ArticlePDF Available

Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease

Authors:

Abstract and Figures

Rheumatoid Arthritis (RA), a chronic systemic inflammatory disease which affects approximately 1% of the population, is classically characterized by inflammation and synovitis that leads to cartilage damage and juxta-articular bone destruction. Accumulating evidence indicates that a major cause of death among RA patients is Cardio Vascular Disease (CVD), in excess of that in the general population. In this review, we discuss the epidemiology of CVD in RA populations, the underlying pathophysiologic mechanisms of CVD in RA including the role of chronic inflammation in driving accelerated atherosclerosis, the obesity paradox and altered metabolic pathways leading to pro-inflammatory HDL formation and insulin resistance. We also discuss the pitfalls in the evaluation of CVD utilizing traditional risk scores which tend to underestimate CVD risk in RA population and the efforts directed to find more accurate predictors for early CVD detection. Finally, we will present the latest developments in the evaluation and management of CVD in RA patients, given recent evidence on the role of inflammation and its impact on CVD.
Content may be subject to copyright.
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Review Article Open Access
Publishers
SCIFED
SciFed Journal of Cardiology
Arielle Urman, SF J Cardiol, 2018, 2:3
page 1 of 23
Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular
Disease
Arielle Urman, Nicholas Taklalsingh, Cristina Sorrento, *Isabel M. McFarlane
*Department of Internal Medicine Division of Rheumatology SUNY-Downstate Medical Center Brooklyn, NY 11203, USA
Keywords
Rheumatoid Arthritis; Cardiovascular Risk
Factors; Cardiovascular Disease; Risk Assessment;
Cardiovascular Outcomes; Pathogenesis; Treatment;
Evaluation
Introduction
Rheumatoid Arthritis (RA), a chronic systemic
inflammatory disease which affects approximately
1% of the population, is classically characterized by
inflammation and synovitis that leads to cartilage damage
with joint space narrowing and juxta-articular bone
erosions. CVD risk is increased among RA patients as
demonstrated by numerous epidemiological studies, as the
presence of traditional Cardio Vascular (CV) risk factors
do not explain the higher rate of CV events seen in this
population. The inflammatory milieu of RA, marked
by elevations of serum inflammatory mediators and
endothelial dysfunction, creates an opportune climate for
the development of atherosclerosis and cardio myocyte
dysfunction. Accordingly, CVD in RA is associated
with active RA disease as measured by joint swelling,
extra-articular disease, and elevated serum inflammatory
markers. Clinically, RA patients with CVD present with an
increased rate of silent cardiac disease, atypical symptoms
and diastolic heart failure. Predicting CVD and evaluating
the risk have proven to be difficult in RA, in part due to
the paucity of studies and challenges with risk calculator
models. Imaging techniques and special functional
tests may provide more reliable tools to assess risk and
progression of CVD in this patient population. Optimizing
*Corresponding author: Isabel M. McFarlane, Department of Internal
Medicine Division of Rheumatology SUNY-Downstate Medical Center
Brooklyn, NY 11203, USA. E-mail: Isabel.McFarlane@downstate.edu;
Tel: 718-221-6515
Received September 10, 2018; Accepted September 27, 2018; Published
Citation: Arielle Urman (2018) Inflammation beyond the Joints:
Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
Copyright: © 2018 Arielle Urman. This is an open-access article
distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are credited.
Abstract
Rheumatoid Arthritis (RA), a chronic systemic inflammatory disease which affects approximately 1% of the
population, is classically characterized by inflammation and synovitis that leads to cartilage damage and juxta-articular
bone destruction. Accumulating evidence indicates that a major cause of death among RA patients is Cardio Vascular
Disease (CVD), in excess of that in the general population. In this review, we discuss the epidemiology of CVD in RA
populations, the underlying pathophysiologic mechanisms of CVD in RA including the role of chronic inflammation in
driving accelerated atherosclerosis, the obesity paradox and altered metabolic pathways leading to pro-inflammatory
HDL formation and insulin resistance. We also discuss the pitfalls in the evaluation of CVD utilizing traditional
risk scores which tend to underestimate CVD risk in RA population and the efforts directed to find more accurate
predictors for early CVD detection. Finally, we will present the latest developments in the evaluation and management
of CVD in RA patients, given recent evidence on the role of inflammation and its impact on CVD.
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 2 of 23
prevention and management in RA includes a combination
approach that addresses traditional risk factors and
inflammation.
Epidemiology
The recognition that RA carries a heightened CVD
morbidity and mortality derived from a number of reviews
and meta-analysis. In a review that included 91,618
patients, CVD accounted for 39.6% of all deaths [1]. One
meta-analysis comprising 111,758 patients found a 50%
increased risk of CVD death, with Ischemic Heart Disease
(IHD) and Cerebral Vascular Accidents (CVA) accounting
for 59% and 52% increased risks, respectively [2]. Another
meta-analysis of 14 observational studies concluded a
48% increased risk of incident CVD in patients with RA,
with the risk of Myocardial Infarction (MI) and CVA being
increased by 68% and 41%, respectively, with a single
study identifying the risk of Congestive Heart Failure
(CHF) increased by 87% [3]. These statistics are supported
by a recent prospective population-based cohort study of
CVD end-points showing that RA patients had higher
rates, via adjusted incidence ratio (IRR) of MI (IRR: 1.43),
unheralded coronary death (1.60), heart failure (1.61),
cardiac arrest (2.26), peripheral arterial disease (1.36)
and lower rates of stable angina (hazard ratio: 0.83) [4].
Increased incidence of CV events in RA patients have
been linked to that in diabetics, with a two-fold increase
compared to the general population [5].
Most recently, a large population-based study
matched RA patient’s ≥15 years of age to individuals without
RA. The mortality rate for RA patients was 232-compared
to184 in the non-RA population (14% versus 9%).
Overall, RA patients had increased all-cause mortality, but
age specific mortality ratios suggested excess mortality
among patients younger than 45 years due to respiratory
and circulatory diseases [6]. The majority of population-
based studies are derived primarily from European and
North American cohorts. A recent cross-sectional study
of Chinese patients showed an approximately two-fold
increased risk of CVD, IHD and CHF in RA patients
compared to age and sex-matched controls [7]. One South
African study looked at CVD in RA patients belonging to
an African Black population cohort. Their review argues
that CVD in RA occurring in developed population cohorts
cannot be extrapolated to developing countries population
as further research is needed to ascertain the true disease
prevalence given the degree of heterogeneity in ethnicity
and geographic locations [8].
Pathophysiology
The pathophysiology of CVD in RA involves
immune dysregulation and chronic inflammation which
results from the interaction of genetic and environmental
factors [9]. Inflammation favors atherosclerotic CVD,
with inflammatory markers like C Reactive Protein
(CRP) considered independent predictors for coronary
heart disease in the general population [10, 11]. Evidence
supports that inflammation is the major driver of excess
CVD in RA [12, 13].
Systemic Inflammation and Endothelial
Dysfunction
Elevated levels of cytokines such as tumor necrosis
factor-α (TNF-α), interleukin-17 (IL-17), interleukin-6
(IL-6), and interleukin-1β (IL-1β) are found in both RA
and CVD, with higher levels being present in RA [10, 14-
16]. These cytokines have been implicated in endothelial
cell activation, a crucial step for pannus formation in the
synovial tissue and in the pathogenesis of atherosclerotic
CVD [10, 17, 18]. (Figure 1) Endothelial activation induces
cellular expression of chemokines and adhesion molecules
that enable leukocyte migration into the joint space or
vascular intima which favor further neutrophil recruitment,
activation and propagation of the local inflammatory
process [19-21]. IL-17 induces endothelial cells to express
chemokines that promote neutrophil recruitment [22].
Local T1 helper cells release interferon-gamma (IFN-γ)
which, together with TNF-α and IL-17, causes endothelial
cell apoptosis, [23, 24] eliminating the endothelial cells’
anti-thrombotic properties [25]. In the inflammatory milieu
of RA, atherosclerotic plaques are particularly unstable
and vulnerable to rupture [26, 27]. TNF- α and IL-17 also
prevent nitric oxide and thrombomodulin synthesis and,
along with IL-1 increase tissue factor production and
activation [17, 28-31]. Similarly, IL-6 increases levels of
fibrinogen and IL-6 receptor signaling pathways play a
causal role in coronary heart disease [29, 32]. In concert,
these actions create a hypercoagulable environment in the
vascular lumen.
Results of cytokine-targeted therapy studies
provide proof-of-concept in vivo, confirming the role
of the cytokines and inflammation in RA pathogenesis.
For example, TNF-α blockade improves endothelial-
dependent vascular function in RA patients [33]. In patients
with Coronary Artery Disease (CAD), anti-IL-1β therapy
significantly reduced the rate of subsequent CV events,
independent of lipid-lowering [34]. Furthermore, IL-6
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 3 of 23
receptor inhibition lessened the inflammatory response
and release of troponin-T after acute MI [35].
In addition to direct pro-atherogenic effects,
TNF-α and IL-17 increase insulin resistance, alter lipid
levels and function, and create oxidative stress. IL-17
also affects cardio myocytes by inducing inflammation
and apoptosis which result in myocardial remodeling
due to increased cardiac fibroblast activity and collagen
production [36-39]. These processes likely contribute to
the development of cardiomyopathy and heart failure [40].
There is also widespread vascular dysfunction with
impaired vasodilation, which positively correlates with
elevated inflammatory markers such as high sensitivity
CRP (hsCRP) [41-43]. Furthermore, Coronary Flow
Reserve (CFR) is decreased and carotid Intima-Media
Thickness (cIMT) is increased in RA population [44].
Deranged Lipids and Atheroma Instability
RA and other systemic inflammatory conditions
are associated with a unique type of dyslipidemia
characterized by high levels of triglycerides and low
levels of Low-Density Lipoproteins (LDL) and High-
Density Lipoproteins (HDL). Although lipoprotein levels
are low, there is a paradoxical increase in CV risk [45].
This “lipid paradox” is explained by the fact that low
lipoprotein levels are associated with inflammatory
states, and inflammation is independently associated with
CV events, and perhaps in a more causal fashion than
cholesterol levels. Low lipoprotein levels are associated
with elevations in ESR and CRP [46]. Furthermore, a
clinically significant portion of HDL is altered in structure
and function so that instead of being anti-inflammatory
and atheroprotective, it enhances LDL-oxidation and foam
cell formation [47-50]. This pro-inflammatory HDL has
an altered proteome and does not bind efficiently with
Lecithin-Cholesterol Acyl Transferase (LCAT) resulting in
inadequate clearance of lipids from developing atheromas,
a decreased efflux capacity. Pro-inflammatory HDL levels
positively correlate with acute phase proteins, including
serum amyloid A and complement factors, as well as RA
disease activity [51, 52].
With increased control of inflammation, LDL
levels rise while HDL production is shifted toward the anti-
inflammatory form [53]. Accordingly, decreasing hsCRP
by at least 10 mg/L over two years is associated with a
rise in LDL and increased HDL cholesterol efflux capacity
[54]. Lipid levels rise with the use of Disease Modifying
Anti-Rheumatic Drugs (DMARDs), anti-TNF, anti-IL-6R,
and Janus kinase inhibitors [53, 55-57].
The altered lipid profile in RA may be responsible
for increased CVD events by causing increased instability
of atherosclerotic plaques. A post-mortem study
demonstrated lower overall grades of stenosis and fewer
vessels with severe-grade stenosis, but significantly more
vulnerable plaques, with increased medial and adventitial
inflammation, in RA subjects compared to controls [27].
On ultrasound examination, RA patients had more carotid
plaques than age and sex matched controls without RA.
Controls were found to have more stable plaques [58].
Although RA patients had fewer atheromas and less
severe stenosis, they had higher frequency of CVD events
when compared to the general population suggesting that
atherosclerotic plaques in RA are more prone to rupture.
Hypercoagulability
There is evidence that RA patients are
hypercoagulable. Some of the related pathophysiology was
discussed above in the context of the effects of cytokines
on endothelial cells. To review, elevated concentrations of
fibrinogen, von Will brand factor, fibrin D-dimer, tissue
plasminogen activator antigen, and platelets are seen
in RA patients [59]. Another pathway that contributes
to hypercoagulability is the CD40-CD40L, which is
upregulated in RA and associated with a genetic variant
of CD40. Studies of this pathway in CVD have shown
that levels of soluble CD40L are predictive of MI and
associated with plaque rupture and thrombosis [60-62].
The Role of T Cells
The major risk allele for RA, HLA DRB1, is
associated with the proliferation of an autoreactive
CD4+CD28- T cell population [9]. These T cells are
senescent meaning that, contrary to what the name
Figure 1: Shared Pathogenesis
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 4 of 23
suggests, they are terminal effectors that are highly active
in secreting pro-inflammatory cytokines and resistant
to apoptosis [63]. Significantly, the responsible allele
has also been linked to increased CVD [64]. Expansion
of the CD4+CD28-T cell population is associated with
preclinical cardiovascular disease; these T cells are found
within atherosclerotic plaques [65] where they release
IFN-γ and IL-17, promoting an inflammatory milieu [66].
In one study of RA patients, it was found that patients with
greater expansion of this T cell population had increased
atherosclerosis and endothelial dysfunction, as measured
by carotid IMT and brachial artery Flow Mediated
Vasodilation (FMV), than those with smaller populations
[67]. TNF-α blocking therapy restored CD28 expression
on T cells [68].
It is thought that activation of T cells and
macrophages inside the atheroma by antigens generated
from oxidized LDL may trigger macrophage-mediated
destruction of the fibrous cap with erosion of the overlying
endothelium, bringing thrombogenic particles from
the endothelium into contact with blood, triggering the
coagulation cascade and arterial occlusion, leading to
dreaded cardiovascular events [69].
Risk Factors
CV risk scores valuable for the general population
(Systematic Coronary Risk Evaluation score, Framingham
Risk Score, Reynolds Risk Score) generally underestimate
or overestimate (QRisk II) CV risk in patients with RA
[70]. According to a recent, large, international cohort
study, the Framingham Risk Score in Adult Treatment
Panel (FRS-ATP), RA-related characteristics have been
shown to account for roughly a third of CVD events in
RA patients [71]. There have been many attempts to adjust
pre-existing CVD risk calculators as well as to derive new
risk calculators, but all have resulted in underestimations
[72-74].
In 2015, the Extended Risk Score - Rheumatoid
Arthritis (ERS-RA) was released. The project used data
from the Consortium of Rheumatology Researchers of
North America (CORRONA) cohort, the largest US-based
RA registry to assess newly diagnosed RA patients without
CVD for all possible traditional and RA-related CV risk
factors. Factors such as age, sex, diabetes, hyperlipidemia,
hypertension, and patient-reported tobacco use were
included; in addition to, RA-related factors such as,
level of disease activity, extent of functional disability,
RA duration, presence of subcutaneous nodules, joint
erosions on X-ray, RF or anti-CCP antibody positivity,
any use of corticosteroids or DMARDS, and current use
of methotrexate, NSAIDS, or anti-TNF drugs were also
recorded. Race, education, physical activity, BMI, family
history of early MI, and aspirin use were also considered.
Patients were followed for a mean of 2.9 years. The
researchers concluded that rather than multiplying by
a correction factor [75, 76] or including inflammatory
biomarkers in the calculation, using clinical disease activity
(moderate or high instead of none or low), functional
disability (moderate or high instead of none or low), disease
duration greater than 10 years, and any prednisone use, in
addition to traditional risk factors, resulted in an improved
risk prediction model [77]. However, a later attempt to
externally validate the ERS-RA algorithm found that the
algorithm does not predict CVD in RA more accurately
than risk calculators made for the general population [72].
Other recent efforts that used DAS28, ESR, or health
assessment questionnaires in addition to traditional risk
factors have also failed [73].
Abnormal Body Adiposity
CVD is traditionally associated with abdominal
adiposity and increased BMI. While these associations
hold true in RA patients, there is also an association
between CVD mortality, weight loss and low BMI in RA
[78-82]. One study found that obesity is associated with
decreased mortality in RA patients, however, others have
shown that obesity contributes the same or to a lower
magnitude of CVD risk in RA patients compared to
Figure 2: Risk Factors
Arrows Represent the Causal Relationship between the Activity of
Inflammatory Factors, and the Development of Traditional Risk Factors
(Increased Adiposity, Dyslipidemia, Insulin Resistance) and RA-
Specific Risk Factors that Reflect Disease Activity.
Hs-CRP High Sensitivity C Reactive Protein, ESR Estimated
Sedimentation Rate, TNF Tumor Necrosis Factor, IL-6 Interleukin-6,
IL-1 Interleukin-1, IL-17 Interleukin-17
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 5 of 23
controls [83, 84]. The observed “obesity paradox” appears
to be a consequence of the association between catabolic
weight loss and increased inflammation and mortality [81].
Chronic inflammation in RA is associated with loss of lean
body mass and increased fat mass, specifically abdominal
adiposity and visceral fat; it has been termed rheumatoid
cachexia, there is no net weight loss so that BMI is
maintained. Rheumatoid cachexia may affect 10-67% of
RA patients [85, 86]. (Figure 3) Moreover, RA patients
have decreased muscle and fat mass resulting in low BMI.
Among RA patients, classical cachexia with BMI <20 is
associated with increased CVD-related mortality [82]. Any
seemingly protective effect of obesity likely represents the
absence of intense disease activity [87].
Bioelectrical impedance studies have shown
that RA patients may have more fat mass for a given
BMI than controls, suggesting that BMI measurements
may underestimate fat mass and CVD risk in RA [37,
88]. It has been suggested that the BMI cut-off for
overweight and obesity should be 2 kg/m2 lower in RA
patients in order to appropriately estimate the health risks
conferred by being overweight or obese [88]. Increased
BMI in RA is associated with typical CVD risk factors,
hypercholesterolemia, diastolic hypertension, and elevated
CRP [78, 79, 89].
The relationship between BMI and CVD in
RA is U-shaped in that at low BMI, inflammatory and
RA-related factors predominate contributing to CVD
risk whereas at high BMI, traditional CVD risk factors
contribute significantly. This results in decreased CVD risk
in RA patients with higher BMI compared to those with
lower BMIs, supporting the postulate that inflammation
associated with RA is a more powerful risk factor for
CVD than traditional CVD risk factors. Additionally,
hypertension and smoking confer attributable risk for
CVD to RA patients as seen for the general population [73,
84].
Dyslipidemia
In RA, CVD risk is associated with lower than
expected levels of LDL-C and Total-Cholesterol (T-C)
which means that using LDL-C to extrapolate CVD risk in
RA results in an underestimation [90]. Recent studies have
shown that a U-shaped association between cholesterol
levels and CVD risk also exists in non-RA population [90].
Low cholesterol levels may reflect chronic inflammation
and are associated with high levels of triglycerides and
LDL Particles (LDL-P) [54]. LDL-P and ApoB have been
shown to be a better surrogate for estimating CVD risk than
LDL-C when the two measures are discordant [91-95].
HDL-C also presents a challenge in RA patients.
As discussed above, a significant portion of the HDL-C
is pro-inflammatory and dysfunctional. HDL particles
(HDL-P) concentration appears to be inversely related to
carotid-IMT and CHD than HDL-C [96].
That the higher risk of CVD associated with lower
cholesterol levels in RA reflects increased inflammation
is supported by evidence. In patients receiving DMARDs,
decreasing hsCRP correlated with improvements in HDL
efflux capacity and rising LDL [54].
Insulin Resistance
Insulin resistance is more prevalent in RA with
a rate of 54% in RA compared to 40-45% in the general
population [97]. It has been hypothesized that the increased
prevalence of insulin resistance is due to inflammation. In
RA, insulin resistance has been shown to be associated
Figure 3: Inflammation Alters Traditional Risk Factors
Figure 4: The Good, the Bad, and the Neutral – A New Way of Thinking
of Cholesterol
New evidence has shed light on the ambiguity of cholesterol and
suggests that conventional categorizations oversimplify the matter.
HDL-P High Density Lipoprotein-Particles, Apoa1 Apolipoprotein A1,
T-C Total-Cholesterol, LDL-C Low Density Lipoprotein-Cholesterol,
HDL-C High Density Lipoprotein-Cholesterol, LDL-P Low Density
Lipoprotein-Particles, Apo B Apolipoprotein B
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 6 of 23
with elevations in TNF-α, IL-6, ESR, and CRP, measures
of RA disease activity and importantly, coronary artery
calcification [97]. One study demonstrated no significant
difference between the CVD risk conferred by diabetes to
RA patients compared to non-RA patients [84].
Smoking
Smoking, a strong predictor of CVD in RA is
also more prevalent in RA than in the general population,
although it only contributes in a smaller magnitude to the
CV risk [73, 84, 98].
Genetic Markers
One study demonstrated an increased risk of CV
mortality in RA patients with HLA-DRB1*04 shared
epitope alleles and the association was even stronger
with HLA-DRB1*0404. The HLA-DRB1*04 allele is
also associated with treatment resistant RA, endothelial
dysfunction and extra-articular RA manifestations [12, 99-
105]. The IL-6-174C allele is associated with increased
CVD in RA patients and this corresponds with elevated
IL-6 levels in the carrier’s serum [106].
Clinical Disease Activity
The CORRONA researchers found that per each
10-point reduction in Clinical Disease Activity Index
(CDAI range 0-76) score there was a 21% reduction in
CVD event risk, with a reduction of 53% between high
disease activity and remission [107]. Similarly, other
studies found Disease Activity Score 28 (DAS28), joint
pain severity, functional disability, severe extra articular
RA and, in some cases, disease duration were independently
associated with increased risk of cardiovascular events
and new onset CAD [108-110]. Absence of joint pain is
associated with decreased cardiac risk [109]. Accordingly,
a study of patients treated with anti-IL-6 therapy found that
decreased risk of cardiovascular events correlated with the
extent of reduction in disease activity measured by DAS28
[111]. There is a small but significant increase in CVD risk
in RA patients for time spent in acute flare compared to
time spent in remission. CVD risk for patients in remission
is similar to the risk in patients without RA [112].
On the other hand, a higher score on the Health
Assessment Questionnaire (score 0-3) at one year after
RA diagnosis is an independent predictor of future CVD-
related and all-cause mortality [113]. The association
between CVD risk in RA and extra-articular disease has
been observed in patients with RA vasculitis, pulmonary
disease, rheumatoid nodules among others, with a hazard
ratio of 2.32 in patients with RA lung disease [45].
Disease severity is also associated with subclinical,
premature atherosclerosis, as measured by Coronary
Artery Calcification (CAC) score and cIMT [114, 115].
The greatest difference in CAC between RA and non-RA
patients occurs in the youngest age group [114].
The “time-to-risk profile” depends on the CVD
phenotype [116]. Risk for certain categories of CVD is
increased even prior to RA diagnosis, while risk for others
is not increased until years after diagnosis. For example,
risks for IHD and venous thromboembolism are not
increased prior to diagnosis but increase rapidly after RA
onset [117-119]. The risk for ACS within the first year
after diagnosis is particularly increased in patients with a
high disease activity score [116, 120]. On the other hand,
risk for ischemic stroke is first detectable at ten years after
RA onset [121].
Corticosteroids
It is unclear how corticosteroid use affects CVD
risk although it appears to be related to pre-existing heart
conditions. Corticosteroid use has been shown to increase
the risk of CVD death in patients with RA without CHD
history, even after adjustments for CV risk factors.
However, in patients with history of CHD, corticosteroid
use actually decreases the risk of death from CVD [45,
122, 123]. Use of corticosteroids has been associated
with an increased risk of CV events, as has use of COX-
2 inhibitors and shorter duration of DMARD use [108].
On the other hand, studies have found no significant
association between corticosteroids and CV events
[124]. Low-dose corticosteroid use had no effect on
atherosclerosis, ventricular function, heart rate variability
or arterial stiffness however, an association with major
CV events, including MI, stroke, and death was found
[125]. Glucocorticoid use was associated with increased
CVD risk and poorer survival for RA patients over age
65 at diagnosis, while methotrexate was associated
with decreased CVD risk [126]. We hypothesize, that
RA patients who required corticosteroid treatment had
more active disease and higher levels of inflammation
which in addition to the effects on insulin sensitivity and
sympathetic tone from glucocorticoids led to an increased
rate of cardiovascular events.
Inflammatory Markers
Inflammation is at the heart of the matter in
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 7 of 23
RA patients. (Figure 5) This is reflected by the strong
correlation between disease activity, quantified to some
extent by plasma biomarkers of inflammation, and CVD
risk. Seropositivity for Rheumatoid Factor (RF) and Anti-
Citrullinated Peptide Antibodies (ACPA), along with higher
white blood cell count at diagnosis, and higher cumulative
C-Reactive Protein (CRP), Erythrocyte Sedimentation
Rate (ESR) and pain-visual analog scale scores were
associated with higher CVD risk among patient <65 years
of age at RA diagnosis [126]. Inflammatory markers (ESR,
CRP, WBC, IL-6, TNF-alpha) have a stronger association
with fatal CVD and heart failure than with atherosclerosis
and MI [45, 127].
C Reactive Protein (CRP)/High-Sensitivity CRP
(hsCRP)
Elevated serum CRP levels are associated
with increased CVD risk, including MI, heart failure,
atherosclerosis, and mortality in RA [124, 128]. It is also
an independent predictor of CVD risk, specifically of MI,
in healthy people without RA. The association holds true
even in sub-groups of patients with LDL cholesterol levels
below 130 mg/dl and is a stronger predictor of CVD events
than LDL-cholesterol, atherogenic cholesterol index,
serum amyloid A, IL-6, homocysteine, and Inter Cellular
Adhesion Molecule-1 (ICAM-1) [129]. An hsCRP level
of >5 mg/dL has been shown to be an independent and
statistically significant predictive marker of CV death in
RA patients with a hazard ratio of 3.9 and 4.22 for men and
women, respectively [128]. A CRP > 10 mg/L has been
associated with increased MI with a hazard ratio of 2.12
(95% CI 1.02 to 4.38) [130].
Elevated CRP levels has also been associated
with a significantly increased risk of heart failure and
mortality with hazard ratios 1.25 and 1.08, respectively, in
RA patients [131]. In both RA and the general population,
elevated CRP is associated with number of atherosclerotic
plaques and with increased carotid IMT. A proposed
mechanism for these strong associations is that CRP
promotes atherogenesis by increasing uptake of oxidized
LDL cholesterol by macrophages and inhibiting cholesterol
efflux from foam cells, and also promotes plaque rupture
by promoting the production of matrix metalloproteinases
by macrophages [132, 133].
ESR
Elevated ESR is independently associated
with increased cardiovascular risk [108, 122]. Studies
have shown that ESR > 42 mm/h carries increased MI
and ischemic stroke risks [130] and ESR=50 mm/h is
associated with approximately 10-fold increase in CVD
risk [130, 131]. A 15 year follow-up study found increased
risk for CVD death with a hazard ratio of 2.03 ((95% CI)
1.45-2.83) for patients with at least 3 ESR values of at
least 60 mm/hr [45].
RF Positivity
Many studies have shown that RF positivity (+)
is significantly associated with increased cardiovascular
and all-cause mortality in RA [134, 135]. In fact, the
mortality gap between RF (-) RA patients and the general
population has remained stable in recent years while the
gap for RF (+) patients has increased [136]. Interestingly,
the association between RF (+) and increased CV and all-
cause mortality holds true even in patients without RA
[137]. RF (+) is positively associated with smoking and
diabetes but correlates inversely with cholesterol levels
[138]. Seropositivity confers increased cardiovascular risk
and all-cause mortality, even after adjusting for smoking
and diabetes [139]. RF (+) also strengthens the association
between CRP levels and CV death [128] However, some
more recent studies have not found an association between
RF positivity and CVD risk [77, 120].
ACPA Positivity
Studies evaluating ACPA positivity as a risk
factor for CVD in RA have produced mixed results. ACPA
positivity was shown to be associated with increased risk
of fatal CVD and death but lower or no increased risk
of non-fatal CVD, CHD, or stroke compared to ACPA
negative patients [108, 109, 140, 141]. Other studies
found borderline associations with CVD events without
statistical significance [120, 140]. Studies that looked at
Figure 5: Inflammation is the Hub of the Risk Factor Wheel
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 8 of 23
coronary atherosclerosis and ACPA positivity found no
association even though citrullinated proteins were found
within atherosclerotic plaques [142]. However, another
study found that ACPA antibodies are independently
associated with IHD (6.5% versus 2.6%) and higher
mortality rates (11.2% versus 6.8%) compared to ACPA
antibody negative RA patients [143]. Interestingly, an
association between high levels of ACPA and reduced left
ventricular mass, end diastolic volume, and stroke volume,
but not reduced ejection fraction has been described.
Cytokines
Elevated levels of IL-6 are associated with CVD
in RA and in the general population, and IL-6 receptor
signaling pathways are known to have a causal role in the
development of CAD [32]. Elevated levels of IL-6 predict
cardiovascular event risk in healthy men and women.
[129, 144, 145] Among women with RA, IL-6 is strongly
associated with increased cardiovascular and all-cause
mortality rather than non-fatal CVD or CHD [109].
Elevated cytokine concentrations are associated
with subclinical CVD in RA patients. Elevated levels
of inflammatory markers such as IL-6, Serum Amyloid
A (SAA), intercellular adhesion molecule-1 (ICAM-1),
E-selectin, TNF-α, and myeloperoxidase were found in RA,
but only IL-6 and TNF-α concentrations correlated with
high coronary calcium scores, independent of Framingham
risk score and diabetes [146]. Elevated IL-6 is associated
with higher levels of markers of endothelial dysfunction,
ICAM-1 and Vascular Cell Adhesion Molecule-1 (VCAM-
1) [147].
Other biomarkers being investigated are high-
sensitivity cardiac troponin I (hs-cardiac troponin 1) and
N-Terminal pro-Brain Natriuretic Peptide (NT-proBNP).
Hs-cardiac troponin 1 has been shown to be independently
associated with subclinical coronary plaques and a
predictor of CV events in RA patients [148]. NT-proBNP
is an independent predictor of CV mortality and has been
shown to be most useful as a predictor when levels are at
least 100 pg/ml [149-152].
Clinical Manifestations of Cardiovascular Disease
in Rheumatoid Arthritis
Ischemic Heart Disease
RA is associated with an increased risk of IHD.
Prospective cohort studies place the relative risk of MI in
RA between 1.7 and 2.0, and meta-analyses data estimate
a standardized mortality ratio of IHD between 1.59 and
1.77 [2, 153-156]. There is evidence that subclinical
atherosclerotic disease is more common in RA. RA
patients who underwent Computed Tomography (CT)-
angiography for evaluation of coronary plaques had a
significantly higher proportion of plaques when compared
with controls [157]. RA patients were also more likely
to have single, double, and triple-vessel disease, >50%
stenosis, and extensive segmental disease. Moderate RA
disease activity (DAS28 ≥3.2) is most closely associated
with non-calcified coronary plaque [157]. RA patients
who were identified to have unilateral or bilateral carotid
plaques by ultrasound examination were more likely to
have newly incident Acute Coronary Syndrome (ACS)
compared to RA patients without carotid artery plaques
[115]. This may point to carotid artery plaque identification
as a potential predictive tool for estimating CAD and CVD
event risk in RA patients.
Just as higher RA disease activity has been
associated with an increased risk of subclinical
atherosclerotic disease, several studies have demonstrated
that high disease activity is associated with an increased risk
of incident IHD. RA patients with ACS have significantly
higher DAS28 scores [116]. RA patients with IHD have
more tender joints [158]. ACS and IHD are associated with
elevated ESR in RA patients [116, 158]. A 10-point drop in
Clinical Disease Activity Index (CDAI) resulted in a 21%
decrease in composite cardiovascular risk. The effect of
disease activity may be cumulative as increased risk with
extended disease duration is observed; RA patients at >10
years after diagnosis had a significantly increased risk of
MI, while those at <10 years after diagnosis did not have
a significantly increased risk. These findings suggest that
both disease length and severity affect the risk of developing
IHD in RA. Furthermore, it has been demonstrated that the
relative risk of MI, stroke, or CV death in RA patients is
highest among young RA patients and lowest for those >
75 years old. This risk-age discrepancy could be explained
by the fact that high-risk RA patients suffered premature
CV death leaving a relatively healthy older population
[107].
It appears that RA patients experience IHD
differently from the general population. RA patients
are five times more likely to have unrecognized MIs,
identified by characteristic ECG findings in a patient with
no documented history of previous MI) [45]. RA patients
are more likely to suffer from sudden cardiac death and
have increased frequency of ST-segment elevation MI,
higher troponin levels, more inpatient complications, and
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 9 of 23
higher short-term mortality after first acute MI or stroke
than non-RA patients [45, 116, 159]. In addition, a review
of Acute Coronary Syndrome (ACS) treatment patterns
and outcomes among RA patients found not only a higher
case fatality rate for MI but also that these patients were
less likely to receive coronary artery bypass grafting,
percutaneous reperfusion, and secondary prevention of
MI including beta-blockers and lipid-lowering agents
[116, 160]. On the other hand, one study that found no
difference in ACS management still found worse mortality
and increased recurrent ischemia in RA compared to non-
RA patients [161].
Congestive Heart Failure
RA has also been associated with an increased
risk of ischemic and non-ischemic CHF, with a more
significantly increased risk for ischemic CHF [162-169].
RA patients have a higher rate of diastolic dysfunction, but
no difference in left ventricular ejection fraction compared
to the general population [137, 166, 170-175].
In non-RA populations, ischemic CHF can be
attributed to hypertension, smoking, high BMI, and
alcohol abuse however, in RA, those traditional risk
factors do not fully explain the occurrence of the disease
prevalence and RA patients are less likely to have obesity
and hypertension [176]. As in IHD, RA patients are more
likely to experience CHF differently from non-RA patients
with greater incidence of sub-clinical disease; they present
with less orthopnea, dyspnea on exertion, and paroxysmal
nocturnal dyspnea compared to controls. This absence of
the cardinal symptoms of CHF could potentially delay
diagnosis and treatment in RA patients [176]. Stroke and
atrial fibrillation have also been found to be more prevalent
in RA patients, with a 40% higher risk of atrial fibrillation
and a 30% increased risk of stroke in RA compared to the
general population [177].
Evaluation and Workup
It is of paramount importance that CV disease be
promptly diagnosed and evaluated in patients with RA
given the heightened risk of mortality [2]. The workup
of CV disease in RA should include history, physical
examination, laboratory tests, electrocardiography
and echocardiography, with a specific focus on IHD,
heart failure, myocarditis, micro-vascular disease and
pulmonary hypertension, conditions more prevalent in
RA [163, 178, 179]. Risk assessment tools, such as the
Systematic Coronary Risk Evaluation (SCORE) index
and Framingham models were developed to estimate
CV risk in the general population [180, 181]. These
tools underestimate the CV risk in RA largely due to the
exclusion of non-traditional risk factors [70, 182]. Studies
have shown that RA patients deemed to have moderate
risk by these models, actually have carotid plaques which
place them at a much higher CV risk [183, 184]. The
QRESEARCH CV Risk Algorithm 2 (QRISK2), a risk
calculator that incorporates RA in its assessment, was
found to be of value in the determination of fatal versus
non-fatal CV risk but appears to overestimate CV events
in RA [70, 76].
The Modified SCORE (mSCORE) index,
developed by the European League Against Rheumatism
(EULAR), has shown a higher 10-year CV risk in patients
with RA. This emphasized the need to include RA disease-
specific factors to the estimation of CV risk in calculation
tools [75, 185, 186]. The 2016 updated EULAR
recommendations advised a multiplication factor of 1.5 for
CV risk assessment calculators when used in patients with
RA [186]. However, current research suggests that many
of these risk assessment tools, including the Expanded
CV Risk Prediction Score for Rheumatoid Arthritis (ERS-
RA), EULAR multiplier and QRISK2, are still not ideal
[72, 77]. Future research is likely to provide a more
accurate CV risk assessment calculator to be used among
RA population.
Figure 6: Elevated Inflammatory Biomarkers Found in Rheumatoid
Arthritis
These markers are being studied for their usefulness in contributing to
cardiovascular disease risk assessment tools in rheumatoid arthritis
patients CRP C-reactive protein, ESR Estimated Sedimentation Rate,
NT-probnp N-terminal pro brain natriuretic peptide, TNF-Α tumor
necrosis factor-alpha, IL-6 interleukin-6, IL-Β interleukin-beta, IL-17
interleukin-17, ICAM-1 Intercellular Adhesion Molecule-1, VCAM-
1 Vascular Cell Adhesion Molecule-, Hs-cardiac troponin 1 high
sensitivity-cardiac troponin 1, RF Rheumatoid Factor, ACPA Anti-
Citrullinated Protein Antibodies
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 10 of 23
Serum biomarkers have been studied as surrogate
CV end points in RA [187] (Figure 6). The Reynold’s risk
assessment incorporated hsCRP given its usefulness across
a full range of CV risk profiles. However, the Reynold’s
risk assessment failed to accurately predict CVD in RA
[188-190].
Imaging studies are also useful in determining CV
risk with high sensitivity and specificity. An ideal imaging
tool would be able to provide: accurate prediction of CV
mortality, early subclinical detection of atherosclerosis,
longitudinal evaluation of interval changes in CVD
and detection of impact of atherosclerosis and other
manifestations of CVD [191]. Endothelial dysfunction has
been reliably associated with impending atherosclerotic
disease [192]. (See Figure 1) Flow-mediated dilation
(FMD) and Brachial Artery Reactivity Testing (BART) can
help estimate endothelial function [193, 194]. However,
rigorous research found that of the parameters measured
in BART, only forearm hyperemic flow could estimate
the 10-year CV risk in RA by correlation with AHA/ACC
risk categories [195]. While impaired in RA, FMD did not
show significant correlation with estimated 10-year CV
risk [195, 196]. Utilizing ultrasonography (US), the aortic
Pulse Wave Velocity (aPWV) and augmentation index
(AIx) can provide a measurement of arterial stiffness [193,
194]. A meta-analysis concluded that arterial stiffness was
abnormal in patients with RA [197]. Studies have shown
that although aPWV and AIx can predict CV risk in the
general population, they are not useful to predict CV
events in RA [198].
Carotid plaque and cIMT, as measured by US, have
been independently associated with CV risk determination
in the general population [199]. A 5-year prospective study
of RA patients without CV risk factors showed that cIMT
≥0.90 mm, predicted CV events in >60% of the cohort. A
cIMT ≤0.77 mm was associated with no CV events. cIMT
is useful in estimating CV risk in RA patients who may
be underestimated by conventional risk scoring calculators
[115, 200]. Studies showed that 65% of patients classified
as moderate risk and 85% of those in high or very high-
risk categories have increased cIMT and/or carotid plaque,
signifying its utility in re-estimating CV risk in this
intermediate-risk cohort [183]. Carotid plaques, defined as
a localized intima-media thickening of >1 mm, was shown
to have significant correlation with poor CV survival
and ischemic CV events. These findings are particularly
important in the presence of bilateral carotid plaques,
where the incidence of CV events rises by over a factor
of 4 [115, 201]. The updated EULAR recommendations
suggest that carotid US be used to screen for asymptomatic
atherosclerotic plaques as part of CV disease evaluation in
patients with RA [186].
Cardiac CT and CAC score are useful modern
imaging techniques to quantify risk in RA patients. Similar
to the ultra-sonographic techniques, CAC is well known
to be a useful risk prediction marker of CV events in the
general population [202]. A study of 227 patients showed
that patients with both early and established RA had more
severe CAC scores. Patients with established disease had
an odds ratio of 3.42 for severe CAC when adjusting for
CV risk factors, making cardiac CT a useful discriminator
of CV risk in RA patients [203]. Cardiac MRI (CMR)
can be used to quantify the extent of myocardial damage
in patients with RA [204]. Although limited literature
currently exists on CMR use in RA, patients tend to have
reduced left ventricular mass and ejection fraction [172].
Myocardial strain imaging with the use of
Speckle-Tracking Echocardiography (STE) is a new
technique to evaluate myocardial function that can detect
early myocardial dysfunction in patients with RA [205].
The Mayo Clinic used this modality to detect subclinical
myocardial diastolic dysfunction in a retrospective study
of 87 patients with RA without history of CV disease.
Global left ventricular and right ventricular strain were
reduced in RA patients, with worse strain correlating
with other markers of RA disease severity [205]. An
Italian study examining RA patients free of CV disease
and normal transthoracic echocardiograms, demonstrated
statistically significant reductions in LV end-systolic radial
and longitudinal strains when compared to the general
population controls [206]. Larger prospective trials are
needed to confirm its usefulness as a routine modality
and clinical utility in predicting CV events. Table 1 for a
summary of special tests that may be used to assess CVD
in RA).
Additionally, genetic and serological markers are
being studied to evaluate CV disease in RA patients. Human
leukocyte antigens, cytokines, adipokines, endothelial
cell activation markers, and their related genes all show
a significant correlation with the development of CV risk
in RA patients [206, 207] (See Figure 6). Age-adjustments
should also be considered when evaluating CV risk in RA
patient since younger RA patients tend to have higher rates
of CAD, and dyslipidemia deserves special attention as
previously discussed [208].
An accurate CV risk assessment tool would involve
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 11 of 23
a composite score that factors in clinical and demographic
data, carotid US results and serological and genetic serum
markers, providing a more individualized approach to CV
risk for patients with RA [207]. We encourage clinicians
to adopt a cautious approach when investigating patients
with RA and maintain a low threshold for treatment and
start prevention strategies to minimize the burden of CV
disease in patients with RA.
Management
Chronic inflammation is now regarded as the
driver of accelerated atherosclerosis in the RA population
[209, 210]. Effective disease control seems to be pivotal
to preventing atherosclerosis and its consequences. One
of the first studies to show that disease control improved
mortality was a retrospective review of RA patients
receiving intramuscular gold therapy [211]. Disease
Modifying Anti-Rheumatic Drugs (DMARD) and
particularly methotrexate (MTX) soon were also found to
reduce mortality up to 50%. In addition, MTX treatment
reduced ESR and CRP, and raised TC and HDL levels
[212, 213]. Hydroxychloroquine (HCQ) has also been
found to have a favorable effect on lipid profile and has an
additional antithrombotic effect [214]. HCQ reduces total
cholesterol (T-C), LDL-C and increases HDL-C leading to
a favorable atherogenic index [215-217].
The availability of DMARDs and anti-TNF
therapies have been associated with decreases in CV
fatalities in RA [218]. Clinical reports support the
notion that arrhythmic events are significantly reduced
in patients taking anti-TNF therapies or MTX [111, 219,
220]. Elevated levels of hsCRP and IL-6 were found to
be strong and independent predictors of sudden cardiac
death among a healthy cohort reinforcing the idea that
inflammatory cytokines induce structural, chronic cardiac
sympathetic activation, and myoelectrical alteration which
favor arrhythmogenic events in the non-RA population
[221, 222]. Moreover, triple therapy with MTX plus
sulfasalazine and HCQ for 52 weeks was demonstrated
to reduce disease activity and increase HDL-C, lower
LDL-C, and lower the atherogenic index [223].
A five-year study that compared RA patients on
synthetic DMARDs to those on anti-TNF therapy found
that the risk of MI was decreased by 39% in the latter
group. The study also demonstrated that 16 months of anti-
TNF therapy was not only associated with a lower CV risk,
but also influenced disease severity and mortality post MI.
These findings reinforced the concept that a reduction of
vascular inflammation in RA is linked to better outcomes
by decreasing atherosclerosis which entails improved
endothelial function, plaque stabilization and post-MI
remodeling [224, 225]. The randomized control trial
of golimumab in MTX refractory RA patients (GO-
FORWARD) revealed that at 14 weeks, the T-C, LDL-C
and HDL-C, had increased in the MTX-golimumab group
compared to the placebo-MTX control [226, 227]. In
MTX naïve patients (GO-BEFORE), rises in the lipid
parameters at 24 weeks were similar in the golimumab-
MTX and placebo-MTX groups, which supported the
notion that the improvements in lipid profile were due
to MTX effectiveness in naïve patients [228, 229].
Tocilizumab (TCZ)’s effects (IL-6R inhibitor) on lipids
and CV risk was examined in the MEASURE trial. TCZ
resulted in a decrease of HDL-associated serum amyloid
A, phospholipase A2, lipoprotein A, fibrinogen, D-dimer
and elevation of paraoxonase while ApoB/ApoA1 ratio
remained stable. This shows that TCZ induced elevations
in LDL-C but altered HDL particles towards an anti-
inflammatory composition, ameliorating many vascular
risk surrogates [53, 229].
The relationship of TCZ and major CV adverse
events (MACE) was studied in about 4000 RA patients
with moderate to severe RA. TCZ’s ability to decrease
disease activity positively correlated with a lower risk
of future MACE. The higher the disease activity despite
TCZ, the higher the risk of future MACE. Moreover, lipid
parameters while on TCZ were not useful as markers of
risk for MACE [111].
JAK inhibitors also increase HLD-C and LDL-C
Table 1: Special Tests
Predictive Requires Further
Testing Not Predictive
Forearm
Hyperemic Flow
[195]
Cardiac MRI [204]
Flow-Mediated
Dilation [195, 196]
Carotid Intima
Media Thickness
[115, 183, 186,
201]
Brachial Artery
Reactivity [195]
Cardiac CT and
Coronary Speckle-Tracking
Echocardiography
[205]
Aortic Pulse Wave
Velocity [198]
Artery
Calcification
Score [203]
Augmentation Index
[198]
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 12 of 23
in a similar fashion as seen with tocilizumab, with a
reduction in the CV risk and decreased cIMT despite the
elevation in lipid profile. Pooled analysis found the MACE
events were similar for patients on tofacitinib or placebo
and this risk did not increase over time. However, in 2017
concerns for venous thromboembolic events lead the FDA
to establish mandatory reporting of all MACE for the JAK
inhibitors class. Venous thromboembolism has been seen
as an adverse effect with the entire class, and portal vein
thrombosis was linked to ruxolitinib [230]. This should be
kept in mind as pulmonary embolism appears to be a class-
wide issue for JAK inhibitors.
The anti-inflammatory effect of statins was tested
in the TARA trial (Trial of Atorvastatin in Rheumatoid
Arthritis), in which RA patients 50 years old without
known CVD or diabetes, who had RA for ≥ 10 years and
were maintained on DMARD therapy, were initiated on
atorvastatin or placebo as adjuvant therapy and followed
for 6 months. Modest but significant improvements were
seen in disease activity index and swollen joint count in
addition to a decrease in ESR and CRP of 28% and 50%,
respectively [231]. Furthermore, a large population-
based study in which non-RA patients on statins were
followed for 12 years looked at the crude incidence of
RA in the population. It found that the high dose statin
group (atorvastatin 20-80 mg, rosuvastatin 5-40 mg, or
simvastatin 80 mg) was associated with a 23% reduction
in the risk of incident RA, compared to the low-dose statin
group, suggesting that statins may reduce inflammation
directly [232]. Statins have also been demonstrated to
improve arterial stiffness and endothelial dysfunction
[233, 234]. Remarkably but not surprisingly, a population
study of RA patients who had their statins discontinued for
more than 3 months revealed a 67% increase in the risk of
MI with a 2% increased risk of acute MI for each month of
discontinuation [235]. The addition of a second non-statin
lipid lowering therapy such as ezetimibe was also studied
in patient with hsCRP >6 mg/dL, leading to significant
reductions in LDL-C, hsCRP and disease severity [236].
A propensity score analysis of RA patients on at least one
DMARD demonstrated that the concomitant use of statins
was associated with a 21% lower risk of all-cause mortality
[237].
The benefits of early therapy in RA were confirmed
by a 20-year follow up study of a cohort of RA patients who
had low disease activity throughout, but in whom disability
was apparent by year seven of disease and continued to
rise regardless of whether they received treatment for their
RA. In this study 44% of patients died, however, patients
who had received DMARD treatment within 6 months
of disease onset tended to have a lower risk of death that
those who did not receive treatment [238]. Early initiation
of statins should also be considered for patients with RA,
even in the absence of dyslipidemia given that this patient
population carries high risk features for CVD and statins
have been demonstrated to have effective preventative and
protective properties in RA patients [239].
A recent large population study encountered that
the risk of ACS was increased 23% in the siblings of RA
patients compared to the general population suggesting
a possible shared susceptibility between RA and ACS.
Therefore, inquiring about family history of RA during
health maintenance visits might help detect and address
subclinical CVD risk in patients otherwise considered low
risk [240]. Rheumatology clinics also failed to discuss
hypertension in 2 out of 3 visits, which represent a huge
missed opportunity since 40% of RA patients met criteria
for uncontrolled hypertension. Better strategies are needed
to address modifiable risk factors such as hypertension,
including prompt referral to primary care and patient
education in order to improve outcomes in this vulnerable
patient population [241].
Conclusion
While RA occurs in about 1% of the general
population, CVD disproportionally affects RA patients
and is currently the major cause of morbidity and mortality
among RA patients. CVD presents much earlier in RA
patients and has been reported even before full clinical
presentation and diagnosis of RA. Among the major CVD
presentations, silent MI, diastolic dysfunction, arrythmias,
stroke and sudden cardiac death appear to be prevalent
and occur at a relatively younger age. Other features of
CVD in RA include the obesity paradox and the lipid
paradox making this disease a particularly interesting
and rather challenging entity to diagnose and manage.
Chronic inflammation appears to be the major underlying
pathogenic factor linking RA and CVD. This is associated
with endothelial dysfunction, lipid abnormalities,
hypercoagulability and early atherosclerosis with unstable
plaque formation. Physicians should be cognizant of RA
disease activity as it correlates with higher CVD risk.
The use of DMARDs and biologics have been shown to
control not only RA but also CVD risk. Early detection
and aggressive management of CVD should be employed
in this high-risk population together with control of the
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 13 of 23
traditional CVD risk factors including hypertension,
hyperlipidemia, smoking and diabetes. Further research
is needed to help characterize and establish the ideal
management strategies in this vulnerable population and
inform future guideline development in this field.
Acknowledgment
This work is supported in part by Dr. Moro O.
Salifu's efforts through NIH Grant # S21MD012474
References
1. Sokka T, Abelson B, Pincus T (2008) Mortality in rheumatoid
arthritis: 2008 update. Clin Exp Rheumatol 26: S35-S61.
2. Avina-Zubieta JA, Choi HK, Sadatsafavi M, et al. (2008) Risk
of cardiovascular mortality in patients with rheumatoid arthritis:
a meta-analysis of observational studies. Arthritis Rheum 59:
1690-1697.
3. Avina-Zubieta JA, Thomas J, Sadatsafavi M, et al. (2012) Risk
of incident cardiovascular events in patients with rheumatoid
arthritis: a meta-analysis of observational studies. Ann Rheum
Dis 71: 1524-1529.
4. Pujades-Rodriguez M, Bram Duyx, Sara LT, et al. (2016)
Rheumatoid Arthritis and Incidence of Twelve Initial
Presentations of Cardiovascular Disease: A Population Record-
Linkage Cohort Study in England. PLoS One 11: e0151245.
5. Kang EH, Liao KP, Kim SC (2018) Cardiovascular Safety
of Biologics and JAK Inhibitors in Patients with Rheumatoid
Arthritis. Curr Rheumatol Rep 20: 42.
6. Widdifield JPJ, Huang A, Bernatsky S (2018) Causes of death
in rheumatoid arthritis: How do they compare to the general
population?
7. Zou K, Fu-Kun Xiao, Hong-Ying Li, et al. (2017) Risk of
cardiovascular disease in Chinese patients with rheumatoid
arthritis: A cross-sectional study based on hospital medical
records in 10 years. PLoS One 12: e0180376.
8. Solomon A, Linda Tsang, Angela J. Woodiwiss, et al. (2014)
Cardiovascular disease risk amongst African black patients
with rheumatoid arthritis: the need for population specific
stratification. Biomed Res Int 10.
9. McInnes IB, Schett G (2011) The pathogenesis of rheumatoid
arthritis. N Engl J Med 365: 2205-2219.
10. Libby P, Ridker PM, Maseri A (2002) Inflammation and
atherosclerosis. Circulation 105: 1135-1143.
11. Ridker PM, Rifai N, Rose L, et al. (2002) Comparison of
C-reactive protein and low-density lipoprotein cholesterol levels
in the prediction of first cardiovascular events. N Engl J Med
347: 1557-1565.
12. Gonzalez-Gay MA, Gonzalez-Juanatey C, Lopez-Diaz MJ,
et al. (2007) HLA-DRB1 and persistent chronic inflammation
contribute to cardiovascular events and cardiovascular mortality
in patients with rheumatoid arthritis. Arthritis Rheum 57:
125-132.
13. Sattar N, McInnes IB (2005) Vascular comorbidity in
rheumatoid arthritis: potential mechanisms and solutions. Curr
Opin Rheumatol 17: 286-292.
14. Arend WP (2001) Physiology of cytokine pathways in
rheumatoid arthritis. Arthritis Rheum 45: 101-106.
15. Houssiau FA, Devogelaer JP, Van Damme J, et al. (1988)
Interleukin-6 in synovial fluid and serum of patients with
rheumatoid arthritis and other inflammatory arthritides. Arthritis
Rheum 31: 784-788.
16. Robert M, Miossec P (2017) Effects of Interleukin 17 on the
cardiovascular system. Autoimmun Rev 16: 984-991.
17. Hot A, Lenief V, Miossec P (2012) Combination of IL-17
and TNFalpha induces a pro-inflammatory, pro-coagulant and
pro-thrombotic phenotype in human endothelial cells. Ann
Rheum Dis 71: 768-776.
18. McInnes IB, Schett G (2007) Cytokines in the pathogenesis
of rheumatoid arthritis. Nat Rev Immunol 7: 429-442.
19. Pober JS, Cotran RS (1990) Cytokines and endothelial cell
biology. Physiol Rev 70: 427-451.
20. Fossiez F, Djossou O, Chomarat P, et al. (1996) T cell
interleukin-17 induces stromal cells to produce proinflammatory
and hematopoietic cytokines. J Exp Med 183: 2593-2603.
21. Munro JM, Pober JS, Cotran RS (1989) Tumor necrosis
factor and interferon-gamma induce distinct patterns of
endothelial activation and associated leukocyte accumulation in
skin of Papio anubis. Am J Pathol 135: 121-133.
22. Yuan S, Zhang S, Zhuang Y, et al. (2015) Interleukin-17
Stimulates STAT3-Mediated Endothelial Cell Activation for
Neutrophil Recruitment. Cell Physiol Biochem 36: 2340-2356.
23. Li JH, Pober JS (2005) The cathepsin B death pathway
contributes to TNF plus IFN-gamma-mediated human
endothelial injury. J Immunol 175: 1858-1866.
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 14 of 23
24. Zhu F, Wang Q, Guo C, et al. (2011) IL-17 induces apoptosis
of vascular endothelial cells: a potential mechanism for human
acute coronary syndrome. Clin Immunol 141: 152-160.
25. Bombeli T, Mueller M, Haeberli A (1997) Anticoagulant
properties of the vascular endothelium. Thromb Haemost 77:
408-423.
26. Kitas GD, Gabriel SE (2011) Cardiovascular disease in
rheumatoid arthritis: state of the art and future perspectives. Ann
Rheum Dis 70: 8-14.
27. Aubry MC, Maradit-Kremers H, Reinalda MS, et al. (2007)
Differences in atherosclerotic coronary heart disease between
subjects with and without rheumatoid arthritis. J Rheumatol 34:
937-942.
28. Bevilacqua MP, J S Pober, G R Majeau, et al. (1986)
Recombinant tumor necrosis factor induces procoagulant activity
in cultured human vascular endothelium: characterization and
comparison with the actions of interleukin 1. Proc Natl Acad Sci
USA 83: 4533-4537.
29. Grignani G, Maiolo A (2000) Cytokines and hemostasis.
Haematologica 85: 967-972.
30. Yoshizumi M, Perrella MA, Lee ME, et al. (1993) Tumor
necrosis factor downregulates an endothelial nitric oxide
synthase mRNA by shortening its half-life. Circ Res 73: 205-209.
31. Sohn RH, Deming CB, Johns DC, et al. (2005) Regulation
of endothelial thrombomodulin expression by inflammatory
cytokines is mediated by activation of nuclear factor-kappa B.
Blood 105: 3910-3917.
32. Go AS, Assimes TL, Quertermous T, et al. (2012) Interleukin-6
receptor pathways in coronary heart disease: a collaborative
meta-analysis of 82 studies. Lancet 379: 1205-1213.
33. Hurlimann D, Forster A, Noll G, et al. (2002) Anti-tumor
necrosis factor-alpha treatment improves endothelial function in
patients with rheumatoid arthritis. Circulation 106: 2184-2187.
34. Ridker PM (2017) Canakinumab for Residual Inflammatory
Risk. Eur Heart J 38: 3545-3548.
35. Kleveland O, Kunszt G, Bratlie M, et al. (2016) Effect of a
single dose of the interleukin-6 receptor antagonist tocilizumab
on inflammation and troponin T release in patients with non-
ST-elevation myocardial infarction: a double blind, randomized,
placebo-controlled phase 2 trial. Eur Heart J 37: 2406-2413.
36. Zuniga LA, Shen WJ, Joyce-Shaikh B, et al. (2010) IL-17
regulates adipogenesis, glucose homeostasis, and obesity. J
Immunol 185: 6947-6959.
37. Liao KP, Solomon DH (2013) Traditional cardiovascular
risk factors, inflammation and cardiovascular risk in rheumatoid
arthritis. Rheumatology 52: 45-52.
38. Su SA, Yang D, Zhu W, et al. (2016) Interleukin-17A mediates
cardiomyocyte apoptosis through Stat3-iNOS pathway. Biochim
Biophys Acta 1863: 2784-2794.
39. Zhou SF, Yuan J, Liao MY, et al. (2014) IL-17A promotes
ventricular remodeling after myocardial infarction. J Mol Med
92: 1105-1116.
40. Wu L, SuFey Ong, Monica V. Talor, et al. (2014) Cardiac
fibroblasts mediate IL-17A-driven inflammatory dilated
cardiomyopathy. J Exp Med 211: 1449-1464.
41. Arosio E, De Marchi S, Rigoni A, et al. (2007) Forearm
haemodynamics, arterial stiffness and microcirculatory reactivity
in rheumatoid arthritis. J Hypertens 25: 1273-1278.
42. Datta D, Ferrell WR, Sturrock RD, et al. (2007) Inflammatory
suppression rapidly attenuates microvascular dysfunction in
rheumatoid arthritis. Atherosclerosis 192: 391-395.
43. Galarraga B, Khan F, Kumar P, et al. (2008) C-reactive
protein: the underlying cause of microvascular dysfunction in
rheumatoid arthritis. Rheumatology 47: 1780-1784.
44. Ciftci O, Yilmaz S, Topcu S, et al. (2008) Impaired coronary
microvascular function and increased intima-media thickness in
rheumatoid arthritis. Atherosclerosis 198: 332-337.
45. Maradit-Kremers H, Crowson CS, Nicola PJ, et al. (2005)
Increased unrecognized coronary heart disease and sudden
deaths in rheumatoid arthritis: a population-based cohort study.
Arthritis Rheum 52: 402-411.
46. Lee YH, Choi SJ, Ji JD, et al, (2000) Lipoprotein (a) and
lipids in relation to inflammation in rheumatoid arthritis. Clin
Rheumatol 19: 324-325.
47. Van Lenten BJ, Hama SY, de Beer FC, et al. (1995) Anti-
inflammatory HDL becomes pro-inflammatory during the acute
phase response. J Clin Invest 96: 2758-2767.
48. Navab M, Hama SY, Hough GP, et al. (2001) A cell-free
assay for detecting HDL that is dysfunctional in preventing the
formation of or inactivating oxidized phospholipids. J Lipid Res
42: 1308-1317.
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 15 of 23
49. Navab M, Van Lenten BJ, Srinivasa Reddy T, et al. (2001)
High-density lipoprotein and the dynamics of atherosclerotic
lesions. Circulation 104: 2386-2387.
50. McMahon M, Grossman J, FitzGerald J, et al. (2006)
Proinflammatory high-density lipoprotein as a biomarker for
atherosclerosis in patients with systemic lupus erythematosus
and rheumatoid arthritis. Arthritis Rheum 54: 2541-2549.
51. Watanabe J, Charles-Schoeman C, Miao Y, et al. (2012)
Proteomic profiling following immunoaffinity capture of
high-density lipoprotein: association of acute-phase proteins
and complement factors with proinflammatory high-density
lipoprotein in rheumatoid arthritis. Arthritis Rheum 64:
1828-1837.
52. Charles-Schoeman C, Watanabe J, Lee YY, et al. (2009)
Abnormal function of high-density lipoprotein is associated with
poor disease control and an altered protein cargo in rheumatoid
arthritis. Arthritis Rheum 60: 2870-2879.
53. McInnes IB, Thompson L, Giles JT, et al. (2015) Effect of
interleukin-6 receptor blockade on surrogates of vascular risk
in rheumatoid arthritis: MEASURE, a randomised, placebo-
controlled study. Ann Rheum Dis 74: 694-702.
54. Liao KP, Playford MP, Michelle F, et al. (2015) The
association between reduction in inflammation and changes
in lipoprotein levels and HDL cholesterol efflux capacity in
rheumatoid arthritis. J Am Heart Assoc 4.
55. Navarro-Millan I, Charles-Schoeman C, Yang S, et al.
(2013) Changes in lipoproteins associated with methotrexate or
combination therapy in early rheumatoid arthritis: results from
the treatment of early rheumatoid arthritis trial. Arthritis Rheum
65: 1430-1438.
56. Pollono EN, Lopez-Olivo MA, Lopez JA, et al. (2010) A
systematic review of the effect of TNF-alpha antagonists on lipid
profiles in patients with rheumatoid arthritis. Clin Rheumatol
29: 947-955.
57. Kremer JM, Bloom BJ, Breedveld FC, et al. (2009) The
safety and efficacy of a JAK inhibitor in patients with active
rheumatoid arthritis: Results of a double blind, placebo-
controlled phase IIa trial of three dosage levels of CP-690,550
versus placebo. Arthritis Rheum 60: 1895-1905.
58. Semb AG, Rollefstad S, Provan SA, et al. (2013) Carotid
plaque characteristics and disease activity in rheumatoid
arthritis. J Rheumatol 40: 359-368.
59. McEntegart A, Capell HA, Creran D, et al. (2001)
Cardiovascular risk factors, including thrombotic variables,
in a population with rheumatoid arthritis. Rheumatology 40:
640-644.
60. Schonbeck U, Libby P (2001) CD40 signaling and plaque
instability. Circ Res 89: 1092-1103.
61. Schonbeck U, Varo N, Libby P, et al. (2001) Soluble CD40L
and cardiovascular risk in women. Circulation 104: 2266-2268.
62. Vasan RS (2006) Biomarkers of cardiovascular disease:
molecular basis and practical considerations. Circulation 113:
2335-2362.
63. Vallejo AN (2005) CD28 extinction in human T cells: altered
functions and the program of T-cell senescence. Immunol Rev
205: 158-169.
64. Liuzzo G, Kopecky SL, Frye RL, et al. (1999) Perturbation of
the T-cell repertoire in patients with unstable angina. Circulation
100: 2135-2139.
65. Pasceri V, Yeh ET (1999) A tale of two diseases: atherosclerosis
and rheumatoid arthritis. Circulation 100: 2124-2126.
66. Eid RE, Rao DA, Zhou J, et al. (2009) Interleukin-17 and
interferon-gamma are produced concomitantly by human
coronary artery-infiltrating T cells and act synergistically on
vascular smooth muscle cells. Circulation 119: 1424-1432.
67. El-Menoufy MA, EK, Ahmed, et al. (2018) Unusual
CD4+CD28- T lymphocyte subset is implicated in the
pathogenesis of early atherosclerosis in patients with rheumatoid
arthritis. The Egyptian Rheumatologist.
68. Gerli R, Schillaci G, Giordano A, et al. (2004) CD4+CD28-
T lymphocytes contribute to early atherosclerotic damage in
rheumatoid arthritis patients. Circulation 109: 2744-2748.
69. Pober JS, Sessa WC (2007) Evolving functions of endothelial
cells in inflammation. Nat Rev Immunol 7: 803-815.
70. Arts EE, Popa C, Den Broeder AA, et al. (2015) Performance
of four current risk algorithms in predicting cardiovascular
events in patients with early rheumatoid arthritis. Ann Rheum
Dis 74: 668-674.
71. Crowson CS, Rollefstad S, Ikdahl E, et al. (2018) Impact of
risk factors associated with cardiovascular outcomes in patients
with rheumatoid arthritis. Ann Rheum Dis 7: 48-54.
72. Crowson CS, Gabriel SE, Semb AG, et al. (2017) Rheumatoid
arthritis-specific cardiovascular risk scores are not superior to
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 16 of 23
general risk scores: a validation analysis of patients from seven
countries. Rheumatology 56: 1102-1110.
73. Crowson CS, Rollefstad S, Kitas GD, et al. (2017) Challenges
of developing a cardiovascular risk calculator for patients with
rheumatoid arthritis. PLoS One 12: e0174656.
74. Crowson CS, Rollefstad S, Kitas GD, et al. (2017) Correction:
Challenges of developing a cardiovascular risk calculator for
patients with rheumatoid arthritis. PLoS One 12: e0175605.
75. Peters MJ, Symmons DP, McCarey D, et al. (2010) EULAR
evidence-based recommendations for cardiovascular risk
management in patients with rheumatoid arthritis and other
forms of inflammatory arthritis. Ann Rheum Dis 69: 325-331.
76. Hippisley-Cox J, Vinogradova Y, Minhas R, et al. (2008)
Predicting cardiovascular risk in England and Wales: prospective
derivation and validation of QRISK2. BMJ 336: 1475-1482.
77. Solomon DH, Greenberg J, Curtis JR, et al. (2015) Derivation
and internal validation of an expanded cardiovascular risk
prediction score for rheumatoid arthritis: a Consortium of
Rheumatology Researchers of North America Registry Study.
Arthritis Rheumatol 67: 1995-2003.
78. Stavropoulos-Kalinoglou A, Metsios GS, Panoulas VF, et
al. (2009) Associations of obesity with modifiable risk factors
for the development of cardiovascular disease in patients with
rheumatoid arthritis. Ann Rheum Dis 68: 242-245.
79. Armstrong DJ, McCausland EM, Quinn AD, et al. (2006)
Obesity and cardiovascular risk factors in rheumatoid arthritis.
Rheumatology 45: 782-783.
80. Sparks JA, Chang SC, Nguyen US, et al. (2018) Weight
Change During the Early Rheumatoid Arthritis Period and Risk
of Subsequent Mortality in Women With Rheumatoid Arthritis
and Matched Comparators. Arthritis Rheumatol 70: 18-29.
81. Baker JF, Billig E , Michaud K, et al. (2015) Weight Loss, the
Obesity Paradox, and the Risk of Death in Rheumatoid Arthritis.
Arthritis Rheumatol 67: 1711-1717.
82. Kremers HM, Nicola PJ, Crowson CS, et al. (2004)
Prognostic importance of low body mass index in relation to
cardiovascular mortality in rheumatoid arthritis. Arthritis Rheum
50: 3450-3457.
83. Wolfe F, Michaud k (2012) Effect of body mass index on
mortality and clinical status in rheumatoid arthritis. Arthritis
Care Res 64: 1471-1479.
84. Gonzalez A, Maradit Kremers H, Crowson CS, et al. (2007)
The widening mortality gap between rheumatoid arthritis patients
and the general population. Arthritis Rheum 56: 3583-3587.
85. Roubenoff R, Roubenoff RA, Ward LM, et al. (1992)
Rheumatoid cachexia: depletion of lean body mass in rheumatoid
arthritis. J Rheumatol 19: 1505-1510.
86. Engvall IL, Elkan AC, Tengstrand B, et al. (2008) Cachexia
in rheumatoid arthritis is associated with inflammatory activity,
physical disability, and low bioavailable insulin-like growth
factor. Scand J Rheumatol 37: 321-328.
87. Escalante A, Haas RW, Rincon I del (2005) Paradoxical
effect of body mass index on survival in rheumatoid arthritis:
role of comorbidity and systemic inflammation. Arch Intern
Med 165: 1624-1629.
88. Stavropoulos-Kalinoglou A, Metsios GS, Koutedakis Y,
Nevill AM, et al. (2007) Redefining overweight and obesity in
rheumatoid arthritis patients. Ann Rheum Dis 66: 1316-1321.
89. Dessein PH, Norton GR, Woodiwiss AJ, et al. (2007)
Independent role of conventional cardiovascular risk factors as
predictors of C-reactive protein concentrations in rheumatoid
arthritis. J Rheumatol 34: 681-688.
90. Liao KP, Liu J, Lu B, et al. (2015) Association between lipid
levels and major adverse cardiovascular events in rheumatoid
arthritis compared to non-rheumatoid arthritis patients. Arthritis
Rheumatol 67: 2004-2010.
91. Sniderman AD, Islam S, Yusuf S, et al. (2012) Discordance
analysis of apolipoprotein B and non-high density lipoprotein
cholesterol as markers of cardiovascular risk in the
INTERHEART study. Atherosclerosis 225: 444-449.
92. Pencina MJ, D’Agostino RB, Zdrojewski T, et al. (2015)
Apolipoprotein B improves risk assessment of future coronary
heart disease in the Framingham Heart Study beyond LDL-C
and non-HDL-C. Eur J Prev Cardiol 22: 1321-1327.
93. Lawler PR, Akinkuolie AO, Ridker PM, et al. (2017)
Discordance between Circulating Atherogenic Cholesterol Mass
and Lipoprotein Particle Concentration in Relation to Future
Coronary Events in Women. Clin Chem 63: 870-879.
94. Otvos JD, Mora S, Shalaurova I, et al. (2011) Clinical
implications of discordance between low-density lipoprotein
cholesterol and particle number. J Clin Lipidol 5: 105-113.
95. Mackey RH, Kuller LH, Moreland LW (2018) Update
on Cardiovascular Disease Risk in Patients with Rheumatic
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 17 of 23
Diseases. Rheum Dis Clin North Am 44: 475-487.
96. Mackey RH, Greenland P, Lloyd-Jones D, et al. (2012)
High-density lipoprotein cholesterol and particle concentrations,
carotid atherosclerosis, and coronary events: MESA (multi-
ethnic study of atherosclerosis). J Am Coll Cardiol 60: 508-516.
97. Chung CP, Oeser A, Solus JF, et al. (2008) Inflammation-
associated insulin resistance: differential effects in rheumatoid
arthritis and systemic lupus erythematosus define potential
mechanisms. Arthritis Rheum 58: 2105-2112.
98. Costenbader KH, Feskanich D, Mandl LA, et al. (2006)
Smoking intensity, duration, and cessation, and the risk of
rheumatoid arthritis in women. Am J Med 119: e1-e9.
99. Gonzalez-Gay MA, Garcia-Porrua C, Hajeer AH (2002)
Influence of human leukocyte antigen-DRB1 on the susceptibility
and severity of rheumatoid arthritis. Semin Arthritis Rheum 31:
355-360.
100. Gonzalez-Gay MA, Gonzalez-Juanatey C, Ollier WE
(2004) Endothelial dysfunction in rheumatoid arthritis: influence
of HLA-DRB1 alleles. Autoimmun Rev 3: 301-304.
101. Ollier W, Venables PJ, Mumford PA, et al. (1984) HLA
antigen associations with extra-articular rheumatoid arthritis.
Tissue Antigens 24: 279-291.
102. Gonzalez-Juanatey C, Testa A, Garcia-Castelo A, et al.
(2003) HLA-DRB1 status affects endothelial function in treated
patients with rheumatoid arthritis. Am J Med 114: 647-652.
103. Salvarani C, Macchioni P, Mantovani W, et al. (1992)
Extraarticular manifestations of rheumatoid arthritis and HLA
antigens in northern Italy. J Rheumatol 19: 242-246.
104. Weyand CM, Hicok KC, Conn DL, et al. (1992) The
influence of HLA-DRB1 genes on disease severity in rheumatoid
arthritis. Ann Intern Med 117: 801-806.
105. Perdriger A, Chalès G, Semana G, et al. (1997) Role of
HLA-DR-DR and DR-DQ associations in the expression
of extraarticular manifestations and rheumatoid factor in
rheumatoid arthritis. J Rheumatol 24: 1272-1276.
106. Panoulas VF, Stavropoulos-Kalinoglou A, Metsios GS,
et al. (2009) Association of interleukin-6 (IL-6)-174G/C
gene polymorphism with cardiovascular disease in patients
with rheumatoid arthritis: the role of obesity and smoking.
Atherosclerosis 204: 178-183.
107. Solomon DH, Reed GW, Kremer JM, et al. (2015) Disease
activity in rheumatoid arthritis and the risk of cardiovascular
events. Arthritis Rheumatol 67: 1449-1455.
108. Innala L, Bozena Möller, Lotta Ljung, et al. (2011)
Cardiovascular events in early RA are a result of inflammatory
burden and traditional risk factors: a five year prospective study.
Arthritis Res Ther 13: 131.
109. Mackey RH, Kuller LH, Deane KD, et al. (2015) Rheumatoid
Arthritis, Anti-Cyclic Citrullinated Peptide Positivity, and
Cardiovascular Disease Risk in the Women’s Health Initiative.
Arthritis Rheumatol 67: 2311-2322.
110. Turesson C, McClelland RL, Christianson TJ, et al. (2007)
Severe extra-articular disease manifestations are associated with
an increased risk of first ever cardiovascular events in patients
with rheumatoid arthritis. Ann Rheum Dis 66: 70-75.
111. Rao VU, Pavlov A, Klearman M, et al. (2015) An evaluation
of risk factors for major adverse cardiovascular events during
tocilizumab therapy. Arthritis Rheumatol 67: 372-380.
112. Myasoedova E, Chandran A, Ilhan B, et al. (2016) The role
of rheumatoid arthritis (RA) flare and cumulative burden of RA
severity in the risk of cardiovascular disease. Ann Rheum Dis
75: 560-565.
113. Farragher TM, Lunt M, Bunn DK, et al. (2007) Early
functional disability predicts both all-cause and cardiovascular
mortality in people with inflammatory polyarthritis: results from
the Norfolk Arthritis Register. Ann Rheum Dis 66: 486-492.
114. Giles JT, Moyses S, Wendy P, et al. (2009) Coronary arterial
calcification in rheumatoid arthritis: comparison with the Multi-
Ethnic Study of Atherosclerosis. Arthritis Res Ther 11: 36.
115. Evans MR, et al. (2011) Carotid atherosclerosis predicts
incident acute coronary syndromes in rheumatoid arthritis.
Arthritis Rheum 63: 1211-1220.
116. Mantel A, Escalante A, Battafarano DF, et al. (2015)
Rheumatoid arthritis is associated with a more severe
presentation of acute coronary syndrome and worse short-term
outcome. Eur Heart J 36: 3413-3422.
117. Holmqvist ME, Wedrén S, Jacobsson LT, et al. (2009)
No increased occurrence of ischemic heart disease prior to
the onset of rheumatoid arthritis: results from two Swedish
population-based rheumatoid arthritis cohorts. Arthritis Rheum
60: 2861-2869.
118. Holmqvist ME, Neovius M, Eriksson J, et al. (2012) Risk of
venous thromboembolism in patients with rheumatoid arthritis
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 18 of 23
and association with disease duration and hospitalization. JAMA
308: 1350-1356.
119. Holmqvist ME, Wedrén S, Jacobsson LT, et al. (2010)
Rapid increase in myocardial infarction risk following diagnosis
of rheumatoid arthritis amongst patients diagnosed between
1995 and 2006. J Intern Med 268: 578-585.
120. Mantel A, Holmqvist M, Nyberg F, et al. (2015) Risk factors
for the rapid increase in risk of acute coronary events in patients
with new-onset rheumatoid arthritis: a nested case-control study.
Arthritis Rheumatol 67: 2845-2854.
121. Holmqvist M, Gränsmark E, Mantel A, et al. (2013)
Occurrence and relative risk of stroke in incident and prevalent
contemporary rheumatoid arthritis. Ann Rheum Dis 72: 541-546.
122. Wallberg-Jonsson S, Johansson H, Ohman ML, et al.
(1999) Extent of inflammation predicts cardiovascular disease
and overall mortality in seropositive rheumatoid arthritis. J
Rheumatol 26: 2562-2571.
123. Jacobsson LT, Turesson C, Hanson RL, et al. (2001) Joint
swelling as a predictor of death from cardiovascular disease
in a population study of Pima Indians. Arthritis Rheum 44:
1170-1176.
124. Meissner Y, Zink A, Kekow J, et al. (2016) Impact of
disease activity and treatment of comorbidities on the risk of
myocardial infarction in rheumatoid arthritis. Arthritis Res Ther
18: 183.
125. Ruyssen-Witrand A, Bruno F, Alain S et al. (2011)
Cardiovascular risk induced by low-dose corticosteroids in
rheumatoid arthritis: a systematic literature review. Joint Bone
Spine 78: 23-30.
126. Ajeganova S, Andersson ML, Frostegård J, et al. (2013)
Disease factors in early rheumatoid arthritis are associated
with differential risks for cardiovascular events and mortality
depending on age at onset: a 10-year observational cohort study.
J Rheumatol 40: 1958-1966.
127. Maradit-Kremers H, Nicola PJ, Crowson CS, et al. (2007)
Raised erythrocyte sedimentation rate signals heart failure in
patients with rheumatoid arthritis. Ann Rheum Dis 66: 76-80.
128. Goodson NJ, Symmons DP, Scott DG, et al. (2005)
Baseline levels of C-reactive protein and prediction of death
from cardiovascular disease in patients with inflammatory
polyarthritis: a ten-year follow up study of a primary care-based
inception cohort. Arthritis Rheum 52: 2293-2299.
129. Ridker PM, Hennekens CH, Buring JE, et al. (2000)
C-reactive protein and other markers of inflammation in the
prediction of cardiovascular disease in women. N Engl J Med
342: 836-843.
130. Zhang J, Rheum AD, Myasoedova E, et al. (2014)
Republished: The association between inflammatory markers,
serum lipids and the risk of cardiovascular events in patients
with rheumatoid arthritis. Postgrad Med J 90: 722-729.
131. Myasoedova E, Crowson CS, Kremers HM, et al. (2011)
Lipid paradox in rheumatoid arthritis: the impact of serum
lipid measures and systemic inflammation on the risk of
cardiovascular disease. Ann Rheum Dis 70: 482-487.
132. Singh U, Dasu MR, Yancey PG, et al. (2008) Human
C-reactive protein promotes oxidized low density lipoprotein
uptake and matrix metalloproteinase-9 release in Wistar rats. J
Lipid Res 49: 1015-1023.
133. Wang X, Liao D, Bhardwaj U, et al. (2008) C-reactive
protein inhibits cholesterol efflux from human macrophage-
derived foam cells. Arterioscler Thromb Vasc Biol 28: 519-526.
134. Wolfe F, Mitchell DM, Sibley JT, et al. (1994) The mortality
of rheumatoid arthritis. Arthritis Rheum 37: 481-494.
135. Goodson NJ, Wiles NJ, Lunt M, et al. (2002) Mortality
in early inflammatory polyarthritis: cardiovascular mortality
is increased in seropositive patients. Arthritis Rheum 46:
2010-2019.
136. Gonzalez A, Maradit Kremers H, Crowson CS, et al.
(2008) Do cardiovascular risk factors confer the same risk for
cardiovascular outcomes in rheumatoid arthritis patients as in
non-rheumatoid arthritis patients? Ann Rheum Dis 67: 64-69.
137. Liang KP, Myasoedova E, Crowson CS, et al. (2010)
Increased prevalence of diastolic dysfunction in rheumatoid
arthritis. Ann Rheum Dis 69: 1665-1670.
138. Wolfe F (2000) The effect of smoking on clinical, laboratory,
and radiographic status in rheumatoid arthritis. J Rheumatol 27:
630-637.
139. Tomasson G, Aspelund T, Jonsson T, et al. (2010) Effect of
rheumatoid factor on mortality and coronary heart disease. Ann
Rheum Dis 69: 1649-654.
140. Liang KP, Kremers HM, Crowson CS, et al. (2009)
Autoantibodies and the risk of cardiovascular events. J
Rheumatol 36: 2462-2469.
141. Humphreys JH, Van Nies JA, Chipping J, et al. (2014)
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 19 of 23
Rheumatoid factor and anti-citrullinated protein antibody
positivity, but not level, are associated with increased mortality
in patients with rheumatoid arthritis: results from two large
independent cohorts. Arthritis Res Ther 16: 483.
142. Sokolove J, Brennan MJ, Sharpe O, et al. (2013) Brief report:
citrullination within the atherosclerotic plaque: a potential target
for the anti-citrullinated protein antibody response in rheumatoid
arthritis. Arthritis Rheum 65: 1719-1724.
143. Lopez-Longo FJ, Oliver-Miñarro D, Sánchez-Ramón
S, et al. (2009) Association between anti-cyclic citrullinated
peptide antibodies and ischemic heart disease in patients with
rheumatoid arthritis. Arthritis Rheum 61: 419-424.
144. Ridker PM, Rifai N,e Stampfer MJ, et al. (2000) Plasma
concentration of interleukin-6 and the risk of future myocardial
infarction among apparently healthy men. Circulation 101:
1767-1772.
145. Danesh J, Kaptoge S, Mann AG, et al. (2008) Long-term
interleukin-6 levels and subsequent risk of coronary heart
disease: two new prospective studies and a systematic review.
PLoS Med 5: e78.
146. Rho YH, Chung CP, Oeser A, et al. (2009) Inflammatory
mediators and premature coronary atherosclerosis in rheumatoid
arthritis. Arthritis Rheum 61: 1580-1585.
147. Dessein PH, Joffe BI, Singh S (2005) Biomarkers of
endothelial dysfunction, cardiovascular risk factors and
atherosclerosis in rheumatoid arthritis. Arthritis Res Ther 7:
634-643.
148. Karpouzas GA, Estis J, Rezaeian P, et al. (2018) High-
sensitivity cardiac troponin I is a biomarker for occult coronary
plaque burden and cardiovascular events in patients with
rheumatoid arthritis. Rheumatology 57: 1080-1088.
149. Zethelius B, Berglund L, Sundström J, et al. (2008) Use
of multiple biomarkers to improve the prediction of death from
cardiovascular causes. N Engl J Med 358: 2107-2116.
150. Avouac J, Meune C, Chenevier-Gobeaux C, et al. (2014)
Inflammation and disease activity are associated with high
circulating cardiac markers in rheumatoid arthritis independently
of traditional cardiovascular risk factors. J Rheumatol 41:
248-255.
151. Mirjafari H, Welsh P, Verstappen SM, et al. (2014)
N-terminal pro-brain-type natriuretic peptide (NT-pro-BNP)
and mortality risk in early inflammatory polyarthritis: results
from the Norfolk Arthritis Registry (NOAR). Ann Rheum Dis
73: 684-690.
152. Provan S, Angel K, Semb AG, et al. (2010) NT-proBNP
predicts mortality in patients with rheumatoid arthritis: results
from 10-year follow-up of the EURIDISS study. Ann Rheum
Dis 69: 1946-1950.
153. Solomon DH, Goodson NJ, Katz JN, et al. (2006) Patterns
of cardiovascular risk in rheumatoid arthritis. Ann Rheum Dis
65: 1608-1612.
154. Lindhardsen J, Ahlehoff O, Gislason GH, et al. (2011) The
risk of myocardial infarction in rheumatoid arthritis and diabetes
mellitus: a Danish nationwide cohort study. Ann Rheum Dis 70:
929-934.
155. Solomon DH, Karlson EW, Rimm EB, et al. (2003)
Cardiovascular morbidity and mortality in women diagnosed
with rheumatoid arthritis. Circulation 107: 1303-1307.
156. Meune C, Touzé E, Trinquart L, et al. (2010) High risk of
clinical cardiovascular events in rheumatoid arthritis: Levels
of associations of myocardial infarction and stroke through a
systematic review and meta-analysis. Arch Cardiovasc Dis 103:
253-261.
157. Karpouzas GA, Malpeso J, Choi TY, et al. (2014) Prevalence,
extent and composition of coronary plaque in patients with
rheumatoid arthritis without symptoms or prior diagnosis of
coronary artery disease. Ann Rheum Dis 73: 1797-1804.
158. Crepaldi G, Alberto Scirè C, Carrara G, et al. (2016)
Cardiovascular Comorbidities Relate More than Others with
Disease Activity in Rheumatoid Arthritis. PLoS One 11:
e0146991.
159. Van Doornum S, Brand C, King B, et al. (2006) Increased
case fatality rates following a first acute cardiovascular event
in patients with rheumatoid arthritis. Arthritis Rheum 54:
2061-2068.
160. Van Doornum S, Caroline B, Sundararajan V, et al. (2010)
Rheumatoid arthritis patients receive less frequent acute
reperfusion and secondary prevention therapy after myocardial
infarction compared with the general population. Arthritis Res
Ther 12: 183.
161. McCoy SS, Crowson CS, Maradit-Kremers H, et al. (2013)
Longterm outcomes and treatment after myocardial infarction
in patients with rheumatoid arthritis. J Rheumatol 40: 605-610.
162. Nicola PJ, Crowson CS, Maradit-Kremers H, et al. (2006)
Contribution of congestive heart failure and ischemic heart
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 20 of 23
disease to excess mortality in rheumatoid arthritis. Arthritis
Rheum 54: 60-67.
163. Nicola PJ, Maradit-Kremers H, Roger VL, et al. (2005)
The risk of congestive heart failure in rheumatoid arthritis:
a population-based study over 46 years. Arthritis Rheum 52:
412-420.
164. Crowson CS, Nicola PJ, Kremers HM, et al. (2005) How
much of the increased incidence of heart failure in rheumatoid
arthritis is attributable to traditional cardiovascular risk factors
and ischemic heart disease? Arthritis Rheum 52: 3039-3044.
165. Wolfe F, Michaud K (2004) Heart failure in rheumatoid
arthritis: rates, predictors, and the effect of anti-tumor necrosis
factor therapy. Am J Med 116: 305-311.
166. Schau T, Gottwald M, Arbach O, et al. (2015) Increased
Prevalence of Diastolic Heart Failure in Patients with
Rheumatoid Arthritis Correlates with Active Disease, but Not
with Treatment Type. J Rheumatol 42: 2029-2037.
167. Khalid U, Egeberg A, Ahlehoff O, et al. (2018) Incident
Heart Failure in Patients with Rheumatoid Arthritis: A
Nationwide Cohort Study. J Am Heart Assoc 7.
168. Logstrup BB, Ellingsen T, Pedersen AB, et al. (2018)
Development of heart failure in patients with rheumatoid
arthritis: A Danish population-based study. Eur J Clin Invest 48:
e12915.
169. Mantel A, Holmqvist M, Jernberg T, et al. (2017) Long-
term outcomes and secondary prevention after acute coronary
events in patients with rheumatoid arthritis. Ann Rheum Dis 76:
2017-2024.
170. Gonzalez-Juanatey C, Testa A, Garcia-Castelo A, et al.
(2004) Echocardiographic and Doppler findings in long-term
treated rheumatoid arthritis patients without clinically evident
cardiovascular disease. Semin Arthritis Rheum 33: 231-238.
171. Myasoedova E, Crowson CS, Roger VL, et al. (2013) Brief
report: rheumatoid arthritis is associated with left ventricular
concentric remodeling: results of a population-based cross-
sectional study. Arthritis Rheum 65: 1713-1718.
172. Giles JT, Malayeri AA, Fernandes V, et al. (2010) Left
ventricular structure and function in patients with rheumatoid
arthritis, as assessed by cardiac magnetic resonance imaging.
Arthritis Rheum 62: 940-951.
173. Midtbo H, Gerdts E, Kvien TK, et al. (2015) Disease
activity and left ventricular structure in patients with rheumatoid
arthritis. Rheumatology 54: 511-519.
174. Midtbo H, Semb AG, Matre K, et al. (2017) Disease activity
is associated with reduced left ventricular systolic myocardial
function in patients with rheumatoid arthritis. Ann Rheum Dis
76: 371-376.
175. Cioffi G, Viapiana O, Ognibeni F, et al. (2016) Combined
Circumferential and Longitudinal Left Ventricular Systolic
Dysfunction in Patients with Rheumatoid Arthritis without
Overt Cardiac Disease. J Am Soc Echocardiogr 29: 689-698.
176. Davis JM, Roger VL, Crowson CS, et al. (2008) The
presentation and outcome of heart failure in patients with
rheumatoid arthritis differs from that in the general population.
Arthritis Rheum 58: 2603-2611.
177. Lindhardsen J, Rintek Madsen O, Torp-Pedersen C, et
al. (2012) Risk of atrial fibrillation and stroke in rheumatoid
arthritis: Danish nationwide cohort study. BMJ 344: e1257.
178. Mavrogeni S, Spargias K, Markussis V, et al. (2009)
Myocardial inflammation in autoimmune diseases: investigation
by cardiovascular magnetic resonance and endomyocardial
biopsy. Inflamm Allergy Drug Targets 8: 390-397.
179. Peters MJ, van Halm VP, Voskuyl AE, et al. (2009) Does
rheumatoid arthritis equal diabetes mellitus as an independent
risk factor for cardiovascular disease? A prospective study.
Arthritis Rheum 61: 1571-1579.
180. Conroy RM, Pyörälä K, Fitzgerald AP, et al. (2003)
Estimation of ten-year risk of fatal cardiovascular disease in
Europe: the SCORE project. Eur Heart J 24: 987-1003.
181. D’Agostino RB, Vasan RS, Pencina MJ, et al. (2008)
General cardiovascular risk profile for use in primary care: the
Framingham Heart Study. Circulation 117: 743-753.
182. Arts EE, Popa CD, Den Broeder AA, et al. (2016) Prediction
of cardiovascular risk in rheumatoid arthritis: performance of
original and adapted SCORE algorithms. Ann Rheum Dis 75:
674-680.
183. Corrales A, González-Juanatey C, Peiró ME, et al.
(2014) Carotid ultrasound is useful for the cardiovascular risk
stratification of patients with rheumatoid arthritis: results of a
population-based study. Ann Rheum Dis 73: 722-727.
184. Corrales A, Parra JA, González-Juanatey C, et al. (2013)
Cardiovascular risk stratification in rheumatic diseases: carotid
ultrasound is more sensitive than Coronary Artery Calcification
Score to detect subclinical atherosclerosis in patients with
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 21 of 23
rheumatoid arthritis. Ann Rheum Dis 72: 1764-1770.
185. de Campos, de Magalhães Souza Fialho SC, Nazário NO,
et al. (2016) Assessment of cardiovascular risk in patients with
rheumatoid arthritis using the SCORE risk index. Rev Bras
Reumatol Engl Ed 5: 138-144.
186. Agca R, Heslinga SC, Rollefstad S, et al. (2017) EULAR
recommendations for cardiovascular disease risk management
in patients with rheumatoid arthritis and other forms of
inflammatory joint disorders: 2015/2016 update. Ann Rheum
Dis 76: 17-28.
187. Kozera L, Andrews J, Morgan AW (2011) Cardiovascular
risk and rheumatoid arthritis the next step: differentiating
true soluble biomarkers of cardiovascular risk from surrogate
measures of inflammation. Rheumatology 50: 1944-1954.
188. Ridker PM, Cook N (2004) Clinical usefulness of very high
and very low levels of C-reactive protein across the full range of
Framingham Risk Scores. Circulation 109: 1955-1959.
189. Ridker PM, Buring JE, Rifai N, et al. (2007) Development
and validation of improved algorithms for the assessment of
global cardiovascular risk in women: the Reynolds Risk Score.
JAMA 297: 611-619.
190. Crowson CS, Matteson EL, Roger VL, et al. (2012)
Usefulness of risk scores to estimate the risk of cardiovascular
disease in patients with rheumatoid arthritis. Am J Cardiol 110:
420-424.
191. Fent GJ, Greenwood JP, Plein S, et al. (2017) The role
of non-invasive cardiovascular imaging in the assessment of
cardiovascular risk in rheumatoid arthritis: where we are and
where we need to be. Ann Rheum Dis 76: 1169-1175.
192. Matsuzawa Y, Lerman A (2014) Endothelial dysfunction
and coronary artery disease: assessment, prognosis, and
treatment. Coron Artery Dis 25: 713-724.
193. Kullo IJ, Malik AR (2007) Arterial ultrasonography and
tonometry as adjuncts to cardiovascular risk stratification. J Am
Coll Cardiol 49: 1413-1426.
194. Kullo IJ, Malik AR, Bielak LF, et al. (2007) Brachial artery
diameter and vasodilator response to nitroglycerine, but not
flow-mediated dilatation, are associated with the presence and
quantity of coronary artery calcium in asymptomatic adults. Clin
Sci 112: 175-182.
195. Scanlon EM, Mankad R, Crowson CS, et al. (2017)
Cardiovascular risk assessment in patients with rheumatoid
arthritis: a correlative study of noninvasive arterial health
testing. Clin Rheumatol 36: 763-771.
196. Di Minno MN, Ambrosino P, Lupoli R, et al. (2015) Clinical
assessment of endothelial function in patients with rheumatoid
arthritis: A meta-analysis of literature studies. Eur J Intern Med
26: 835-842.
197. Ambrosino P, Tasso M, Lupoli R, et al. (2015) Non-invasive
assessment of arterial stiffness in patients with rheumatoid
arthritis: a systematic review and meta-analysis of literature
studies. Ann Med 47: 457-467.
198. Ikdahl E, Rollefstad S, Wibetoe G, et al. (2016)
Predictive Value of Arterial Stiffness and Subclinical Carotid
Atherosclerosis for Cardiovascular Disease in Patients with
Rheumatoid Arthritis. J Rheumatol 43: 1622-1630.
199. Polak JF, Szklo M, Kronmal RA, et al. (2013) The value
of carotid artery plaque and intima-media thickness for incident
cardiovascular disease: the multi-ethnic study of atherosclerosis.
J Am Heart Assoc 2: e000087.
200. Gonzalez-Juanatey C, Llorca J, Martin J, et al. (2009)
Carotid intima-media thickness predicts the development of
cardiovascular events in patients with rheumatoid arthritis.
Semin Arthritis Rheum 38: 366-371.
201. Ajeganova S, de Faire U, Jogestrand T, et al. (2012)
Carotid atherosclerosis, disease measures, oxidized low-
density lipoproteins, and atheroprotective natural antibodies for
cardiovascular disease in early rheumatoid arthritis an inception
cohort study. J Rheumatol 39: 1146-1154.
202. Lehmann N, Erbel R, Mahabadi AA, et al. (2018) Value of
Progression of Coronary Artery Calcification for Risk Prediction
of Coronary and Cardiovascular Events: Result of the HNR
Study (Heinz Nixdorf Recall). Circulation 137: 665-679.
203. Chung CP, Oeser A, Raggi P, et al. (2005) Increased
coronary-artery atherosclerosis in rheumatoid arthritis:
relationship to disease duration and cardiovascular risk factors.
Arthritis Rheum 52: 3045-3053.
204. Kobayashi Y, Giles JT, Hirano M, et al. (2010) Assessment
of myocardial abnormalities in rheumatoid arthritis using a
comprehensive cardiac magnetic resonance approach: a pilot
study. Arthritis Res Ther 12: 171.
205. Fine NM, Crowson CS, Lin G, et al. (2014) Evaluation of
myocardial function in patients with rheumatoid arthritis using
strain imaging by speckle-tracking echocardiography. Ann
Rheum Dis 73: 1833-1839.
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 22 of 23
206. Sitia S, Tomasoni L, Cicala S, et al. (2012) Detection of
preclinical impairment of myocardial function in rheumatoid
arthritis patients with short disease duration by speckle tracking
echocardiography. Int J Cardiol 160: 8-14.
207. Lopez-Mejias R, Castañeda S, González-Juanatey C,
et al. (2016) Cardiovascular risk assessment in patients with
rheumatoid arthritis: The relevance of clinical, genetic and
serological markers. Autoimmun Rev 15: 1013-1030.
208. Fransen J, Kazemi-Bajestani SM, Bredie SJ, et al. (2016)
Rheumatoid Arthritis Disadvantages Younger Patients for
Cardiovascular Diseases: A Meta-Analysis. PLoS One 11:
e0157360.
209. Ross R (1999) Atherosclerosis an inflammatory disease. N
Engl J Med 340: 115-126.
210. Sattar N, McCarey DW, Capell H, et al. (2003) Explaining
how high-grade systemic inflammation accelerates vascular risk
in rheumatoid arthritis. Circulation 108: 2957-2963.
211. Lehtinen K, Isomaki H (1991) Intramuscular gold therapy
is associated with long survival in patients with rheumatoid
arthritis. J Rheumatol 18: 524-529.
212. Choi HK, Hernán MA, Seeger JD, et al. (2002) Methotrexate
and mortality in patients with rheumatoid arthritis: a prospective
study. Lancet 359: 1173-1177.
213. Krause D, Schleusser B, Herborn G, et al. (2000) Response
to methotrexate treatment is associated with reduced mortality
in patients with severe rheumatoid arthritis. Arthritis Rheum 43:
14-21.
214. Atzeni F, Turiel M, Caporali R, et al. (2010) The effect
of pharmacological therapy on the cardiovascular system of
patients with systemic rheumatic diseases. Autoimmun Rev 9:
835-839.
215. Kerr G, Aujero M, Richards J, et al. (2014) Associations
of Hydroxychloroquine use with lipid profiles in rheumatoid
arthritis: pharmacologic implications. Arthritis Care Res 66:
1619-1626.
216. Morris SJ, Wasko MC, Antohe JL, et al. (2011)
Hydroxychloroquine use associated with improvement in lipid
profiles in rheumatoid arthritis patients. Arthritis Care Res 63:
530-534.
217. Wallace DJ, Metzger AL, Stecher VJ, et al. (1990)
Cholesterol-lowering effect of Hydroxychloroquine in patients
with rheumatic disease: reversal of deleterious effects of steroids
on lipids. Am J Med 89: 322-326.
218. Jacobsson LT, Turesson C, Gülfe A, et al. (2005) Treatment
with tumor necrosis factor blockers is associated with a
lower incidence of first cardiovascular events in patients with
rheumatoid arthritis. J Rheumatol 32: 1213-1218.
219. Dixon WG, Watson KD, Lunt M, et al. (2007) Reduction
in the incidence of myocardial infarction in patients with
rheumatoid arthritis who respond to anti-tumor necrosis factor
alpha therapy: results from the British Society for Rheumatology
Biologics Register. Arthritis Rheum 56: 2905-2912.
220. Roubille C, Richer V, Starnino T, et al. (2015) The effects
of tumour necrosis factor inhibitors, methotrexate, non-steroidal
anti-inflammatory drugs and corticosteroids on cardiovascular
events in rheumatoid arthritis, psoriasis and psoriatic arthritis:
a systematic review and meta-analysis. Ann Rheum Dis 74:
480-489.
221. Empana JP, Jouven X, Canouï-Poitrine F, et al. (2010)
C-reactive protein, interleukin 6, fibrinogen and risk of sudden
death in European middle-aged men: the PRIME study.
Arterioscler Thromb Vasc Biol 30: 2047-2052.
222. Masoud S, Boon Lim P, Kitas GD, et al. (2017) Sudden
cardiac death in patients with rheumatoid arthritis. World J
Cardiol 9: 562-573.
223. Charles-Schoeman C, Wang X, Lee YY, et al. (2016)
Association of Triple Therapy with Improvement in Cholesterol
Profiles over Two-Year Followup in the Treatment of Early
Aggressive Rheumatoid Arthritis Trial. Arthritis Rheumatol 68:
577-586.
224. Kleinbongard P, Heusch G, Schulz R (2010) TNFalpha
in atherosclerosis, myocardial ischemia/reperfusion and heart
failure. Pharmacol Ther 127: 295-314.
225. Low AS, Symmons DPM, Lunt M, et al. (2017) Relationship
between exposure to tumour necrosis factor inhibitor therapy
and incidence and severity of myocardial infarction in patients
with rheumatoid arthritis. Ann Rheum Dis 76: 654-660.
226. Charles-Schoeman C, Yin Lee Y, Shahbazian A, et al. (2017)
Improvement of High-Density Lipoprotein Function in Patients
With Early Rheumatoid Arthritis Treated With Methotrexate
Monotherapy or Combination Therapies in a Randomized
Controlled Trial. Arthritis Rheumatol 69: 46-57.
227. Popa C, van Tits LJ, Barrera P, et al. (2009) Anti-inflammatory
therapy with tumour necrosis factor alpha inhibitors improves
high-density lipoprotein cholesterol antioxidative capacity in
ISSN:XXXX-XXXX SFJC, an open access journal
Volume 2 · Issue 3 · 1000019SF J Cardiol
Citation: Arielle Urman (2018) Inflammation beyond the Joints: Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
page 23 of 23
rheumatoid arthritis patients. Ann Rheum Dis 68: 868-872.
228. Keystone EC, Genovese MC, Klareskog L, et al. (2009)
Golimumab, a human antibody to tumour necrosis factor {alpha}
given by monthly subcutaneous injections, in active rheumatoid
arthritis despite methotrexate therapy: the GO-FORWARD
Study. Ann Rheum Dis 68: 789-796.
229. Weinblatt ME, Mease P, Mysler E, et al. (2015) The
efficacy and safety of subcutaneous clazakizumab in patients
with moderate-to-severe rheumatoid arthritis and an inadequate
response to methotrexate: results from a multinational, phase
IIb, randomized, double-blind, placebo/active-controlled, dose-
ranging study. Arthritis Rheumatol 67: 2591-2600.
230. Verden A, Dimbil M, Kyle R, et al. (2018) Analysis of
Spontaneous Postmarket Case Reports Submitted to the FDA
Regarding Thromboembolic Adverse Events and JAK Inhibitors.
Drug Saf 41: 357-361.
231. McCarey DW, McInnes IB, Madhok R, et al. (2004) Trial
of Atorvastatin in Rheumatoid Arthritis (TARA): double-blind,
randomised placebo-controlled trial. Lancet 363: 2015-2021.
232. Tascilar K, Dell’Aniello S, Hudson M, et al. (2016) Statins
and Risk of Rheumatoid Arthritis: A Nested Case-Control Study.
Arthritis Rheumatol 68: 2603-2611.
233. Hermann F, Forster A, Chenevard R, et al. (2005)
Simvastatin improves endothelial function in patients with
rheumatoid arthritis. J Am Coll Cardiol 45: 461-464.
234. Van Doornum S, McColl G, Wicks IP (2004) Atorvastatin
reduces arterial stiffness in patients with rheumatoid arthritis.
Ann Rheum Dis 63: 1571-1575.
235. De Vera, Choi H, Abrahamowicz M, et al. (2011) Statin
discontinuation and risk of acute myocardial infarction in
patients with rheumatoid arthritis: a population-based cohort
study. Ann Rheum Dis 70: 1020-1024.
236. Maki-Petaja KM, BA, Hall FC, et al. (2007) Ezetimibe
and simvastatin reduce inflammation, disease activity, and
aortic stiffness and improve endothelial function in rheumatoid
arthritis. J Am Coll Cardiol 50: 852-858.
237. Schoenfeld SR, Lu L, Rai SK, et al. (2016) Statin use and
mortality in rheumatoid arthritis: a general population-based
cohort study. Ann Rheum Dis 75: 1315-1320.
238. Gwinnutt JM, Symmons DPM, MacGregor AJ, et al.
(2017) Twenty-Year Outcome and Association between Early
Treatment and Mortality and Disability in an Inception Cohort
of Patients with Rheumatoid Arthritis: Results from the Norfolk
Arthritis Register. Arthritis Rheumatol 69: 1566-1575.
239. Whelton SP, VMF, McEvoy JW, et al. (2017) Cardiovascular
Risk Assesment and Allocation of Lipid-Lowering Therapy
in Patients with Chronic Inflammatory Diseases. Am Coll
Cardiology.
240. Westerlind Hea (2018) Siblings of patients with rheumatoid
arthritis share increased risk for acute coronary syndrome.
Amsterdam.
241. Bartels CM, Heather J, Jacobs EA, et al. (2018) Frequency
and Predictors of Communication about High Blood Pressure
in Rheumatoid Arthritis Visits. J Clin Rheumatol 24: 210-217.
Citation: Arielle Urman (2018) Inflammation beyond the Joints:
Rheumatoid Arthritis and Cardiovascular Disease. SF J Cardiol 2:3.
... Cytokines present in chronic inflammation are implicated in the pathogenesis of atherosclerosis, while vascular changes manifesting as impaired vasodilation affect coronary flow reserve [3,10]. ...
... In this cross-sectional, single-center study, we identified patients with RA according to the International Classification of Diseases, Ninth Revision, and Tenth Revision Codes (ICD-9-CM 714.0, and ICD-10-C M06.00-M06.09) and HF (ICD-9-CM 428.0 -428.9 and ICD-10-CM I50.1 -I50.9). We included all inpatients and outpatients aged 18- [10]. For each patient diagnosed with RA-HF, we identified one age-and sex-matched patient from the historical cohort. ...
... Rheumatoid arthritis (RA) is a persistent inflammatory disease with various co-morbidities [3]. The job of caring is fundamental in the early diagnosis and treatment of this crippling illness [3]. ...
... Rheumatoid arthritis (RA) is a persistent inflammatory disease with various co-morbidities [3]. The job of caring is fundamental in the early diagnosis and treatment of this crippling illness [3]. Non-steroidal anti-inflammatory drugs (NSAIDs) are effective pain-relieving medications [4]. ...
Article
Purpose: To investigate the risk and safety profile of non-steroidal anti-inflammatory drugs (NSAIDs) for osteoporotic fractures in patients with rheumatoid arthritis (RA). Methods: 298 RA patients admitted to The First People's Hospital of Huzhou, Huzhou, China from August 2020 to September 2022, were investigated. Patients were assigned to groups based on drugs used viz: control group received anti-rheumatic drugs other than NSAIDs; corticosteroid group received dexamethasone, disease-modifying anti-rheumatic drugs (DMARDs) groups received sulfasalazine, NSAID group received either diclofenac or ibuprofen etc. Each group of patients received the respective treatment frequently for at least 3 years. The primary outcome in this study was the incidence of osteoporotic fracture resulting from fragile bone and deterioration of bone mass. Results: The incidence of osteoporotic fracture was highest in NSAID group (11.58 %) when compared to 5.44, 4.96, and 2.36 % in the corticosteroid, DMARD and control groups, respectively (p < 0.05). This shows that users of NSAIDs had a 5-fold higher risk of osteoporotic fracture than patients in control group (OR = 1.26 (95 % CI: 1.22 - 1.81)) and 2-fold possibility of osteoporotic fracture when compared to corticosteroid users ((OR = 1.46 (95 % CI: 1.57 - 2.32)) and DMARD users (1.83 (95 % CI: 1.26 - 1.67)). Conclusion: Rheumatoid arthritis patients on NSAIDs such as celecoxib, diclofenac, ibuprofen, indomethacin, and oxaprozin are at risk of developing osteoporotic fracture. Therefore, NSAIDs must be used with proper counseling in RA patients to minimize the risk of osteoporotic fracture. In future, the study duration will need to be extended to determine the long-term effects and potential changes in bone properties.
Article
Full-text available
The challenges associated with diagnosing and treating cardiovascular disease (CVD)/Stroke in Rheumatoid arthritis (RA) arise from the delayed onset of symptoms. Existing clinical risk scores are inadequate in predicting cardiac events, and conventional risk factors alone do not accurately classify many individuals at risk. Several CVD biomarkers consider the multiple pathways involved in the development of atherosclerosis, which is the primary cause of CVD/Stroke in RA. To enhance the accuracy of CVD/Stroke risk assessment in the RA framework, a proposed approach involves combining genomic-based biomarkers (GBBM) derived from plasma and/or serum samples with innovative non-invasive radiomic-based biomarkers (RBBM), such as measurements of synovial fluid, plaque area, and plaque burden. This review presents two hypotheses: (i) RBBM and GBBM biomarkers exhibit a significant correlation and can precisely detect the severity of CVD/Stroke in RA patients. (ii) Artificial Intelligence (AI)-based preventive, precision, and personalized (aiP³) CVD/Stroke risk AtheroEdge™ model (AtheroPoint™, CA, USA) that utilizes deep learning (DL) to accurately classify the risk of CVD/stroke in RA framework. The authors conducted a comprehensive search using the PRISMA technique, identifying 153 studies that assessed the features/biomarkers of RBBM and GBBM for CVD/Stroke. The study demonstrates how DL models can be integrated into the AtheroEdge™–aiP³ framework to determine the risk of CVD/Stroke in RA patients. The findings of this review suggest that the combination of RBBM with GBBM introduces a new dimension to the assessment of CVD/Stroke risk in the RA framework. Synovial fluid levels that are higher than normal lead to an increase in the plaque burden. Additionally, the review provides recommendations for novel, unbiased, and pruned DL algorithms that can predict CVD/Stroke risk within a RA framework that is preventive, precise, and personalized.
Article
Background: Rheumatoid arthritis (RA) is a chronic autoimmune disease that can lead to joint pain and disability, and seriously impact patients' quality of life. Strychni Semen combined with Atractylodes Macrocephala koidz (SA) have pronounced curative effect on RA, and there is no poisoning of Strychni Semen (SS). However, its pharmacological mechanisms are still unclear. Objective: In this study, we aimed to investigate the pharmacological mechanisms of Strychni Semen combined with Atractylodes Macrocephala Koidz (SA) for the treatment of RA. Methods: We used network pharmacology to screen the active components of SA and predict the targets and pathways involved. Results originating from network pharmacology were then verified by animal experiments. Results: Network pharmacology identified 81 active ingredients and 141 targets of SA; 2640 disease-related genes were also identified. The core targets of SA for the treatment of RA included ALB, IL-6, TNF and IL-1β. A total of 354 gene ontology terms were identified by Gene ontology (GO) enrichment analysis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis results showed that SA was closely associated with TNF signaling pathways in the treatment of RA. Furthermore, according to the predicted results of network pharmacology, we established a rat model of Adjuvant arthritis (AA) for in vivo experiments. Analysis showed that each treatment group led to an improvement in paw swelling, immune organ coefficient and synovial tissue morphology in AA rats to different degrees, inhibit the expression levels of IL-1β, TNF-α and IL-6, upregulated the levels of Fas, Bax and Caspase 3, down-regulated the expression levels of Fas-L, Bcl-2 and p53. Conclusion: SA has an anti-RA effect, the mechanism underlying the therapeutic action of SA in AA rats was related to the regulation of apoptosis signaling pathways.
Article
Full-text available
ABSTRACT The aim:The purpose of this study is to find out the association between procalcitonin and hepcidin in patients with COVID-19, in addition to their role as diagnostic markers. Materials and methods: A total of 75 patients infected with coronavirus were included in the current study, their age is ranging between 20 to 78 years. Those patients was hospitalized in Al-Sadr Teaching Hospital in Najaf, in Iraq. This study also included 50 healthy subjects which are volunteers and considered as a (control group). Biomarker (procalcitonin and hepcidin) measurements were achieved by electrochemiluminescent immunoassay (ECLIA) in the Elecsys immunoassay system. Results: The present study showed a significant increase the serum cencentration of hepcidin and procalcitonin in patients with COVID-19 as compared tohealthy subjects. There was a highly significant increasing(p < 0.01) in hepcidin and PCT level in patients with severe infection comparing to other catgaries.The current study also revealed that the sensitivity values of the markers were: 0.88%, 0.85 for procalcitonin and hepcidin respectively, which indicate high diagnostic power. Conclusions: Serum levels of hepcidin and procalcitonin are increased as inflammatory markers in COVID-19 patients with relatively high sensitivity. It seems that these imflammatory markers obviously elevate in the severe cases COVID-19dusease. KEY WORDS: Covid-19, hepcidin, procalcitonin, imflammatory markers
Article
Objective: The aim: To establish indicators and significance of sonography in the evaluation of muscle necrosis in ischemia of the limb acording to quantitative ultrasonographic indicators and density of collagen by histological method. Patients and methods: Materials and methods: In experiments, rabbits modeled with 6-hour limb ischemia by applying an elastic tourniquet. On days 5, 15, and 30, ultrasound and histological studies of the muscles and correlation analysis were performed between the muscles' entropy and the degree of their damage (atrophy, fibrosis and necrosis). Results: Results: The relative amount of structurally altered tissue was estimated morphometrically and compared with entropy. A high correlation of muscle damage with vertical δ-entropy indicates that sonography is highly likely to detect areas of necrosis and, to a lesser extent, fibrosis in the development of ischemic limb contracture in the early stages. Conclusion: Conclusions: Vertical δ-entropy in sonography is a significant indicator of muscle damage after traumatic ischemia and has strong relationship with muscle fibrosis.
Article
Objective: The aim: To determine the features of daily shifts in blood pressure (BP), the influence of the presence of rheumatoid arthritis (RA) on BP control and identify factors that affect BP among patients with RA in combination with resistant hypertension (RH). Patients and methods: Materials and methods: All material for writing this scientific work were the results of a comprehensive survey of 201 people with RH and RA, hypertension (H) and RA, RA without H, H without RA and relatively healthy individuals. A laboratory study was performed: rheumatoid factor, C-reactive protein (CRP), K+ serum, and creatinine levels. All patients underwent office BP measurement and ambulatory BP monitoring for 24 hours. Statistical processing of the study results was carried out using "IBM SPSS Statistics 22". Results: Results: Among patients with RA in combination with RH non-dippers (38.7%) are the most common type of BP profile. Patients with RH in combination with RA are characterized by an increase in BP more at night (p <0.003), which corresponds to the high frequency of night-peackers (17.7%). The presence of RA determines worse control of diastolic BP (p <0.01) and more vascular overload on organs and systems during the night (p <0.05). Conclusion: Conclusions: An increase in BP in patients with RA in combination with RH is more significant at night, characterized by poorer BP control and greater vascular load at night indicating the need for tighter control of BP during sleep. Non-dippers are most often detected among patients with RA in combination with RH, which is prognostically unfavorable for the development of nocturnal "vascular accidents".
Article
Objective: The aim: This study aimed to develop mouth-dissolving tablets of Acrivastine, an antihistamine medication, in order to increase its oral bioavailability. Patients and methods: Materials and methods: Different super disintegrants, such as crospovidone, croscarmellose sodium, and sodium starch glycolate, were used to make Acrivastine oral dispersible tablets (ODTs). These super disintegrants were utilized in various concentrations. The formulation (F3) with 6% w/w crospovidone had a fast disintegration time (less than 30 seconds) and practically total drug release within 10 minutes. All of the formulations were made using the direct compression method and proper diluents, binders, and lubricants. Fourier transform infrared spectroscopy (FTIR) tests were used to investigate the drug-ex¬cipient interaction, and all formulations demonstrated improved drug-excipient compatibility. Results: Results: The average weight of all formulations was between 175 and 180 mg. All formulations' hardness and friability were within acceptable ranges. Direct compression tablets had a hardness of 3.2 to 4 kg/cm2. All formulations were determined to have a friability of less than 1.0%. For oral dissolving tablets, the in vitro disintegration time is critical, and this time preferred to be < 60 seconds. The results also showed that crospovidone disintegrated after 24 seconds and sodium starch glycolate disintegrated in 40 seconds in vitro. Conclusion: Conclusions: When compared to croscarmellose sodium and sodium starch glycolate, crospovidone performs better as a super disintegrant. In comparison to other formula, tablets breakdown in the mouth in 30 seconds and have a maximum in vitro drug release time in 1-3 minutes.
Article
Full-text available
Background Rheumatoid arthritis (RA) is associated with accelerated atherosclerosis which is not fully explained by traditional risk factors. Such excess risk appears to be driven by systemic inflammation. Aim of the work Aim of the work was to compare between RA patients with and without CD4⁺CD28⁻ T-cell expansion regarding carotid intima-media thickness (IMT) and brachial artery flow-mediated endothelium-dependent dilatation (FMEDD), as markers of early atherosclerosis. Patients and methods The study was conducted on 39 female patients with no overt cardiovascular disease or risk factor and 28 age matched females as controls. Atherosclerotic changes were assessed through measurement of carotid IMT and FMEDD. CD4⁺CD28⁻ T-cells were assessed by flow cytometry. Results The mean age of the patients was 34.9 ± 5 years and the disease duration of 6.1 ± 2.1 years. Traditional risk factors were comparable between patients and controls. Serum homocysteine level tended to be higher in the patients (11 ± 4.21 μmol/L) compared to the control (9.91 ± 3.61 μmol/L). Patients had significantly higher carotid IMT (0.83 ± 0.24 mm vs 0.6 ± 0.15 mm, p = 0.008) and lower FMEDD (3.27 ± 1.49% vs 6.01 ± 1.79%, p = 0.002). Similarly, patients with CD4⁺CD28⁻ expansion (n = 12) had significantly higher IMT (1 ± 0.23 mm) and lower FMEDD (2.25 ± 1.06%) compared to those without (n = 27) (0.76 ± 0.21 mm and 3.67 ± 1.47%); p = 0.01, p = 0.01 respectively; but not affected by receiving methotrexate or not. Laboratory investigations were comparable in patients with and without expansion. Conclusion CD4⁺CD28⁻ cells may contribute to the development of premature atherosclerosis in RA patients. Further studies are recommended to evaluate the benefit of CD4⁺CD28⁻ T-cell modulation on the future development of atherosclerosis in these patients.
Article
Full-text available
Purpose of review: Increased cardiovascular (CV) risk and associated mortality in rheumatoid arthritis (RA) are not fully explained by traditional CV risk factors. This review discusses the epidemiology and mechanisms of increased CV risk in RA and treatment effects on CV risk focusing on biologic disease-modifying anti-rheumatic drugs (DMARDs) and JAK inhibitors. Recent findings: Intermediary metabolic changes by inflammatory cytokines are observed in body composition, lipid profile, and insulin sensitivity of RA patients, leading to accelerated atherosclerosis and increased CV risk. Successful treatment with DMARDs has shown beneficial effects on these metabolic changes and ultimately CV outcomes, in proportion to the treatment efficacy in general but also with drug-specific mechanisms. Recent data provide further information on comparative CV safety between biologic DMARDs or JAK inhibitors as well as their safety signals for non-atherosclerotic CV events. CV benefits or safety signals associated with DMARD treatments can differ despite similar drug efficacy against RA, suggesting that both anti-inflammatory and drug-specific mechanisms are involved in altering CV risk.
Article
Full-text available
Objectives: Patients with RA display greater occult coronary atherosclerosis burden and experience higher cardiovascular morbidity and mortality compared with controls. We here explored whether pro-inflammatory cytokines and high-sensitivity cardiac troponin I (hs-cTnI), a biomarker of myocardial injury, correlated with plaque burden and cardiovascular events (CVEs) in RA. Methods: We evaluated 150 patients with 64-slice coronary CT angiography. Coronary artery calcium, number of segments with plaque (segment involvement score), stenotic severity and plaque burden were assessed. Lesions were described as non-calcified, mixed or fully calcified. Blood levels of hs-cTnI and pro-inflammatory cytokines were assessed during coronary CT angiography. Subjects were followed over 60 (s.d. 26) months for both ischaemic [cardiac death, non-fatal myocardial infarction (MI), stroke, peripheral arterial ischaemia] and non-ischaemic (new-onset heart failure hospitalization) CVEs. Results: Plasma hs-cTnI correlated with all coronary plaque outcomes (P < 0.01). Elevated hs-cTnI (⩾1.5 pg/ml) further associated with significant calcification, extensive atherosclerosis, obstructive plaque and any advanced mixed or calcified plaques after adjustments for cardiac risk factors or Framingham D'Agostino scores (all P < 0.05). Eleven patients suffered a CVE (1.54/100 patient-years), eight ischaemic and three non-ischaemic. Elevated hs-cTnI predicted all CVE risk independent of demographics, cardiac risk factors and prednisone use (P = 0.03). Conversely, low hs-cTnI presaged a lower risk for both extensive atherosclerosis (P < 0.05) and incident CVEs (P = 0.037). Conclusion: Plasma hs-cTnI independently associated with occult coronary plaque burden, composition and long-term incident CVEs in patients with RA. Low hs-cTnI forecasted a lower risk for both extensive atherosclerosis as well as CVEs. hs-cTnI may therefore optimize cardiovascular risk stratification in RA.
Article
Full-text available
Background: Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with a wide range of comorbidities, including cardiovascular disease, but its association with heart failure (HF) is not fully clear. We investigated the risk of incident HF in a nationwide cohort of patients with RA. Methods and results: The study comprised the entire Danish population aged ≥18 years followed from January 1, 2008 until first hospitalization for HF, emigration, December 31, 2012, or death. Information on comorbidity, medication, and socioeconomic status was identified by individual-level linkage of administrative registers. Patients with a rheumatologist diagnosis of RA between 1978 and 2008 were included. The primary study outcome was incident HF defined as first hospital admission for HF. Incidence rates of HF per 1000 person-years were calculated and incidence rate ratios adjusted for age, sex, calendar year, comorbidity, medications, socioeconomic status, smoking, and alcohol consumption were estimated. A total of 4 305 225 subjects with no history of HF were eligible for analysis at the study start. Of these subjects, 24 343 developed RA and 50 623 were hospitalized for HF. Overall incidence rates of incident HF were 2.43 and 6.64 for the reference population (n=49 879) and patients with RA (n=744), respectively. Correspondingly, the fully adjusted incidence rate ratio for incident HF was increased in patients with RA with incidence rate ratio 1.30 (95% confidence interval, 1.17-1.45). Conclusions: In this cohort study, RA was associated with an increased hospitalization for HF. These findings add significantly to the existing evidence of RA as a clinically relevant risk factor for HF.
Conference Paper
Background Patients with rheumatoid arthritis (RA) are at increased risk of cardiovascular disease such as acute coronary syndromes (ACS), which cannot entirely be explained by traditional cardiovascular risk factors. Studies have shown an association between RA disease severity and risk of ACS, speaking for a contribution of the RA disease per se to the excess ACS risk. In a recent report, however, we demonstrated that despite more efficient control of inflammation in RA during the recent years, the excess risk for ACS among patients with RA compared to the general population remains¹. This finding suggests that besides effects related to the RA disease per se, there may be a shared susceptibility. If the excess risk of ACS in patients with RA were increased due to this, an increased risk of ACS would be observed also in individuals with a similar genetic set-up and background as the patients with RA, such as their siblings. Objectives To investigate any potential shared susceptibility between RA and ACS by estimating the risk of ACS in full siblings of patients with (vs. without) RA. Methods We used the Swedish Rheumatology Quality register (SRQ) to identify an early RA cohort diagnosed between 1996–2015, which was linked to the Swedish Multigeneration Register, Patient Register, the Cause of Death register, and the Total Population Register. Through this, we sampled five general population comparator subjects to each patient with RA, matched by birth year and sex, and identified all full siblings to patients with RA and for their comparator subjects born within five years of their index case. The comparators, and all siblings, were required to be alive and living in Sweden at the time of the index patient’s RA diagnosis (=start of follow-up). All unique individuals were then followed for ACS (defined as first ever hospitalisation for ACS (ICD10: I21 or I20.0) or MI listed as the cause of death), and censored at death, migration, RA diagnosis (for non-RA subjects) or the end of the study (Dec 31 st 2015). We calculated hazard ratios (HR) using a Cox proportional hazards model, adjusting for age, sex and calendar period of diagnosis. Confidence intervals (CI) were estimated using a robust sandwich estimator. Results We identified 7492 patients with RA who had 10 671 full siblings, and 35 120 population comparator subjects with 47 137 full siblings. The HR for ACS was 1.44 (95% CI: 1.25 to 1.66) and 1.23 (95% CI: 1.09 to 1.40) for patients with RA and their siblings, respectively, compared to the comparator subjects. A direct comparison between the RA patients and their RA-free siblings confirmed the familial association between RA and ACS, HR 1.19 (95% CI: 1.02 to 1.38). Conclusions The increased risk of ACS in siblings of patients with RA a) provide evidence of shared susceptibility between RA and ACS, the nature of which needs to be further investigated, and b) suggests that to bring down the CV risk in RA to that in the general population, cardiopreventive measures must go beyond optimised RA disease control. Reference [1] Holmqvist M, Ljung L, Askling J Acute coronary syndrome in new-onset rheumatoid arthritis: a population-based nationwide cohort study of time trends in risks and excess risks, Ann Rheum Dis2017 Disclosure of Interest H. Westerlind: None declared, M. Holmqvist: None declared, L. Ljung: None declared, T. Frisell: None declared, J. Askling Grant/research support from: Johan Askling has or has had research agreements with Abbvie, BMS, MSD, Pfizer, Roche, Astra-Zeneca, Eli Lilly, Samsung Bioepis, and UCB, mainly in the context of safety monitoring of biologics via ARTIS. Karolinska Institutet has received remuneration for JA participating in advisory boards arranged by Pfizer and Eli Lilly.
Article
Cardiovascular disease (CVD) risk is 1.5-fold higher in rheumatoid arthritis (RA), partly due to subclinical atherosclerosis that develops before the diagnosis of RA. Dyslipidemia in RA is better quantified by lipoproteins and apolipoproteins than by cholesterol levels. Current risk factors likely underestimate CVD risk by underestimating prior risk factor levels. Some of the 2-fold higher risk of heart failure and total mortality in RA may be due to myocardial disease caused by inflammation. Per recent recommendations, to reduce CVD risk in RA, control disease activity, reduce inflammation, and aggressively treat CVD risk factors.
Article
Background/objective: Given heightened cardiovascular disease (CVD) risk in rheumatoid arthritis (RA) and that higher blood pressure (BP) represents greater CVD risk, we hypothesized that higher BP would predict more BP-related communication in rheumatology visits. We examined predictors of documented BP communication during RA clinic visits. Methods: This was a retrospective cohort study of RA patients identified in electronic health record records with uncontrolled hypertension (HTN) receiving both primary and rheumatology care. Trained abstractors reviewed RA visit notes for "BP communication" using a standardized tool to elicit documentation about BP or HTN beyond recording vital signs. We used multivariate logistic regression to examine the impact of BP category (American Heart Association: ideal normotension, pre-HTN, and stages I and II HTN) on odds ratios (95% confidence intervals) of BP communication. Results: Among 1267 RA patients, 40% experienced BP elevations meeting the definition of uncontrolled HTN. Of 2677 eligible RA visits, 22% contained any documented BP communication. After adjustment, models predicted only 31% of visits with markedly high BPs 160/100 mm Hg or greater would contain BP communication. Compared with stage I, stage II elevation did not significantly increase communication (odds ratio, 2.0 [95% confidence interval, 1.4-2.8] vs. 1.5 [1.2-2.2]), although both groups' odds exceeded pre-HTN and normotension. Less than 10% of eligible visits resulted in documented action steps recommending follow-up of high BP. Conclusions: Regardless of BP magnitude, most RA clinic visits lacked documented communication about BP despite compounded CVD risk. Future work should study how rheumatology clinics can facilitate follow-up of high BPs to address HTN as the most common and reversible CVD risk factor.
Article
Objective: To compare mortality rates, underlying causes of death, excess mortality and years of potential life lost (YPLL) among rheumatoid arthritis (RA) patients relative to the general population. Methods: We studied an inception cohort of 87,114 Ontario RA patients and 348,456 age/sex/area-matched general population comparators over 2000 to 2013. All-cause, cause-specific, and excess mortality rates, mortality rate ratios (MRRs), and YPLL were estimated. Results: A total of 11,778 (14% of) RA patients and 32,472 (9% of) comparators died during 508,385 and 1,769,365 person-years (PY) of follow-up, respectively, for corresponding mortality rates of 232 (95% CI 228, 236) and 184 (95% CI 182, 186) per 10,000 PYs. Leading causes of death in both groups were diseases of the circulatory system, cancer, and respiratory conditions. Increased mortality for all-cause and specific causes was observed in RA relative to the general population. MRRs were elevated for most causes of death. Age-specific mortality ratios illustrated a high excess mortality among RA patients under 45 years of age for respiratory disease and circulatory disease. RA patients lost 7,436 potential years of life per 10,000 persons, compared with 4,083 YPLL among those without RA. Conclusion: Mortality rates were increased in RA patients relative to the general population across most causes of death. The potential life years lost (before the age of 75) among RA patients was roughly double that among those without RA, reflecting higher rate ratios for most causes of death and RA patients dying at earlier ages. This article is protected by copyright. All rights reserved.
Article
Objectives: To investigate the incidence of heart failure (HF) and ischemic heart disease (IHD) in different time-spans following incident rheumatoid arthritis (RA). Furthermore, to investigate the impact of IHD on the development of HF and the impact of different treatment era's of RA. Methods: This matched cohort study used nationwide, prospectively collected data. From the total Danish population of approximately 5.7 million inhabitants we identified 51,859 patients (between 1995 and July 2016) with incident RA and a sex and age matched cohort from the general population (256,653 persons). Results: The hazard ratio (HR) for HF among RA patients compared with persons from comparison cohort were 2.28 within the first year of index date, 1.39 within1-5 years of index date, and 1.38 within 5-10 after index date. No difference was identified regarding different treatment era's of RA. For IHD the sHR was 1.93 within the first year of index date, 1.26 within 1-5 years of index date, and 1.31 within 5-10 after index date. Coronary revascularization was also more common within the first year after diagnosis of RA. An increased risk of percutaneous coronary intervention and coronary by-pass grafting within 10 years following the RA diagnosis were observed. HR for new onset of HF in RA without IHD was 1.23, while the HR for new onset of HF in patients with RA and IHD was 2.06. Conclusions: RA patients had higher rates of HF and IHD throughout the entire observation period compared to the comparison cohort. RA was associated with a larger risk for developing HF. This article is protected by copyright. All rights reserved.