, Transl. Psychiatry 11 (1)
(2021) 58, https://doi.org/10.1038/s41398-020-01164-y.
[190] B.K. Pedersen, M.A. Febbraio, Muscle as an endocrine organ: focus on muscle-
derived interleukin-6, Physiol. Rev. 88 (4) (2008) 13791406, https://doi.org/
10.1152/physrev.90100.2007.
[191] B.K. Pedersen, et al., The metabolic role of IL-6 produced during exercise: is IL-6
an exercise factor? Proc. Nutr. Soc. 63 (2) (2004) 263267, https://doi.org/
10.1079/PNS2004338.
[192] A.L. Carey, et al., Interleukin-6 increases insulin-stimulated glucose disposal in
humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated
protein kinase, Diabetes 55 (10) (2006) 26882697, https://doi.org/10.2337/
db05-1404.
[193] G. Karsenty, P. Mera, Molecular bases of the crosstalk between bone and muscle,
Bone 115 (2018) 4349, https://doi.org/10.1016/j.bone.2017.04.006.
[194] A.M.W. Petersen, B.K. Pedersen, The anti-inammatory effect of exercise, J. Appl.
Physiol. Bethesda Md 1985 98 (4) (2005) 11541162, https://doi.org/10.1152/
japplphysiol.00164.2004.
[195] T. Tsujinaka, et al., Interleukin 6 receptor antibody inhibits muscle atrophy and
modulates proteolytic systems in interleukin 6 transgenic mice, J. Clin. Invest. 97
(1) (1996) 244249, https://doi.org/10.1172/JCI118398.
[196] G.J. Grosicki, et al., Circulating interleukin-6 is associated with skeletal muscle
strength, quality, and functional adaptation with exercise training in mobility-
limited older adults, J. Frailty Aging (2019) 17, https://doi.org/10.14283/
jfa.2019.30.
[197] R. Roubenoff, T.B. Harris, L.W. Abad, P.W. Wilson, G.E. Dallal, C.A. Dinarello,
Monocyte cytokine production in an elderly population: effect of age and
inammation, J. Gerontol. A. Biol. Sci. Med. Sci. 53 (1) (1998) M20M26,
https://doi.org/10.1093/gerona/53a.1.m20.
[198] F. De Benedetti, et al., Impaired skeletal development in interleukin-6-transgenic
mice: a model for the impact of chronic inammation on the growing skeletal
system, Arthritis Rheum. 54 (11) (2006) 35513563, https://doi.org/10.1002/
art.22175.
[199] F. De Benedetti, et al., Interleukin 6 causes growth impairment in transgenic mice
through a decrease in insulin-like growth factor-I. A model for stunted growth in
children with chronic inammation, J. Clin. Invest. 99 (4) (1997) 643650,
https://doi.org/10.1172/JCI119207.
[200] F. De Benedetti, et al., Effect of IL-6 on IGF binding protein-3: a study in IL-6
transgenic mice and in patients with systemic juvenile idiopathic arthritis,
Endocrinology 142 (11) (2001) 48184826, https://doi.org/10.1210/
endo.142.11.8511.
[201] H. Tsangari, D.M. Findlay, J.S. Kuliwaba, G.J. Atkins, N.L. Fazzalari, Increased
expression of IL-6 and RANK mRNA in human trabecular bone from fragility
fracture of the femoral neck, Bone 35 (1) (2004) 334342, https://doi.org/
10.1016/j.bone.2004.02.006.
[202] L. Lazzaro, B.A. Tonkin, I.J. Poulton, N.E. McGregor, W. Ferlin, N.A. Sims, IL-6
trans-signalling mediates trabecular, but not cortical, bone loss after ovariectomy,
Bone 112 (2018) 120127, https://doi.org/10.1016/j.bone.2018.04.015.
[203] H.G. Raterman, I.E. Bultink, W.F. Lems, Osteoporosis in patients with rheumatoid
arthritis: an update in epidemiology, pathogenesis, and fracture prevention,
Expert Opin. Pharmacother. 21 (14) (2020) 17251737, https://doi.org/
10.1080/14656566.2020.1787381.
[204] A. Tournadre, G. Vial, F. Capel, M. Soubrier, Y. Boirie, Sarcopenia, Joint Bone
Spine 86 (3) (2019) 309314, https://doi.org/10.1016/j.jbspin.2018.08.001.
[205] T.P. van Staa, P. Geusens, J.W.J. Bijlsma, H.G.M. Leufkens, C. Cooper, Clinical
assessment of the long-term risk of fracture in patients with rheumatoid arthritis,
Arthritis Rheum. 54 (10) (2006) 31043112, https://doi.org/10.1002/art.22117.
[206] K.A. Coulson, G. Reed, B.E. Gilliam, J.M. Kremer, P.H. Pepmueller, Factors
inuencing fracture risk, T score, and management of osteoporosis in patients
with rheumatoid arthritis in the Consortium of Rheumatology Researchers of
North America (CORRONA) registry, J. Clin. Rheumatol. Pract. Rep. Rheum.
Musculoskelet. Dis. 15 (4) (2009) 155160, https://doi.org/10.1097/
RHU.0b013e3181a5679d.
[207] G. Ozen, S. Pedro, F. Wolfe, K. Michaud, Medications associated with fracture risk
in patients with rheumatoid arthritis, Ann. Rheum. Dis. 78 (8) (2019) 10411047,
https://doi.org/10.1136/annrheumdis-2019-215328.
[208] N. Gua˜
nabens, et al., Vertebral fractures are increased in rheumatoid arthritis
despite recent therapeutic advances: a case-control study, Osteoporos. Int. J.
Establ. Result Coop. Eur. Found. Osteoporos. Natl. Osteoporos. Found. USA
(2021) doi: 10.1007/s00198-021-05824-7.
[209] B. Hauser, P.L. Riches, J.F. Wilson, A.E. Horne, S.H. Ralston, Prevalence and
clinical prediction of osteoporosis in a contemporary cohort of patients with
rheumatoid arthritis, Rheumatol. Oxf. Engl. 53 (10) (2014) 17591766, https://
doi.org/10.1093/rheumatology/keu162.
[210] L. Lindner, et al., Osteoporosis in patients with rheumatoid arthritis: trends in the
German National Database 20072017, Rheumatol. Int. 40 (12) (2020)
20052012, https://doi.org/10.1007/s00296-020-04593-6.
[211] T.-H. Li, et al., The prevalence and risk factors of sarcopenia in rheumatoid
arthritis patients: A systematic review and meta-regression analysis, Semin.
Arthritis Rheum. 51 (1) (2021) 236245, https://doi.org/10.1016/j.
semarthrit.2020.10.002.
[212] Y. Yamada, M. Tada, K. Mandai, N. Hidaka, K. Inui, H. Nakamura, Glucocorticoid
use is an independent risk factor for developing sarcopenia in patients with
rheumatoid arthritis: from the CHIKARA study, Clin. Rheumatol. 39 (6) (2020)
17571764, https://doi.org/10.1007/s10067-020-04929-4.
[213] Y.-F. Chen, et al., Synergistic effect of sarcopenia and poor balance on
osteoporotic vertebral fracture in Chinese patients with rheumatoid arthritis, Clin.
Rheumatol. (2021), https://doi.org/10.1007/s10067-021-05703-w.
[214] A. Shin, et al., Comparative risk of osteoporotic fracture among patients with
rheumatoid arthritis receiving TNF inhibitors versus other biologics: a cohort
study, Osteoporos. Int. J. Establ. Result Coop. Eur. Found. Osteoporos. Natl.
Osteoporos. Found. USA 31 (11) (2020) 21312139, doi: 10.1007/s00198-020-
05488-9.
[215] Y.-M. Chen, et al., Tocilizumab potentially prevents bone loss in patients with
anticitrullinated protein antibody-positive rheumatoid arthritis, PloS One 12 (11)
(2017), e0188454, https://doi.org/10.1371/journal.pone.0188454.
[216] I.A. Choi, A. Sagawa, E.Y. Lee, E.B. Lee, Y.W. Song, Tocilizumab increases body
weight and serum adipokine levels in patients with rheumatoid arthritis
independently of their treatment response: a retrospective cohort study, J. Korean
Med. Sci. 35 (22) (2020), e155, https://doi.org/10.3346/jkms.2020.35.e155.
[217] A. Tournadre, et al., Changes in body composition and metabolic prole during
interleukin 6 inhibition in rheumatoid arthritis, J. Cachexia Sarcopenia Muscle 8
(4) (2017) 639646, https://doi.org/10.1002/jcsm.12189.
[218] A. Sepriano, et al., Safety of synthetic and biological DMARDs: a systematic
literature review informing the 2019 update of the EULAR recommendations for
the management of rheumatoid arthritis, Ann. Rheum. Dis. 79 (6) (2020)
760770, https://doi.org/10.1136/annrheumdis-2019-216653.
[219] K.L. Grøn, et al., Overall infection risk in rheumatoid arthritis during treatment
with abatacept, rituximab and tocilizumab; an observational cohort study,
Rheumatol. Oxf. Engl. 59 (8) (2020) 19491956, https://doi.org/10.1093/
rheumatology/kez530.
[220] S. Rose-John, K. Winthrop, L. Calabrese, The role of IL-6 in host defence against
infections: immunobiology and clinical implications, Nat. Rev. Rheumatol. 13 (7)
(2017) 399409, https://doi.org/10.1038/nrrheum.2017.83.
[221] M.F. Doran, C.S. Crowson, G.R. Pond, W.M. OFallon, S.E. Gabriel, Frequency of
infection in patients with rheumatoid arthritis compared with controls: a
population-based study, Arthritis Rheum. 46 (9) (2002) 22872293, https://doi.
org/10.1002/art.10524.
[222] B. Mehta, et al., Serious infection risk in rheumatoid arthritis compared with non-
inammatory rheumatic and musculoskeletal diseases: a US national cohort
study, RMD Open 5 (1) (2019), e000935, https://doi.org/10.1136/rmdopen-
2019-000935.
[223] J.A. Singh, et al., Risk of serious infection in biological treatment of patients with
rheumatoid arthritis: a systematic review and meta-analysis, Lancet Lond. Engl.
386 (9990) (2015) 258265, https://doi.org/10.1016/S0140-6736(14)61704-9.
[224] A. Pawar, et al., Risk of serious infections in tocilizumab versus other biologic
drugs in patients with rheumatoid arthritis: a multidatabase cohort study, Ann.
Rheum. Dis. 78 (4) (2019) 456464, https://doi.org/10.1136/annrheumdis-
2018-214367.
[225] A.I. Rutherford, S. Subesinghe, K.L. Hyrich, J.B. Galloway, Serious infection
across biologic-treated patients with rheumatoid arthritis: results from the British
Society for Rheumatology Biologics Register for Rheumatoid Arthritis, Ann.
Rheum. Dis. 77 (6) (2018) 905910, https://doi.org/10.1136/annrheumdis-
2017-212825.
[226] R. Sakai, et al., Head-to-head comparison of the safety of tocilizumab and tumor
necrosis factor inhibitors in rheumatoid arthritis patients (RA) in clinical practice:
results from the registry of Japanese RA patients on biologics for long-term safety
(REAL) registry, Arthritis Res. Ther. 17 (2015) 74, https://doi.org/10.1186/
s13075-015-0583-8.
[227] P. Emery, H. van Hoogstraten, K. Thangavelu, E. Mangan, G. St John,
P. Verschueren, Subcutaneous sarilumab in patients with rheumatoid arthritis
who previously received subcutaneous sarilumab or intravenous tocilizumab: an
open-label extension of a randomized clinical trial, ACR Open Rheumatol. 2 (11)
(2020) 672680, https://doi.org/10.1002/acr2.11188.
[228] P. Emery, et al., Safety and tolerability of subcutaneous sarilumab and
intravenous tocilizumab in patients with rheumatoid arthritis, Rheumatol. Oxf.
Engl. 58 (5) (2019) 849858, https://doi.org/10.1093/rheumatology/key361.
[229] R. Fleischmann, M.C. Genovese, K. Maslova, H. Leher, A. Praestgaard, G.
R. Burmester, Long-term safety and efcacy of sarilumab over 5 years in patients
with rheumatoid arthritis refractory to TNF inhibitors, Rheumatol. Oxf. Engl.
(2021), https://doi.org/10.1093/rheumatology/keab355.
[230] M.H. Schiff, J.M. Kremer, A. Jahreis, E. Vernon, J.D. Isaacs, R.F. van Vollenhoven,
Integrated safety in tocilizumab clinical trials, Arthritis Res. Ther. 13 (5) (2011)
R141, https://doi.org/10.1186/ar3455.
[231] S. Danese, B. Gao, Interleukin-6: a therapeutic Jekyll and Hyde in gastrointestinal
and hepatic diseases, Gut 59 (2) (2010) 149151, https://doi.org/10.1136/
gut.2008.173534.
[232] C. Klein, et al., The IL-6-gp130-STAT3 pathway in hepatocytes triggers liver
protection in T cell-mediated liver injury, J. Clin. Invest. 115 (4) (2005) 860869,
https://doi.org/10.1172/JCI23640.
[233] F. Hong, S. Radaeva, H. Pan, Z. Tian, R. Veech, B. Gao, Interleukin 6 alleviates
hepatic steatosis and ischemia/reperfusion injury in mice with fatty liver disease,
Hepatol. Baltim. Md 40 (4) (2004) 933941, https://doi.org/10.1002/hep.20400.
[234] L. Campbell, C. Chen, S.S. Bhagat, R.A. Parker, A.J.K. ¨
Ost¨
or, Risk of adverse
events including serious infections in rheumatoid arthritis patients treated with
M. Jarlborg and C. Gabay
Cytokine 149 (2022) 155742
12
tocilizumab: a systematic literature review and meta-analysis of randomized
controlled trials, Rheumatol. Oxf. Engl. 50 (3) (2011) 552562, https://doi.org/
10.1093/rheumatology/keq343.
[235] F. Iannone, et al., Real-world experience of tocilizumab in rheumatoid arthritis:
sub-analysis of data from the Italian biologicsregister GISEA, Clin. Rheumatol.
37 (2) (2018) 315321, https://doi.org/10.1007/s10067-017-3846-8.
[236] P.R. Wood, L. Caplan, Drug-induced gastrointestinal and hepatic disease
associated with biologics and nonbiologic disease-modifying antirheumatic drugs,
Rheum. Dis. Clin. North Am. 44 (1) (2018) 2943, https://doi.org/10.1016/j.
rdc.2017.09.003.
[237] M.C. Genovese, et al., Transaminase levels and hepatic events during tocilizumab
treatment: pooled analysis of long-term clinical trial safety data in rheumatoid
arthritis, Arthritis Rheumatol. Hoboken NJ 69 (9) (2017) 17511761, https://doi.
org/10.1002/art.40176.
[238] H. Wadstr¨
om, T. Frisell, J. Askling, Anti-Rheumatic Therapy in Sweden (ARTIS)
Study Group, Malignant neoplasms in patients with rheumatoid arthritis treated
with tumor necrosis factor inhibitors, tocilizumab, abatacept, or rituximab in
clinical practice: a nationwide cohort study from Sweden, JAMA Intern. Med. 177
(11) (2017) 16051612, doi: 10.1001/jamainternmed.2017.4332.
[239] M. Alfreijat, M. Habibi, P. Bhatia, A. Bhatia, Severe hepatitis associated with
tocilizumab in a patient with rheumatoid arthritis, Rheumatol. Oxf. Engl. 52 (7)
(2013) 13401341, https://doi.org/10.1093/rheumatology/kes397.
[240] F. Xie, H. Yun, S. Bernatsky, J.R. Curtis, Brief report: risk of gastrointestinal
perforation among rheumatoid arthritis patients receiving tofacitinib,
tocilizumab, or other biologic treatments, Arthritis Rheumatol. Hoboken NJ 68
(11) (2016) 26122617, https://doi.org/10.1002/art.39761.
[241] A. Strangfeld, et al., Risk for lower intestinal perforations in patients with
rheumatoid arthritis treated with tocilizumab in comparison to treatment with
other biologic or conventional synthetic DMARDs, Ann. Rheum. Dis. 76 (3)
(2017) 504510, https://doi.org/10.1136/annrheumdis-2016-209773.
[242] X. Jin, T.A. Zimmers, Z. Zhang, R.H. Pierce, L.G. Koniaris, Interleukin-6 is an
important in vivo inhibitor of intestinal epithelial cell death in mice, Gut 59 (2)
(2010) 186196, https://doi.org/10.1136/gut.2008.151175.
[243] S. Grivennikov, et al., IL-6 and Stat3 are required for survival of intestinal
epithelial cells and development of colitis-associated cancer, Cancer Cell 15 (2)
(2009) 103113, https://doi.org/10.1016/j.ccr.2009.01.001.
[244] G. Jones, et al., Comparison of tocilizumab monotherapy versus methotrexate
monotherapy in patients with moderate to severe rheumatoid arthritis: the
AMBITION study, Ann. Rheum. Dis. 69 (1) (2010) 8896, https://doi.org/
10.1136/ard.2008.105197.
[245] T. Koike, et al., Effectiveness and safety of tocilizumab: postmarketing
surveillance of 7901 patients with rheumatoid arthritis in Japan, J. Rheumatol.
41 (1) (2014) 1523, https://doi.org/10.3899/jrheum.130466.
[246] C. Weng, L. Xue, Q. Wang, W. Lu, J. Xu, Z. Liu, Comparative efcacy and safety of
Janus kinase inhibitors and biological disease-modifying antirheumatic drugs in
rheumatoid arthritis: a systematic review and network meta-analysis, Ther. Adv.
Musculoskelet. Dis. 13 (2021) 1759720X2199956, doi: 10.1177/
1759720X21999564.
[247] A.B. Avci, E. Feist, G.R. Burmester, Targeting IL-6 or IL-6 receptor in rheumatoid
arthritis: whats the difference? BioDrugs 32 (6) (2018) 531546, https://doi.
org/10.1007/s40259-018-0320-3.
[248] C. Ancuța, et al., Exploring the role of interleukin-6 receptor inhibitor tocilizumab
in patients with active rheumatoid arthritis and periodontal disease, J. Clin. Med.
10 (4) (2021), https://doi.org/10.3390/jcm10040878.
[249] S. Schreiber, et al., Therapeutic interleukin 6 trans-signaling inhibition by
olamkicept (sgp130Fc) in patients with active inammatory bowel disease,
Gastroenterology (2021), https://doi.org/10.1053/j.gastro.2021.02.062.
[250] R.Q. Le, et al., FDA approval summary: tocilizumab for treatment of chimeric
antigen receptor T cell-induced severe or life-threatening cytokine release
syndrome, Oncologist 23 (8) (2018) 943947, https://doi.org/10.1634/
theoncologist.2018-0028.
[251] O.J. McElvaney, G.F. Curley, S. Rose-John, N.G. McElvaney, Interleukin-6:
obstacles to targeting a complex cytokine in critical illness, Lancet Respir. Med.
(2021), https://doi.org/10.1016/S2213-2600(21)00103-X.
[252] E.G. Favalli, Understanding the role of interleukin-6 (IL-6) in the joint and
beyond: a comprehensive review of IL-6 inhibition for the management of
rheumatoid arthritis, Rheumatol. Ther. 7 (3) (2020) 473516, https://doi.org/
10.1007/s40744-020-00219-2.
[253] M.D. George, J.F. Baker, Perioperative management of immunosuppression in
patients with rheumatoid arthritis, Curr. Opin. Rheumatol. 31 (3) (2019)
300306, https://doi.org/10.1097/BOR.0000000000000589.
[254] G. Evangelatos, V. Koulouri, A. Iliopoulos, G.E. Fragoulis, Tuberculosis and
targeted synthetic or biologic DMARDs, beyond tumor necrosis factor inhibitors,
Ther. Adv. Musculoskelet. Dis. 12 (2020) 1759720X20930116, doi: 10.1177/
1759720X20930116.
[255] M. Soubrier, J. Pei, F. Durand, L. Gullestad, A. John, Concomitant use of statins in
tocilizumab-treated patients with rheumatoid arthritis: a post hoc analysis,
Rheumatol. Ther. 4 (1) (2017) 133149, https://doi.org/10.1007/s40744-016-
0049-8.
M. Jarlborg and C. Gabay
... IL-6 exerts its biological effect through three signalling pathways (classical signalling, trans-signalling, and trans-presentation), which are products of the interaction between IL-6, IL-6R (or its soluble form sIL-6R), and glycoprotein 130 (gp130; or its soluble form sgp130) (see Fig. 1) (Heink et al., 2017;Schaper and Rose-John, 2015). IL-6 trans-signalling is thought to be responsible for most of the pathogenetic effects of IL-6 and is implicated in autoimmune diseases like rheumatoid arthritis and irritable bowel disease (Jarlborg and Gabay, 2022;Schreiber et al., 2021). In IL-6 trans-signalling, the relative abundance of sgp130 over IL-6:sIL-6R complex determines the availability of the IL-6: sIL-6R complex free to trigger trans-signalling. ...
... Furthermore, this study indicates that treatment with steroids and IL-6 inhibitors is associated with delayed onset of anemia, with a subsequent decrease both in the proportion of transfused patients and in the amount of transfusion per patient. These findings are in line with those in patients with other inflammatory conditions, such as rheumatoid arthritis [20]. Collectively, findings suggest inflammation as a causative factor in the occurrence of anemia. ...
Article
Full-text available
Background Anemia is a hallmark of critical illness, which is largely inflammatory driven. We hypothesized that the use of anti-inflammatory agents limits the development of anemia and reduces the need for red blood cell (RBC) transfusions in patients with a hyper-inflammatory condition due to COVID-19. Methods An observational cohort ( n = 772) and a validation cohort (a subset of REMAP-CAP, n = 119) of critically ill patients with hypoxemic respiratory failure due to COVID-19 were analyzed, who either received no treatment, received steroids or received steroids plus IL-6 blocking agents. The trajectory of hemoglobin (Hb) decline and the need for RBC transfusions were compared using descriptive statistics as well as multivariate modeling. Results In both cohorts, Hb level was higher in the treated groups compared to the untreated group at all time points. In the observational cohort, incidence and number of transfused patients were lower in the group receiving the combination treatment compared to the untreated groups. In a multivariate analysis controlling for baseline Hb imbalance and mechanical ventilation, receipt of steroids remained associated with a slower decline in Hb level and the combination treatment remained associated with a slower decline of Hb and with less transfusions. Results remained the same in the validation cohort. Conclusion Immunomodulatory treatment was associated with a slower decline in Hb level in critically ill patients with COVID-19 and with less transfusion. Findings point toward inflammation as an important cause for the occurrence of anemia in the critically ill.
... • Anemia: A mild to moderate anemia is usually seen in polyarthritis, ERA, and PsJIA. A predominant microcytic hypochromic anemia is seen in sJIA because of the effects of IL-6 on iron metabolism and utilization [28]. ...
Article
Full-text available
Juvenile idiopathic arthritis (JIA) is the most common rheumatic disease in children. The International League of Associations for Rheumatology (ILAR) has defined JIA as “arthritis of unknown etiology persisting for ≥6 wk with an onset at <16 y of age, after excluding other causes of joint inflammation”. Synovial inflammation is the result of a complex interplay of aberrant immune systems (both adaptive and innate) in a genetically susceptible individual, with possible external stimuli/triggers. Diagnosis of JIA essentially remains clinical, and laboratory investigations usually help to assess the severity of disease activity. Few investigations like antinuclear antibodies (ANA), human leukocyte antigen (HLA)-B27, and rheumatoid factor (RF) help to categorize or prognosticate a child with JIA. Timely use of effective therapeutic interventions including biological has shown good long-term outcomes of JIA.
... Rheumatoid arthritis (RA) is a progressive autoimmune disease characterized by a significant disability rate [1]. In the early stages of RA, bone destruction is commonly observed, leading to an irreversible period and a substantial loss of labor force, ranging from 33% to 39% within a span of 5-10 years [2][3][4]. ...
Article
Full-text available
Rheumatoid arthritis (RA) is an autoimmune disease characterized by a notably high disability rate, primarily attributed to cartilage and bone degradation. The involvement of heat shock protein 90 (HSP90) as a molecular chaperone in the inflammatory response of RA has been established, but its role in bone destruction remains uncertain. In the present study, the expression of HSP90 was augmented in osteoclasts induced by the receptor activator of nuclear factor-κB ligand. Additionaly, it was observed that the outcomes revealed a noteworthy inhibition of osteoclast formation and differentation when triptolide was utilized to hinder the expression of HSP90. Furthermore, the positive influence of HSP90 in osteoclast differentiation was substantiated by overexpressing HSP90 in osteoclast precursor cells. Mechanically, HSP90 significantly activated the TNF receptor-associated factor 6 (TRAF6)/Nuclear factor of activated T cells 1 (NFATc1) signaling axis, accompanied by markedly promoting osteoclast differentiation. This effect was consistently observed in the destructive joint of rats with collagen-induced arthritis, where HSP90 effectively activated osteoclasts and contributed to arthritic bone destruction by activating the TRAF6/NFATc1 signaling. Overall, the findings of this study provide compelling evidence that HSP90 exacerbates bone destruction in RA by promoting osteoclast differentiation through the activation of TRAF6/NFATc1 signaling, and interference with HSP90 may be a promising strategy for the discovery of anti-arthritic bone destruction agents.
... However, as IL-6 expression in the OB correlates with body weight loss and food intake (Fig. 4), its expression in the OB may be associated with changes in the brain parenchyma. Although IL-6 expression in serum and joint tissues has been reported in RA and mouse models [40,41], our results suggest that OB may also be a source of IL-6 that can act on the brain parenchyma early in the pathology of arthritis. In patients with RA, appetite loss is a common symptom from the early stage of the disease [7,8]; therefore, we were interested in the relationship between IL-6 expression in the OB and appetite loss in patients with RA. ...
Article
Full-text available
Introduction Rheumatoid arthritis (RA) can be comorbid with psychiatric symptoms. Brain abnormalities in RA patients and in arthritis models have been reported. However, it remains unclear when these abnormalities occur and where they are distributed. In this study, we analyzed spatiotemporal changes in gene expression in the brains of mice with collagen-induced arthritis (CIA). Methods Mice were divided into three groups: i) CIA (all mice developed arthritis on day 35): complete Freund’s adjuvant (CFA) and type II collagen at initial immunization, and incomplete Freund’s adjuvant (IFA) and type II collagen at booster immunization; ii) C(+/-) (50% mice developed arthritis on day 35): only IFA at booster immunization; and iii) C(-/-) (no arthritis): only CFA at initial immunization and only IFA at booster immunization. Whole brains were collected at ten stages of arthritis and divided into six sections. Real-time polymerase chain reaction was performed using RNA extracted from the brain, and the expression of proinflammatory cytokines and glial markers was semi-quantified. Arthritis score, body weight, and food and water intakes were recorded and analyzed for correlations with brain gene expression. We also investigated the effect of interleukin-6 (IL-6) injection in the olfactory bulbs (OB) on the food intake. Results After booster immunization, a transient increase in integrin subunit α-M and IL-1β was observed in multiple areas in CIA. IL-6 is persistently expressed in the OB before the onset of arthritis, which is correlated with body weight loss and decreased food intake. This change in the OB was observed in the C(+/-) but not in the C(-/-) group. In the C(+/-) group, non-arthritic mice showed the same changes in the OB as the arthritic mice. This elevation in IL-6 levels persisted throughout the chronic phase until day 84. In addition, IL-6 injection into the OB reduced food intake. Conclusion Persistent elevation of IL-6 in the OB from the early stage of arthritis may be an important finding that might explain the neuropsychiatric pathophysiology of RA, including appetite loss, which is present in the early stages of the disease and manifests as a variety of symptoms over time.
... Patients with RA often develop iron deficiency and different types of anemia as some of the most common comorbidities. The causes of anemia in RA are complex and multifactorial [1,[18][19][20]. In addition, in the presence of chronic inflammation, the major form of systemic hypoferremia is anemia of chronic disease (ACD) in which there is a retention of iron within the body stores. ...
Article
Full-text available
Soluble receptors are important for the balance between ligands and their membrane receptors. Changes in the expression of soluble receptors are associated with human diseases. The aim of this study was to determine the serum levels of sRAGE, sRANKL and OPG in subgroups of rheumatoid arthritis (RA) patients according to the level of CRP and to assess the relationship of these parameters with markers of iron and disease status. The study involved 114 RA patients. The levels of sRAGE, sRANKL and OPG were higher in the subgroup with increased CRP level compared to the subgroup with normal CRP. sRAGE showed a significant positive correlation with sTfR (r = 0.435, p < 0.0001), prohepcidin (r = 0.232, p = 0.04), sRANKL (r = 0.636, p < 0.0001), RF (r = 0.363, p < 0.002), and antiCCP antibodies (r = 0.429, p = 0.003) in the subgroup with normal CRP. Serum sRANKL was positively associated with sRAGE (r = 0.636, p < 0.0001), sTfR (r = 0.513, p < 0.0001), CRP (r = 0.223, p = 0.048), DAS28 (r = 0.269, p = 0.016), RF (r = 0.390, p = 0.001) and antiCCP antibodies (r = 0.445, p = 0.002) also in the subgroup with normal CRP. Serum OPG was positively correlated with ferritin in the subgroup with normal CRP. The association of sRAGE, sRANKL and OPG with markers of iron and disease status in RA suggests a relationship between inflammatory state, osteoclast activation and impaired iron and immune status. Therefore, sRAGE, sRANKL and OPG can be useful markers for the assessment of early pathological changes in RA and can also assist in monitoring of the therapy.
... However, to our knowledge, these results have yet to be reproduced in human subjects. Additionally, patients suffering from autoimmune diseases characterized by increased IL-6 levels, such as rheumatoid arthritis, tend to have a higher risk of developing CVD, supporting the role of IL-6 as a risk factor for atherosclerosis [103]. ...
Article
Full-text available
Citation: Jigoranu, R.-A.; Roca, M.; Costache, A.-D.; Mitu, O.; Oancea, A.-F.; Miftode, R.-S.; Haba, M.S , .C.; Botnariu, E.G.; Mas , taleru, A.; Gavril, R.-S.; et al. Novel Biomarkers for These authors contributed equally to this work. Abstract: Atherosclerosis is a significant health concern with a growing incidence worldwide. It is directly linked to an increased cardiovascular risk and to major adverse cardiovascular events, such as acute coronary syndromes. In this review, we try to assess the potential diagnostic role of biomarkers in the early identification of patients susceptible to the development of atherosclerosis and other adverse cardiovascular events. We have collected publications concerning already established parameters, such as low-density lipoprotein cholesterol (LDL-C), as well as newer markers, e.g., apolipoprotein B (apoB) and the ratio between apoB and apoA. Additionally, given the inflamma-tory nature of the development of atherosclerosis, high-sensitivity c-reactive protein (hs-CRP) or interleukin-6 (IL-6) are also discussed. Additionally, newer publications on other emerging components linked to atherosclerosis were considered in the context of patient evaluation. Apart from the already in-use markers (e.g., LDL-C), emerging research highlights the potential of newer molecules in optimizing the diagnosis of atherosclerotic disease in earlier stages. After further studies, they might be fully implemented in the screening protocols.
... Therefore, IL-6 acts as a pleiotropic autocrine, paracrine and hormone-like regulator of various Table 1 [17][18][19] and Table 2. 20 41 Second, IL-6Rα is only expressed on the surface of certain cells (i.e. hepatocytes, neutrophils, monocytes, adipocytes, myocytes and some populations of lymphocytes), whereas gp130 is expressed by the vast majority of human cells. ...
Article
Full-text available
Rheumatoid arthritis (RA) is a chronic immunoinflammatory rheumatic disease, which manifests as progressive destruction of joints, systemic inflammation of visceral organs and a wide range of comorbidities associated with chronic inflammation. Among the cytokines involved in the pathogenesis of RA and certain other immunoinflammatory rheumatic diseases, the role of interleukin (IL) 6 is of special interest. The introduction of the monoclonal antibodies tocilizumab and later sarilumab, both of which block the IL-6 receptor, into clinical practice was an important achievement in the treatment of immunoinflammatory rheumatic diseases at the beginning of the 21st century. The humanized monoclonal antibody against IL-6, olokizumab, provides a new mode of action by direct inhibition of IL-6. This article reviews new data on the efficacy and safety of olokizumab in RA and the prospects of its use in rheumatology.
Article
Full-text available
Objective To evaluate the comparative efficacy and safety of Janus kinase (JAK) inhibitors and biological disease-modifying antirheumatic drugs (bDMARDs) in patients with rheumatoid arthritis (RA) and an inadequate response to at least one disease-modifying antirheumatic drug (DMARD). Methods PubMed, Embase, Cochrane library and ClinicalTrials.gov were searched for relevant randomized controlled trials (RCTs) from inception to April 2020. The active drugs included three JAK inhibitors and eight bDMARDs while the control drugs included placebo or conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). Outcomes include American College of Rheumatology 20% response (ACR20), Disease Activity Score in 28 joints (DAS28), Health Assessment Questionnaire–Disability Index (HAQ-DI) and discontinuations for adverse events (AEs). We estimated summary odds ratios (ORs) and weighted mean differences (WMDs) using network meta-analysis with random effects. Results Eighty-eight RCTs with 31,566 patients were included. All JAK inhibitors and bDMARDs were more effective than placebo in ACR20 (ORs ranging between 3.05 and 5.61), DAS28 (WMDs ranging between −1.91 and −0.80) and HAQ-DI (WMDs ranging between −0.34 and −0.21). Tocilizumab, certolizumab pegol and upadacitinib showed relatively good efficacy in these three outcomes according to their relative ranking. Notably, tocilizumab was more effective than other active drugs in DAS28 (WMDs ranging between −1.11 and −0.49). Compared with the lower recommended doses, increasing the doses of JAK inhibitors (baricitinib 4 mg versus 2 mg, tofacitinib 10 mg versus 5 mg and upadacitinib 30 mg versus 15 mg) cannot provide significant additional benefits. In terms of discontinuations for AEs, all active drugs showed no significant difference compared with placebo except certolizumab pegol [OR 1.65, 95% credible interval (CrI) 1.06–2.61] and rituximab (3.17, 1.11–10.80). Conclusions Tocilizumab, certolizumab pegol and upadacitinib may have relatively good efficacy in patients with RA after treatment failure with csDMARDs. RA patients taking a JAK inhibitor may have a preference for a lower recommended dose.
Article
Full-text available
Background: The human anti-IL-6 receptor antibody tocilizumab (TCZ) has been approved for the treatment of rheumatoid arthritis (RA) and giant cell arteritis (GCA). It is observed that CRP levels drop quickly after starting TCZ treatment. This may lead to misinterpretation of laboratory results when accessing the patient with infectious disease while on TCZ. We conducted this study to report cases treated with tocilizumab who developed serious infections with special reference to levels of CRP and to review the literature on the effect of tocilizumab on acute phase response (APR) during infections. Methods: The files of RA and GCA patients hospitalized in the Tel Aviv medical center between 2009-2019 were reviewed. Cases of patients with RA and GCA treated with tocilizumab who were hospitalized due to severe infections were reviewed with special emphasis on the duration of treatment, type of infection, and APR. Results: We identified nine admissions. Seven patients were treated with tocilizumab for RA, two for GCA. The diagnosis was pneumonia in three cases, osteomyelitis in one, cellulitis in one, endocarditis due to Whipple disease in one, abscess of cervix uteri in one, meningitis in one, and perforated diverticulitis in one. The mean CRP levels on admission were 4.75 mg/L (normal range, up to 5 mg/L). All cases were diagnosed correctly on admission. Conclusions: CRP levels may not correctly reflect the severity of infectious diseases during tocilizumab treatment. Increased awareness of the masking effect of tocilizumab on the APR during infection is needed in order to avoid a delay in the diagnosis.
Article
Full-text available
Background A large unmet therapeutic need exists in inflammatory bowel diseases (IBD). Inhibition of interleukin (IL)-6 appears to be effective, but the therapeutic benefit of a complete IL-6/IL-6R blockade is limited by profound immunosuppression. Evidence has emerged, that chronic pro-inflammatory activity of IL-6 is mainly mediated by trans-signalling via a complex of IL-6 bound to soluble IL-6R engaging the gp130 receptor without the need of membrane bound IL6R. We have developed a decoy protein, sgp130Fc, which exclusively blocks IL-6 pro-inflammatory trans-signalling and has shown efficacy in preclinical models of IBD, without signs of immunosuppression. Methods We present a 12-week, open label, prospective phase IIa trial (FUTURE) in 16 patients with active IBD treated with the trans-signalling inhibitor olamkicept (sgp130Fc) to assess molecular mechanisms, safety and effectiveness of IL-6 trans-signalling blockade in vivo. We performed in-depth molecular profiling at various time points before and after therapy induction to identify the mechanism of action of olamkicept. Results Olamkicept was well tolerated and induced clinical response in 44% and clinical remission in 19% of patients. Clinical effectiveness coincided with target inhibition (reduction of phosphorylated STAT3) and marked transcriptional changes in the inflamed mucosa. An olamkicept-specific transcriptional signature, distinguishable from remission signatures of anti-TNF (infliximab) or anti-integrin (vedolizumab) therapies was identified. Conclusion Our data suggest that blockade of IL-6 trans-signaling holds large promise for the therapy of IBD and should undergo full clinical development as a new immunoregulatory therapy of IBD.
Article
Full-text available
Background: The aim of our study was to explore the influence of weekly subcutaneous administration of interleukin-6 (IL-6) receptor inhibitor tocilizumab (TCZ) on periodontal status in a local longitudinal study of patients with rheumatoid arthritis (RA) and periodontal disease (PD). Methods: We performed a 6-month prospective study in 51 patients with chronic periodontitis and moderate-to-severe RA starting TCZ in accordance with local recommendations. Extensive rheumatologic (clinical activity, inflammatory, serological biomarkers) and periodontal (visible plaque index, gingival index, bleeding on probing, probing pocket depth, clinical attachment loss) assessments were done. Changes in RA activity and periodontal status were reassessed after 3 and 6 months. Results: We demonstrated significant correlations between periodontal status, disease activity, and serologic biomarkers (p < 0.05). Tocilizumab significantly improved the gingival index scores and decreased the number of sites with bleeding on probing after only 3 months (p < 0.05), while the probing pocket depth significantly decreased after 6 months; overall, clinical attachment loss presented only slight changes without any statistical significance as well as teeth count and plaque levels (p > 0.05). Conclusion: IL-6 inhibition is able to improve periodontal outcomes in patients with RA and concomitant PD, which is essentially related to a dramatic decrease in serum inflammatory mediators.
Article
Full-text available
Because medical illness is associated with increased inflammation and an increased risk for treatment-resistant major depressive disorder, anti-cytokine therapy may represent a novel, and especially efficacious, treatment for depression. We hypothesized that blockade of the interleukin (IL)-6 signaling pathway with tocilizumab would decrease depression and related symptomatology in a longitudinal cohort of allogeneic hematopoietic stem cell transplantation (HCT) patients, a medically ill population with a significant inflammation and psychopathology. Patients undergoing allogeneic HCT received either a single dose of tocilizumab one day prior to HCT (n = 25), or HCT alone (n = 62). The primary outcome included depressive symptoms at 28 days post HCT; anxiety, fatigue, sleep, and pain were assessed at pretreatment baseline and days +28, +100, and +180 post HCT as secondary outcomes. Multivariate regression demonstrated that preemptive treatment with tocilizumab was associated with significantly higher depression scores at D28 vs. the comparison group (β = 5.74; 95% CI 0.75, 10.73; P = 0.03). Even after adjustment for baseline depressive symptoms, propensity score, and presence of acute graft-versus-host disease (grades II–IV) and other baseline covariates, the tocilizumab-exposed group continued to have significantly higher depression scores compared to the nonexposed group at D28 (β = 4.73; 95% CI 0.64, 8.81; P = 0.02). Despite evidence that IL-6 antagonism would be beneficial, blockade of the IL-6 receptor with tocilizumab among medically ill patients resulted in significantly more—not less—depressive symptoms.
Article
The ability to block specific cytokine pathways has revealed pathophysiological differences among autoimmune diseases (e.g., the efficacy of TNF inhibitors in arthritides and inflammatory bowel disorders and their inefficacy in giant-cell arteritis and multiple sclerosis), providing a framework for reclassification.
Article
Objective: To evaluate long-term safety and efficacy of sarilumab over 5 years in patients with RA refractory to TNF inhibitors (TNFi). Methods: Patients in the 24-week randomized controlled trial (RCT) TARGET (NCT01709578) whoreceived double-blind placebo or sarilumab 150 or 200 mg every 2 weeks (q2w), plus conventionalsynthetic DMARDs (csDMARDs), were eligible to receive open-label sarilumab 200 mg q2w pluscsDMARDs in the open-label extension (OLE), EXTEND (NCT01146652). OLE dose reduction to 150 mg q2w was permitted per investigators' judgement or protocol-mandated safety concerns. Safety and efficacy were assessed through treatment-emergent adverse events (AEs), laboratoryabnormalities and clinical disease activity scores. All statistics are descriptive. Results: Of 546 patients, 454 (83%) were treated with sarilumab in the OLE. Cumulative observation period was 1654.8 patient-years (PY; n = 521); 268 patients (51%) had ≥4 years' exposure. Incidencerates per 100 PY of AEs, AEs leading to discontinuation, infection and serious infection were 160.4, 8.1, 57.8 and 3.9, respectively. Neutropenia was the most common AE (15.3 per 100 PY). Absoluteneutrophil count <1000 cells/mm3 (Grade 3/4 neutropenia) was observed in 74 patients (14.2%) and normalized on treatment in 48. Clinical efficacy was sustained through 5 years' follow-up. Efficacy was similar for patients with 1 and >1 TNFi failure, and between patients who either remained on 200mg or reduced to 150 mg. Conclusion: In patients with RA refractory to TNFi, sarilumab's long-term term safety profile was consistent with previous clinical studies and post-marketing reports. Efficacy was sustained over 5years.
Article
Circulating concentrations of the pleiotropic cytokine interleukin-6 (IL-6) are known to be increased in pro-inflammatory critical care syndromes, such as sepsis and acute respiratory distress syndrome. Elevations in serum IL-6 concentrations in patients with severe COVID-19 have led to renewed interest in the cytokine as a therapeutic target. However, although the pro-inflammatory properties of IL-6 are widely known, the cytokine also has a series of important physiological and anti-inflammatory functions. An adequate understanding of the complex processes by which IL-6 signalling occurs is crucial for the correct interpretation of IL-6 concentrations in the blood or lung, the use of IL-6 as a critical care biomarker, or the design of effective anti-IL-6 strategies. Here, we outline the role of IL-6 in health and disease, explain the different types of IL-6 signalling and their contribution to the net biological effect of the cytokine, describe the approaches to IL-6 inhibition that are currently available, and discuss implications for the future use of treatments such as tocilizumab in the critical care setting.
Article
Objectives This study aimed to investigate the synergistic effect of sarcopenia and poor balance on osteoporotic vertebral fracture (VOPF) in Chinese patients with rheumatoid arthritis (RA).MethodsA total of 238 RA patients and 158 normal subjects were enrolled in the case-control study. Poor balance capability (Berg balance scale (BBS) score < 40) and sarcopenia (skeletal muscle mass index (SMI) <7.0 (male)/5.7 (female)) between RA patients and normal subjects were compared. Associations of poor balance capability or sarcopenia with disease activity, structural damage, and joint function in different groups were also investigated.ResultsThe incidence of sarcopenia in RA was 58.4%, significantly higher than that in controls (P<0.0001). Moreover, the percentages of low balance capacity (BBS<40) in RA were 43.7%, which was higher than that in controls (P<0.0001). The prevalence of VOPF in the case group was 19.3%, which was higher than that in the controls (P<0.0001). In the RA group, compared to RA patients without VOPF, RA patients with VOPF had higher percentages of poor balance and sarcopenia (P<0.05). Compared with RA patients without sarcopenia or good balance, RA patients with sarcopenia or poor balance had a higher incidence of VOPF, higher disease activity, severer structural damage, and worse joint function (P<0.05). The incidence of VOPF in patients combined with good balance and non-sarcopenia (4.8%) was significantly lower than that in patients combined with poor balance and sarcopenia (38.2%) (P<0.0001). Logistic regression indicated that higher SMI and higher BBS scores were protective factors for VOPF in RA patients, while age was a risk factor for VOPF in RA patients (P<0.0001).Conclusion Sarcopenia and poor balance are popular in Chinese patients with RA, and they are associated with disease activity and structural damage. There is a synergistic effect of sarcopenia and poor balance on VOPF in RA. Key Points • Sarcopenia and balance capability were popular (about a half) in patients with RA. • Sarcopenia and poor balance had a synergistic effect on VOPF in RA.
Article
Objectives To investigate the causes and risk of death in a large cohort of Korean patients with rheumatoid arthritis (RA). Methods Patients in the Hanyang BAE (Bae registry of Autoimmune diseases for Epidemiology) RA cohort who fulfilled the American College of Rheumatology criteria were analyzed. A total of 2355 patients were enrolled from October 2001 to December 2015. Mortality data were derived by linking with data from the Korean National Statistical Office. Standardized mortality ratio was estimated by dividing observed deaths by expected number of deaths in the general population. Results Over the observation period, 225 deaths were reported. Total age‐ and sex‐adjusted standardized mortality ratio was 1.65 (95% confidence interval 1.44‐1.87). The most common cause of death was malignancy (40 cases; 17.8%), followed by respiratory disease (38 cases; 16.9%) and cardiovascular disease (32 cases; 14.2%). Mortality rate and causes of death differed according to year and age of RA onset. Compared with survivors, individuals who died were more likely to be male, smokers, diagnosed with RA at an older age, and to have long disease duration, higher erythrocyte sedimentation rate and C‐reactive protein, higher rheumatoid factor positivity rate, more severe radiographic damage, and more comorbidities. Conclusion The mortality rate of patients with RA remains higher than that of the general population. Therefore, to improve the survival of patients with RA, attention should be paid to the management of comorbidities as well as to the RA itself.