ArticlePDF AvailableLiterature Review

Cardiovascular Disease in Systemic Lupus Erythematosus: Recent Data on Epidemiology, Risk Factors and Prevention

Authors:

Abstract and Figures

Systemic Lupus Erythematosus (SLE) is associated with increased risk for accelerated atherosclerosis and cardiovascular (CV) events including coronary heart disease, cerebrovascular and peripheral artery disease. CV events occur both early and late during the disease course, with younger patients being at much higher risk than age-matched counterparts. The risk cannot be fully accounted for by the increased prevalence of traditional atherosclerotic factors and may be due to pathophysiologic intermediates such as type I interferons and other inflammatory cytokines, oxidative stress, activated granulocytes and production of extracellular chromatin traps, antiphospholipid and other autoantibodies causing dysfunction of lipoproteins, altogether resulting in endothelial injury and pro-atherogenic dyslipidaemia. These mechanisms may be further aggravated by chronic intake of prednisone (even at doses <7.5 mg/day), whereas immunomodulatory drugs, especially hydroxychloroquine, may exert antiatherogenic properties. To date, there is a paucity of randomized studies regarding the effectiveness of preventative strategies and pharmacological interventions specifically in patients with SLE. Nevertheless, both the European League Against Rheumatism recommendations and extrapolated evidence from the general population emphasize that SLE patients should undergo regular monitoring for atherosclerotic risk factors and calculation of the 10-year CV risk. Risk stratification should include diseaserelated factors and accordingly, general (lifestyle modifications/smoking cessation, antihypertensive and statin treatment, low-dose aspirin in selected cases) and SLE-specific (control of disease activity, minimization of glucocorticoids, use of hydroxychloroquine) preventive measures be applied as appropriate. Further studies will be required regarding the use of non-invasive tools and biomarkers for CV assessment and of risk-lowering strategies tailored to SLE.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.net
Current Vascular Pharmacology, 2020, 18, 000-000 1
REVIEW ARTICLE
1570-1611/20 $65.00+.00 © 2020 Bentham Science Publishers
Cardiovascular Disease in Systemic Lupus Erythematosus: Recent Data on
Epidemiology, Risk Factors and Prevention
Myrto Kostopoulou1, Dionysis Nikolopoulos1, Ioannis Parodis2 and George Bertsias3,4,*
14th Department of Internal Medicine, Attikon University Hospital, Joint Rheumatology Program, National and Kapo-
distrian University of Athens Medical School, Athens, Greece; 2Division of Rheumatology, Department of Medicine
Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; 3Department of
Rheumatology, Clinical Rheumatology and Allergy, University of Crete Medical School, Iraklio, Greece; 4Laboratory of
Rheumatology, Autoimmunity and Inflammation, Institute of Molecular Biology and Biotechnology-FORTH, Iraklio,
Greece
A R T I C L E H I S T O R Y
Received: July 19, 2019
Revised: November 20, 2019
Accepted: December 12, 2019
DOI:
10.2174/1570161118666191227101636
Abstract: Systemic Lupus Erythematosus (SLE) is associated with increased risk for accelerated athe-
rosclerosis and cardiovascular (CV) events including coronary heart disease, cerebrovascular and pe-
ripheral artery disease. CV events occur both early and late during the disease course, with younger
patients being at much higher risk than age-matched counterparts. The risk cannot be fully accounted for
by the increased prevalence of traditional atherosclerotic factors and may be due to pathophysiologic
intermediates such as type I interferons and other inflammatory cytokines, oxidative stress, activated
granulocytes and production of extracellular chromatin traps, antiphospholipid and other autoantibodies
causing dysfunction of lipoproteins, altogether resulting in endothelial injury and pro-atherogenic
dyslipidaemia. These mechanisms may be further aggravated by chronic intake of prednisone (even at
doses <7.5 mg/day), whereas immunomodulatory drugs, especially hydroxychloroquine, may exert anti-
atherogenic properties. To date, there is a paucity of randomized studies regarding the effectiveness of
preventative strategies and pharmacological interventions specifically in patients with SLE. Neverthe-
less, both the European League Against Rheumatism recommendations and extrapolated evidence from
the general population emphasize that SLE patients should undergo regular monitoring for atheroscle-
rotic risk factors and calculation of the 10-year CV risk. Risk stratification should include disease-
related factors and accordingly, general (lifestyle modifications/smoking cessation, antihypertensive and
statin treatment, low-dose aspirin in selected cases) and SLE-specific (control of disease activity, mini-
mization of glucocorticoids, use of hydroxychloroquine) preventive measures be applied as appropriate.
Further studies will be required regarding the use of non-invasive tools and biomarkers for CV assess-
ment and of risk-lowering strategies tailored to SLE.
Keywords: Autoimmune disease, type I interferon, antiphospholipid antibodies, glucocorticoids, dyslipidaemia, ischemic heart
disease, cerebrovascular disease, statin.
1. INTRODUCTION
Systemic Lupus Erythematosus (SLE) is the prototype
systemic autoimmune disease that can affect any organ [1].
SLE usually starts in early or mid-adulthood and affects
more women than men [1]. Its pathogenesis is complex and
involves an interaction between genetic and environmental
factors, which results in the activation of innate and adaptive
*Address correspondence to this author at the Department of Rheumatology,
Clinical Rheumatology and Allergy, University of Crete Medical School,
71003 Voutes, Iraklio, Greece; Tel: (+30) 2810 394635;
E-mail: gbertsias@uoc.gr
immune pathways and the production of autoantibodies di-
rected against nuclear and other self-antigens [1]. Despite
advances in the treatment and overall management, patients
with SLE still suffer from increased morbidity and mortality
as compared with the general population, and an increased
burden of comorbid conditions including metabolic and car-
diovascular (CV) disease (CVD) [2, 3]. This has been further
augmented by the fact that life expectancy for SLE patients
has increased considerably over the last decades [4]; thus,
patients live long enough to manifest such complications.
In this review, we summarize the evidence on the epide-
miology and risk factors of CVD in SLE. We discuss the
2 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
major pathophysiologic drivers of atherosclerosis in the con-
text of the disease, as well as the existing modalities in risk
assessment and stratification. We conclude based on the evi-
dence-based use of preventative measures and some practical
considerations in the routine management of SLE.
2. DEFINITIONS
Atherosclerosis is a chronic inflammatory state character-
ized by the accumulation of lipids and immune cells in the
sub-endothelial arterial wall, resulting in inflammation-
driven plaque formation [5]. It is the main contributor to the
clinical burden of CVD and subclinical forms of atheroscle-
rosis constitute a risk for and are predictive of future cardio-
vascular events (CVE) [6]. However, there are patients with
atherosclerosis who never develop CVE, and, conversely,
there are patients with CVE without any evidence of sub-
clinical atherosclerosis [7]. The complexity of this interplay
becomes more prominent in SLE, where inflammatory me-
diators may independently contribute to the formation of
atherosclerotic plaques and subsequent CVE [5, 8, 9].
In most epidemiologic studies on SLE outcomes, CVD
comprises coronary heart disease (CHD; myocardial infarc-
tion (MI), angina pectoris and/or heart failure), cerebrovas-
cular disease (a stroke or transient ischemic attack) and pe-
ripheral artery disease (intermittent claudication and critical
limb ischemia) [10]. To this end, study designs on survival
outcomes require large patient cohorts and long-term follow-
up. As a result, most studies investigate subclinical athero-
sclerosis rather than actual CVE rates. The former is defined
by the presence of plaque, however, a number of surrogate
markers are commonly used, particularly carotid intima me-
dia thickness (cIMT) and coronary artery calcium (CAC)
[11]. Other non-invasive assessments of endothelial function
and vascular stiffness such as flow-mediated vasodilation
and pulse wave velocity (PWV) are also used due to their
feasibility and established correlation to CVD risk in the
general population [12-14]. Given the heterogeneity and sub-
jectivity of the techniques used to assess CVD, results may
not always be generalized.
3. EPIDEMIOLOGY OF CVD IN SLE
The impact of CVD on SLE mortality was first docu-
mented in the 1970’s by Urowitz et al. who described a bi-
modal mortality pattern where an early peak (within 1 year of
diagnosis) attributed to disease activity and infections, was
followed by a later one (>5 years after diagnosis) due to CVD
[15]. Since then, death related to lupus activity has been mark-
edly decreased due to the introduction of new treatments and
improved overall management. However, CV mortality re-
mains unchanged and is considered one of the main causes of
death in SLE, especially during late disease stages [16, 17].
In recent studies, the overall prevalence of CVE in SLE
varies between 4% and 20% depending on the definitions
used and the ethnic and age variation [18-22]. According to a
recent meta-analysis including 17,187 SLE patients, CVE
occurrence had a pooled estimate of 25.4% in a median fol-
low-up of 8 years [23]. SLE patients have a 2- to 10-fold
increased risk of clinical CVD compared with the general
population, even after adjustments for traditional risk factors
[21, 24-27]. This risk is even more pronounced among
younger women, as illustrated in the Framingham Offspring
Study, where premenopausal women with SLE were over
50-times more likely to suffer an MI than age- and sex-
matched controls [28]. Notably, the risk of CV morbidity is
increased since the early stages of the disease. In two studies
examining CVE risk during the first and second year after
SLE diagnosis, the adjusted hazard ratios (HR) were 3.8 and
6.0, respectively [25, 29].
As in the general population, subclinical atherosclerosis
in SLE is more prevalent than clinical CVD. A recent study
found the risk of subclinical atherosclerosis in SLE patients
to be comparable to that in matched patients with diabetes
mellitus (DM) and rheumatoid arthritis, suggesting compara-
ble cardiovascular burden [30]. In two autopsy series, athe-
rosclerotic coronary plaques were seen in up to 40% of the
tissue segments of SLE patients [31, 32]. The premature on-
set and the accelerated nature of atherosclerosis have also
been documented in several cross-sectional and prospective
studies [9, 33-37]. Thus, both cIMT and carotid plaques were
more frequent in SLE patients than age- and sex-matched
controls [9, 38]. In a meta-analysis of 80 studies, SLE pa-
tients had a greater cIMT and a 2-fold increased prevalence
of carotid plaques compared with controls [39]. Although
data connecting subclinical atherosclerosis to the incidence
of CVEs in SLE are sparse, one prospective study with a
mean follow-up of 8 years showed that women with cIMT or
carotid plaque at baseline had an increased incidence of
CVD (HR 1.35 and 4.26, respectively) [40].
Traditional atherosclerotic risk factors contribute to in-
creased risk but cannot fully explain the accelerated CVD in
SLE. In a retrospective study of 263 SLE patients, there was a
marked difference between the observed and expected (as-
sessed according to the Framingham risk score) incidence of
CHD, resulting in a 7.5-fold increased risk [27]. In another
prospective study of CHD in patients with SLE and matched
controls followed over a median of 8 years, SLE per se was an
independent risk factor even after correcting for traditional
atherosclerotic risk factors [41]. What accounts for this excess
risk remains elusive, but studies suggest that it can be partly
attributed to disease and treatment-related factors (Table 1).
Specifically, among various SLE-specific factors that have
been found to correlate with CVD (i.e. disease activity and
duration, cumulative damage, presence of antiphospholipid
(aPL) antibodies, lupus nephritis, and various treatments), data
on disease duration are more robust, indicating as expected,
longer duration as an independent predictor for both CVE and
accelerated atherosclerosis [40, 42-45]. Thus, in a cohort of
392 women with SLE, disease duration at enrolment correlated
with the incidence of CVE in age-adjusted Cox regression
models (HR: 1.04 per 1-year) [40]. Higher organ damage (as-
sessed by the Systemic Lupus International Collaborating
Clinics/American College of Rheumatology Damage Index)
was also significantly associated with CVE and subclinical
atherosclerosis [22, 23, 46]. Conversely, data on the impact of
disease activity on atherosclerosis are less consistent in the
literature [21, 28, 47-49], with several studies failing to dem-
onstrate significant associations [39, 50, 51].
4. PATHOGENESIS OF ACCELERATED CVD IN SLE
At an early stage, endothelial injury caused by oxidized
low-density-lipoprotein (oxLDL) induces the expression of
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 3
endothelial cell adhesion molecules and recruitment of
monocytes, which differentiate into macrophages [52]. In-
gestion of oxLDL by these macrophages transforms them
into cholesterol-laden “foam cells [52]. The subsequent
production of growth factors and cytokines leads to the pro-
liferation of vascular smooth muscle cells and plaque devel-
opment [53]. To this end, although the pathogenesis of ac-
celerated atherosclerosis in SLE is not thoroughly under-
stood, systemic inflammatory response, together with the
early appearance of traditional risk factors, is believed to
exacerbate the atherogenic process [54].
4.1. The Role of Pathophysiology Intermediates
4.1.1. Inflammatory Cytokines
Several studies have implicated type I interferons (IFNs)
in premature atherosclerosis in murine and human SLE [55-
58]. Patients with SLE have an increased expression of type I
IFN-regulated genes (so-called interferon signature’),
which play important roles in the pathogenesis of this disease
[59]. Notably, these IFNs may hamper vascular repair and
inhibit angiogenesis by decreasing endothelial progenitor
cells and suppressing neo-vascularization in sites of endothe-
lial injury [55]. In addition to their anti-angiogenic role, type
I IFNs may also contribute to the atherogenic process by
enhancing foam cell formation [60], promoting platelet acti-
vation, impairing smooth muscle cell maturation, and leading
to plaque rupture [61, 62]. In a study that evaluated type I
IFNs in relation to subclinical atherosclerosis in SLE, en-
hanced serum IFN activity was associated with increased
cIMT and severity of CAC in SLE patients but not in con-
trols [57]. Another cytokine, tumour necrosis factor (TNF)-α
also contributes to endothelial injury by stimulating mono-
cyte differentiation into macrophages and foam cells. Al-
though high levels of TNF-α in SLE have been associated
with high triglyceride and low high density lipoprotein cho-
lesterol (HDL-C) levels [63], their actual impact on subclini-
cal atherosclerosis remains uncertain [9, 64]. Other impli-
cated cytokines include interleukin (IL)-6 and IL-17, with
circumstantial evidence suggesting a possible link between
their increased levels and atherosclerosis is SLE [65-67].
4.1.2. Oxidized Low-Density Lipoproteins (oxLDL)
Low-density lipoproteins (LDL) trapped in the subendo-
thelial space and exposed to active reactive oxygen species
(ROS) result in the formation of oxLDLs. Next, chemokines
and proinflammatory cytokines released by endothelial cells,
promote monocytes infiltration to the subendothelial space
where they differentiate into macrophages [52]. The oxLDLs
are phagocytosed by macrophages, which transform into
foam cells [52]. In SLE patients, circulating oxLDLs are
increased and have been correlated with measures of sub-
clinical atherosclerosis [68]. This might be due to the higher
levels of proinflammatory high-dense lipoprotein (pi-HDL)
observed in SLE, which, in contrast to normal HDL, does not
prevent LDL oxidation [69]. Moreover, antibodies against
HDL (anti-HDL) or against apolipoprotein A1 (the main
constituent of HDL) impair the anti-oxidant activity of nor-
mal HDL, therefore enhancing LDL oxidation [70]. OxLDL
may also create complexes with anti-β2-glycoprotein I (anti-
β2-GPI) autoantibodies, which are encountered in a propor-
tion of SLE patients, and induce an antibody-mediated
phagocytosis by neighbouring macrophages, thus accelerat-
ing the atherogenic process [71].
4.1.3. Neutrophils and Neutrophil Extracellular Traps
(NETs)
Neutrophils have been recently implicated in the patho-
genesis of atherosclerosis. Their role in innate immune re-
sponse is well-described and encompasses mechanisms such
as phagocytosis and intracellular degradation, degranulation,
the release of ROS and formation of NETs, all physiologi-
cally induced following a pathogen insult [72]. However,
some of these mechanisms may be triggered by endogenous
molecules such as cytokines [73]. In lupus, the co-existence
of a specific subset of proinflammatory neutrophils, known
as low density granulocytes (LDGs), and atherosclerosis has
been shown to be pathogenic [74]. In a recent study, vascular
Table 1. SLE-related risk factors with significant associations with cardiovascular diseases and atherosclerosis.
CVD
Subclinical Atherosclerosis
Risk Factor
No. Studies
OR
No. Studies
Disease activity
3
7
Disease duration
5
2.30 3.16
12
Cumulative damage
2
1.7
5
a-PL (any)
12
5.2
1
Renal involvement
7
2.16
3
GCs
8
0.33 2.93
7
HCQ
8
3
CYC
0.31
1
AZA
2.33 2.58
4
CVD, cardiovascular disease; OR, odds ratio; a-PL, anti-phospholipid antibodies; GCs, glucocorticoids; HCQ, hydroxychloroquine; CYC, cyclophosphamide; AZA, azathioprine.
4 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
damage and non-calcified plaque burden (an indicator of
plaque vulnerability assessed by computed tomography an-
giography) correlated with the proportion of circulating
LDGs in SLE subjects. Interestingly, the Framingham risk
score did not correlate with non-calcified plaque burden,
suggesting an independent role of LDGs in plaque formation
and instability [75]. To this end, LDGs have been linked to
increased type I IFN production and endothelial damage via
NETosis [76]. NETosis represents a unique type of cell
death, different from apoptosis and necrosis, that causes ex-
ternalization of fibrous networks (NETs) consisting of nu-
clear chromatin, antimicrobial components including mye-
loperoxidase, neutrophil elastase and human cathelecidin
(LL-37) [77]. The mechanism that connects NETs to athero-
sclerosis in the SLE population has yet to be defined. None-
theless, in a study in paediatric SLE patients, NET content
was shown to be capable of inducing IFN-α by plasmacytoid
dendritic cells [78], while in another study, excess of NETs
resulted in vascular leakage and endothelial-to-mesenchymal
transition, thus jeopardizing vascular integrity [79].
4.1.4. Autoantibodies
Several autoantibodies and immune complexes have been
linked to vascular damage and atherosclerosis in lupus. aPL
antibodies including lupus anticoagulant, anticardiolipin and
anti-β2-GPI antibodies, represent a strong risk factor for
thrombotic events driven by immune-mediated mechanisms
rather than atherosclerosis [80]. aPL antibody positive SLE
patients, especially patients with moderate to high serum
antibody levels, are at increased risk of venous and arterial
thrombotic events compared with aPL-negative counterparts
[81]. Although the underlying mechanism that links the pres-
ence of aPL antibodies to endothelial injury in lupus is not
clear, possible pathways include the anti-β2-GPI binding to
β2-GPI and the subsequent endothelial cell activation [82]
and enhanced formation of foam cells induced by a complex
of ox-LDL/β2-GPI/anti-β2-GPI [83]. Conversely, IgM
autoantibodies against phosphorylcholine and malondialde-
hyde have been recently found to play a protective role
against atherosclerosis in SLE, probably by increasing the
clearance of apoptotic cells and by reducing oxidative stress
[84, 85].
4.2. Increased Prevalence of Traditional CVD Risk Fac-
tors
The increased CVD burden in SLE can be partly attrib-
uted to the increased prevalence and deleterious effect of
traditional risk factors (Table 2). In a study from the Toronto
Table 2. Summary of the recent studies reporting on traditional cardiovascular risk factors in patients with SLE.
CVD Risk
Atherosclerosis Risk
(cIMT/Carotid Plaque)
Risk Factor
Significant
Associations
Non-Significant
Associations
Significant
Associations
Non-Significant
Associations
Comments
10 studies
3 studies
12 studies
3 studies
Hypertension
(HR 2.318.05)
(OR 1.44.8)
High level of evidence
4 studies
4 studies
6 studies
2 studies
DM
OR: 7.07-61.8
Inconsistent results
8 studies
3 studies
12 studies
6 studies
Dyslipidemia
OR: 2.18-5.37
OR: 1.52-3.4
Great heterogeneity under the term
dyslipidemia
2 studies
1 study
Metabolic syndrome
HR: 3.88
OR: 3.11
Low level of evidence
7 studies
4 studies
7 studies
4 studies
Smoking
OR: 1.48-3.25
OR: 1.05-5.89
Heterogeneity regarding the expo-
sure variable (ever/current/no
smoking, pack-years)
4 studies
2 studies
Homocysteine
OR: 1.24
No study with CVE as outcome
1 study
3 studies
3 studies
Obesity
OR: 4.09
Low level of evidence
4 studies
3 studies
4 studies
4 studies
CKD
OR: 1.002-2.26
(creatinine levels)
Heterogeneity regarding the out-
comes (creatinine levels, eGFR,
definition of CKD)
CVD, cardiovascular disease; cIMT, carotid intima media thickness; HR, hazard ratio; OR, odds ratio; DM, diabetes mellitus; CKD, chronic kidney disease; eGFR, estimated glome-
rular filtration rate.
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 5
Lupus Cohort, women with SLE had increased prevalence of
hypertension, DM, premature menopause and sedentary life-
style compared to their age-matched counterparts [86]. How-
ever, SLE subjects have increased CVD risk even after ac-
counting for these factors, which explains why risk assess-
ment models used in the general population (e.g. the
Framingham risk score) underestimate the actual hazard in
SLE [27]. Age and gender are the most robust, non-
modifiable risk factors for CVD both in the general popula-
tion and in patients with SLE [22, 87]. Specifically, higher
age and male sex have been associated with multiple meas-
ures of subclinical atherosclerosis and increased incidence of
CVE, yielding an up to 3-fold increase in CVD events in
SLE [88-90]. Among modifiable factors, arterial hyperten-
sion, dyslipidaemia, and smoking have consistently been
reported as predictors of CVD in SLE, whereas data on DM,
metabolic syndrome and homocysteinaemia are less robustly
associated. In a recent meta-analysis of 32 studies, hyperten-
sion (OR 3.5) and dyslipidaemia (OR 3.9) were significant
predictors of CVE [23], while other studies have reported
similar associations even after adjusting for possible con-
founding variables [21, 44, 51, 90-93]. Smoking has also
been associated with an increased risk for CVD (HR ranging
from 1.9 to 8.6) and subclinical atherosclerosis [46, 89, 94,
95].
4.3. The Role of Treatment
4.3.1. Glucocorticoids
Lupus patients are frequently treated with immunosup-
pressive agents and glucocorticoids (GCs) for prolonged
time periods. The use of GCs, in particular, has been linked
to an increased risk for atherosclerosis and higher rates of
CVEs [21]. However, the association between GCs and CVD
may be confounded by many factors intrinsic to active dis-
eases such as vascular inflammation and certain comorbid-
ities. Several studies have evaluated the impact of GCs both
on CVEs and subclinical atherosclerosis with inconsistent
results [9, 30, 96-98]. The discrepancies may be partly ex-
plained by the difference in the assessment of drug exposure
(current dose, cumulative dose, duration of treatment) and
differences in the sampled population (e.g. lupus nephritis).
Both higher cumulative GC intake and longer duration of
treatment have been strongly associated with accelerated
atherosclerosis [30, 96, 97]. Lupus patients treated with 10
mg of prednisone equivalent daily for 10 years had substan-
tially increased CVE risk, which was further augmented
when the daily dose exceeded 20 mg [21]. In contrast, Roma
et al. showed that a higher progression of carotid plaque was
associated with a lower average dose of GCs, pointing out a
possible atheroprotective effect [9]. Notably, a study con-
ducted in a paediatric SLE population revealed a decreased
cIMT in patients treated with moderate doses of GCs com-
pared with those treated with low or high GC doses [98].
GCs have multiple effects on immune-mediated path-
ways which are commonly involved in the pathogenesis of
atherosclerosis. However, little is known about the endothe-
lial modifications in response to GCs, especially in the con-
text of SLE. In experimental models, normal mice treated
with GCs exhibited increased endothelial dysfunction [99,
100]. A possible explanation involves endothelial nitric ox-
ide synthase, a molecule with an atheroprotective profile. In
this model, increased ROS production induced by GCs
down-regulated endothelial nitric oxide synthase mRNA and,
consequently, reduced its bioavailability [101]. An interest-
ing study demonstrated that the deletion of macrophage-
specific GC receptor reduced vascular calcification without
affecting atherosclerotic lesion size [102], highlighting the
role of GCs on vascular calcification. On the other hand,
GCs may have presumed atheroprotective effects by inhibit-
ing the expression of adhesion molecules, mainly Intercellu-
lar Adhesion Molecule 1 (ICAM-1) and vascular cell adhe-
sion molecule-1 (VCAM-1), resulting in decreased cell mi-
gration at the side of plaque [103]. Moreover, GCs inhibit
the nuclear factor kappa B (NF-κB) [102]; a molecule, which
is activated at different stages of atherosclerosis participating
in underlying pathogenesis [104]. Another group treated an
atherosclerotic murine model with dexamethasone, and ob-
served inhibition of proliferation and recruitment of macro-
phages along with decreased aortic foam cell formation
[105].
Although the effect of GCs per se on atherosclerosis re-
mains to be better defined, the long-term exposure to GCs
culminates in the accumulation of various comorbidities in-
cluding hypertension, dyslipidaemia, glucose intolerance and
central obesity [106]. Thus, most of the pro-atherogenic ef-
fects of GCs may be driven by the aggregation of such co-
morbidities.
4.3.2. Antimalarials
Hydroxychloroquine (HCQ) is the cornerstone of lupus
care due to its multiple beneficial effects including decreased
CVD risk and protection against both arterial and venous
thrombosis along with a lower burden of atherosclerotic
plaques [107-109]. Several clinical studies have reported on
the atheroprotective role of HCQ [4]. Its main effects are
mediated by anti-inflammatory and lipid-lowering properties
as well as its ability to control the glycaemic profile [110-
112]. A study utilising lupus-prone mice (NZB/NZW F1
strain), demonstrated that early administration of HCQ in-
creases nitric oxide availability and improves the function of
endothelium via reduction of ROS [113]. Moreover, antima-
larials have been shown to reduce type I IFN production by
plasmacytoid dendritic cells [114], which may represent an-
other atheroprotective mechanism as IFN is a mediator of
vascular damage in lupus [55]. Another positive effect of
HCQ on prevention of CVD may be mediated by the im-
pediment to NET formation [115], as NETs contribute to
plaque development [116]. Interestingly, HCQ has been
shown to inhibit platelet activation and aggregation induced
by aPL antibodies, suggesting an antithrombotic role [117].
4.3.3. Mycophenolate Mofetil (MMF)
The direct effect of MMF on atherosclerotic lesions has
been evaluated in mice and human studies. Specifically, in
Ldlr–/– lupus-prone mice that are prone to atherosclerosis, the
administration of MMF resulted in inhibition of T-cell infil-
tration and activation in atherosclerotic plaques [118].
Moreover, in non-lupus atherogenic mice, treatment with
MMF has been shown to reduce atherosclerosis [119]. One
study in SLE patients who underwent carotid endarterectomy
demonstrated a reduced number of infiltrated activated T-
6 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
cells along with elevated levels of regulatory T-cells at the
atherosclerotic lesion [120]. Furthermore, there was also a
reduced metalloproteinase and pro-inflammatory genes ex-
pression. Nevertheless, Kiani et al. failed to detect any bene-
ficial effect of MMF on either progression of subclinical
atherosclerosis or coronary calcification in a longitudinal
SLE cohort study [121]. In addition, Sazliyana et al. did not
find any significant decrease in cIMT in association with the
duration of treatment or the cumulative dose of MMF [122].
4.3.4. Other Immunosuppressive Drugs
Methotrexate has been evaluated for the prevention of
atherosclerotic events in healthy individuals without yielding
positive results [123]. However, methotrexate seems to be
protective against atherosclerosis in rheumatoid arthritis,
which represents another chronic autoimmune inflammatory
disorder. A meta-analysis has underscored its putative
atheroprotective role in rheumatoid arthritis patients by 21%
reduction in the overall incidence of CVD and in particular,
an 18% reduction in the risk of MI [124]. Several studies
have shown that use of azathioprine is associated with an
increased CV burden [93, 98, 122, 125, 126]. However, these
findings may reflect the systemic inflammation in these pa-
tients. In contrast, one study demonstrated that the use of
cyclosporine may be beneficial in reducing atherosclerosis
progression as measured by cIMT [122, 127]. Furthermore,
carotid plaque formation has been negatively correlated with
the use of cyclophosphamide, thus highlighting a possible
atheroprotective effect, presumably through reduction of the
systemic inflammation burden [9].
4.3.5. Biologic Drugs
Rituximab, an anti-CD20 monoclonal antibody targeting
B-cells, has been introduced as an off label option in the
therapeutic armamentarium of severe and refractory lupus
[128]. Rituximab seems to be protective against premature
atherosclerosis by affecting the total cholesterol/HDL-C ra-
tio, HDL-C and levels of tissue factor [129]. These findings
may reflect the attenuation of systemic inflammation medi-
ated by B-cell depletion. An interesting study conducted in
atherosclerosis-prone mice (apoE/ and Lldlr/ strains)
treated with a CD20-specific monoclonal antibody [130],
demonstrated significantly decreased atherosclerosis along
with decreased titers of anti-oxLDL antibodies. Moreover,
reduced numbers of infiltrating B-cells, T-lymphocytes and
macrophages were detected in atherosclerotic lesions in the
aforementioned mice.
Belimumab is a monoclonal antibody against B-cell acti-
vated factor (BAFF), inhibiting B-cell maturation and sur-
vival, and is approved for the management of SLE [131].
Two interesting studies conducted in hyperlipidemic apoE/
mice showed that both the treatment with antibodies directed
against the BAFF receptor (BAFF-R) and BAFF-R defi-
ciency (Baffr/ mice) led to reduced atherosclerosis [132,
133]. BAFF-R inhibition neutralizes the B2 cells subtype,
which is atherogenic, while preserving the B1 cells that are
considered atheroprotective [131, 133]. Furthermore, be-
limumab may exert a protective role against aPL syndrome
development [134]. It is worth noting that aPL seroconver-
sion from positive to negative levels has been observed in
SLE patients treated with belimumab [135]. These data sug-
gest that belimumab might be beneficial in the prevention of
aPL-medicated thrombotic events and accelerated atheroscle-
rosis. Nevertheless, belimumab efficacy has been shown to
be weakened in patients with previous thrombotic events
[136-138], necessitating further survey of the impact of be-
limumab use on primary and secondary CVD prevention
separately.
Anifrolumab, a monoclonal antibody against type I IFN
receptor [139], may be a promising agent in preventing sub-
clinical atherosclerosis in view of the pro-atherogenic prop-
erties of type I IFNs. To this end, other biologics such as
anti-TNF and anti-IL6 are under investigation as atheropro-
tective agents in patients with rheumatoid arthritis [140].
5. SELECTED CVD MANIFESTATIONS IN PA-
TIENTS WITH SLE
5.1. Coronary Heart Disease (CHD)
In recent studies, the prevalence of clinically significant
CHD in SLE varies from 2-14% [20, 141-143]. Incidence
rates of MI are significantly raised in lupus patients confer-
ring at least a 2-fold risk compared with the general popula-
tion. In a study that comprised 4,863 individuals with SLE,
the adjusted HR for MI was 2.61 compared to non-SLE con-
trols [29]. This risk was even more marked during the first
year of diagnosis and in younger women, highlighting the
premature onset of atherosclerosis [28, 29, 90]. Race and
ethnic variation may contribute to discrepant MI rates across
different lupus cohorts. In a recent study including 65,788
cases of SLE with wide race and ethnic distribution, there
was a reduced MI risk among Hispanics and Asians com-
pared with Caucasian SLE patients (HR: 0.61 and HR: 0.57,
respectively) [144]. Notably, the increased risk seen in SLE
patients seems to be extended to long-term poorer outcomes
and increased cause-specific mortality. Although in-hospital
post-MI outcomes do not differ between SLE patients and
controls [145, 146], patients with SLE are more likely to
experience a new MI or require repeat percutaneous coro-
nary intervention within 1 year after the initial event [147].
Moreover, long-term data suggest that SLE patients with MI
are at least 2.6 times more likely to die than non-SLE pa-
tients with the same coronary event [148, 149].
5.2. Cerebrovascular Disease
Individuals with SLE have an increased risk for cere-
brovascular disease compared with the general population
[29, 150, 151]. In a recent meta-analysis of 5 studies, the
pooled relative risk of stroke in SLE patients was 2.53 [152].
Similar to CHD, the risk of cerebrovascular disease is high-
est early during the course of the disease and amongst indi-
viduals younger than 50 years [29, 150-153]. Race and eth-
nicity seem to modify the risk with Blacks and Hispanics
being more susceptible to the development of stroke com-
pared with Caucasian SLE patients (HR 1.31) [144]. Studies
on immediate post-ischemic stroke mortality failed to prove
a significant difference between SLE and non-SLE subjects
[145, 154]. However, Rossides et al. recently reported an
increased late mortality risk (after the first month of an
ischemic stroke) (HR 1.85) and an even greater risk of death
in cases of haemorrhagic stroke expanded even before the
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 7
first month from the occurrence of the event (HR 2.30)
[154].
5.3. Peripheral Artery Disease
The prevalence of peripheral artery disease in SLE as-
sessed by the ankle-brachial index ranges between 21% and
33% [155-157]. In a population-based cohort study of 10,144
patients with SLE and 10,144 controls, the risk of peripheral
artery disease was higher amongst the former (HR 9.39)
even after adjustment for traditional risk factors and GC use.
In the same study, younger (34 years old) and newly diag-
nosed SLE patients had a strikingly increased risk compared
to age-matched controls (HR 47.6 and 32.0, respectively)
[158]. Moreover, age seems to modify the effect of tradi-
tional risk factors on peripheral artery disease risk, thus ex-
plaining the discordance between studies evaluating predic-
tors of the disease in different age groups.
6. ASSESSMENT OF CVD RISK IN PATIENTS WITH
SLE
6.1. Traditional CVD Risk Factors and Risk Stratifica-
tion Scores
There are several validated risk prediction tools (e.g.
Framingham Risk Score, Systematic COronary Risk Evalua-
tion (SCORE), QRISK which uses multiple risk factors) that
quantify CVD risk and guide preventive measures in the
general population. However, their performance in SLE pa-
tients is considered suboptimal due to underestimating the
actual risk [27, 41, 46, 159, 160]. Modifications on the
aforementioned conventional” risk tools, either by incorpo-
rating SLE as a separate risk factor [161] or by introducing
“multipliers” to increase the calculated score [162, 163],
have been suggested to provide a more accurate risk esti-
mate. An additional data-driven risk equation designed by
Petri et al. [164] used SLE-specific predictors of CVEs from
the Hopkins Lupus Cohort to formulate a more oriented tool.
Although extensive validation studies are lacking, a recent
comparative assessment between different risk scores in
1,887 SLE patients, showed that the modified (multiplied by
a factor of 2) Framingham risk score outperformed in terms
of both usability and sensitivity/specificity (46% and 83%,
respectively) [160].
6.2. Non-Invasive Methods for CVD Assessment
6.2.1. Coronary Artery Assessment
CAC assessed by electron beam or multislice computed
tomography, is an indicator of coronary atherosclerosis and
is associated with increased CVD risk in the general popula-
tion [165]. The prevalence of CAC is higher in SLE subjects
compared with age-matched healthy controls, even after ad-
justment for traditional risk factors [166]. In SLE subjects
followed over a 2-year period, CAC progression correlated
with age, smoking and total cholesterol levels [167].
Whether the CAC progression rate is more accelerated in
SLE and its link to future CVE has yet to be defined.
Other modalities of CV imaging such as Single Photon
Emission Computerized Tomography (SPECT) and Mag-
netic Resonance Imaging (MRI) have been used to detect
perfusion defects and coronary atherosclerosis in SLE pa-
tients [168]. Plazak et al. compared echocardiography, CAC
and SPECT findings in 50 SLE patients and found that
SPECT could reveal myocardial perfusion defects even in
patients without evident CAC. This discrepancy could be due
to that SPECT may additionally show ischemia caused by
thrombosis or vasculitis, which cannot be detected by CAC
[169]. Similarly, Varma et al. in a study evaluating cardiac
magnetic resonance (cMR) found that SLE patients had a
diffuse contrast enhancement pattern indicative of both in-
flammation and atherosclerotic plaque stability [170].
6.2.2. Carotid Plaques and cIMT
The most frequently used modality for assessing and
monitoring subclinical atherosclerosis in SLE is carotid ul-
trasound. Risk stratification through ultrasound focuses on
the detection of carotid plaques and measures of cIMT.
While these indices have been linked to increased CVD haz-
ard both in the general and the SLE population [40, 171], the
presence of plaques is generally considered a powerful pre-
dictor of future CVEs [162]. The higher prevalence of ca-
rotid plaques and increased cIMT in SLE patients has been
verified in a series of studies and meta-analyses [39, 172,
173]. Interestingly, even SLE patients without CVD history
have been shown to have significantly elevated measures of
subclinical atherosclerosis in agreement with a higher CVD
risk [40].
6.2.3. Endothelial Dysfunction and Vascular Stiffness
Endothelial dysfunction, a potentially reversible loss of
vascular homeostasis, is an early event in the atherogenic
process and precedes the formation of atherosclerotic
plaques [174]. Impairment of endothelial integrity is difficult
to be detected through non-invasive techniques, however,
endothelial-dependent flow-mediated vasodilation, measured
in the brachial artery by high-resolution ultrasound is fre-
quently used for the assessment of endothelial dysfunction
[175]. In SLE patients, the flow-mediated vasodilation is
significantly lower compared with matched controls as a
result of both traditional and disease-related risk factors
[176, 177]. In a recent meta-analysis of 25 case-control stud-
ies, the presence of DM, higher diastolic blood pressure and
renal involvement were all associated with impaired vasodi-
lation in SLE subjects [177]. Unlike the assessment of cIMT
and plaques, the validity of flow-mediated vasodilation as a
prognostic tool has not been tested in prospective cohorts,
therefore limiting its usefulness in CVD risk assessment.
In the general population, arterial stiffness measured by
PWV has been associated with an increased risk of CVD and
serves as an indicator of vascular damage [162]. Many stud-
ies have reported higher PWV measurements in SLE pa-
tients, however, absolute values may vary depending on the
modality used and the sampled population [110, 178, 179].
Although the predictive value of PWV regarding CVD in
SLE is still unknown, a recent cross-sectional study reported
that patients with higher measurements of PWV had an in-
creased CVD risk [180].
6.3. Novel Biomarkers
A number of soluble molecules associated with CVD in
SLE can potentially serve as predictive biomarkers. A com-
8 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
prehensive review identified >20 biomarkers associated with
CVEs, CVD mortality and premature atherosclerosis in pa-
tients with SLE (Table 3) [116]. In addition, the hypothesis
that an accurate risk stratification would probably require a
combination of molecules rather than a single biomarker has
recently been tested. In 2014, McMahon et al. found that a
panel of biomarkers comprising 4 inflammatory (inflamma-
tory HDL, leptin, homocysteine and TNF-like weak inducer
of apoptosis) and 2 traditional (age and diabetes) risk factors
yielded a better predictive value than individual biomarkers
and traditional risk factors on detecting subclinical athero-
sclerosis in SLE patients (for high-risk patients positive pre-
dictive value was 64%, negative predictive value was 94%,
sensitivity was 89%, and specificity was 79%) [181].
Moreover, a number of immune mediators found to be inde-
pendently associated with both CVD and SLE are currently
under investigation as surrogate predictive biomarkers for
atherosclerosis in SLE patients [182]. Certain circulating
immunometabolites found to be increased in SLE, promote
the activation of the inflammasomes (large multiprotein
complexes that serve as key signalling platforms) [183]. Fol-
lowing activation, inflammasomes cause the activation of
caspase-1 resulting in aberrant IL-1β production [184].
However, inhibition of the latter has been associated with
lower CVD risk in the general population [183]. Therefore,
immunometabolites could serve as putative biomarkers for
further stratification of SLE patients.
Since neutrophils are known to play a central role in the
pathogenesis of both atherosclerosis and SLE, proteins se-
creted from activated neutrophils (i.e. S100A8, A9, A8/9,
and A12) may also be considered candidate biomarkers of
CVD development in patients with SLE; these proteins have
been shown to be associated with atherosclerosis and in-
flammation [182, 185]. Similarly, follicular helper T-cells
have been shown to be implicated in the pathogenesis of
both atherosclerosis and SLE [186]. Polymorphisms of
OX40L, a trigger that promotes the activation of these cells,
have also been associated with increased susceptibility to
lupus and atherosclerosis [187]. Additional studies may
promote the utilisation of these gene polymorphisms as
novel biomarkers. Although these candidate biomarkers
might be promising, further investigations are needed to
validate these hypotheses.
7. CVD PREVENTION IN PATIENTS WITH SLE
The increased CV burden seen in SLE patients empha-
sizes the need for primary and secondary prevention strate-
gies. However, to date, there is no high-quality evidence to
guide CVD risk management. The majority of prevention
trials on SLE population have used repeated measures of
subclinical atherosclerosis and modifications of traditional
CVD risk factors as primary outcomes rather than the actual
CVD incidence, which is a clear limitation to be corrected in
future trial design. That would necessitate joint multicentre
efforts.
7.1. Non-Pharmacological Interventions and Counselling
Smoking cessation, avoidance of sedentary life and main-
tenance of a normal body mass index have all been associ-
ated with a better cardiovascular risk profile in the general
Table 3. Novel biomarkers and associations with cardiovascu-
lar disease (CVD) in patients with SLE.
Biomarker
Association with CVD
PC4d
Mortality and CVEs
CRP
Mortality and CVEs
Asymmetric dimethyl-arginine
CVEs and atherosclerosis
Homocysteinemia
CVEs and atherosclerosis
VEGF
CVEs and atherosclerosis
Whole blood viscosity
CVEs and atherosclerosis
TWEAK
CVEs and atherosclerosis
Type I interferon
CVEs
VWf
CVEs
Interleukin-6
CVEs
sCD40L
CVEs
Annexin A5
Atherosclerosis
AntiapoAI
Atherosclerosis
C3 complement
Atherosclerosis
Endocan
Atherosclerosis
Eselectin
Atherosclerosis
Fattyacidbinding protein 4
Atherosclerosis
ICAM1
Atherosclerosis
piHDL
Atherosclerosis
VCAM-1
Atherosclerosis
PC4d, Platelets bearing complem ent protein C4d; CVE, cardiovascular event; CRP, C-
reactive protein; VEGF, vascular endothelial growth factor; sCD40L , soluble CD40
ligand; TWEAK, tumor necrosis factor-like weak inducer of apoptosis; Anti-apoA-I,
anti- apolipoprotein A1; ICAM1, intercellular adhesion molecule 1; piHDL, pro-
inflammatory high-density lipoprotein; VCAM, vascular cell adhesion molecule-1.
population. In SLE patients, data regarding the effect of
smoking cessation on CVD hazards are indirect and mostly
extrapolated from studies identifying smoking as a CVD risk
factor [188]. Similarly, indirect evidence from studies on
surrogate markers of CVD suggests that physical exercise
and dietary supplementation with low-dose omega-3-
polyunsaturated fatty acids may reverse endothelial dysfunc-
tion [189, 190].
7.2. Pharmacological Interventions
7.2.1. Statins
Statins are potent inhibitors of the biosynthesis of choles-
terol that have been widely used for CVD management. In
addition to their lipid-lowering effect, they have several plei-
otropic and anti-inflammatory properties. Therefore, it was
reasonably hypothesized that SLE patients could benefit
from their dual effect both on atherosclerosis and disease
activity [191]. However, studies in SLE patients have pro-
vided inconsistent results and the protective role of statins
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 9
against CVD has been disputed. To date, there are no high
quality randomized controlled trials (RCTs) that have tested
or currently test the long-term effect of statins on hard out-
comes such as MI, stroke and CVD mortality in SLE. Re-
sults from 2 longitudinal studies (1 retrospective and 1 small
post-hoc analysis of an RCT) suggested a protective effect of
statins against CVEs and mortality in SLE patients [192,
193]. In line with the previous findings, patients treated with
atorvastatin had a lower rate of progression of atherosclero-
sis in 2 RCTs [194, 195]. In contrast, in a double-blinded
RCT of 200 SLE patients who were randomized to receive
atorvastatin or placebo, the progression of CAC, cIMT or
carotid plaque did not differ in the 2 groups at the end of a 2-
year follow up [167]. Similar results were obtained from
another RCT where the administration of rosuvastatin was
not found to reduce cIMT progression, and no significant
differences were observed between the treatment and control
arms [196].
7.2.2. Aspirin
There are no prospective studies of SLE patients that
examine the effect of aspirin on CVD management. How-
ever, in a retrospective study of 291 SLE patients followed
over a period of 8 years, aspirin use was found to be benefi-
cial regarding CVD prevention (HR: 0.24) [50]. For the sub-
group of SLE patients with aPL antibody positivity, a recent
meta-analysis found a significant protective effect of aspirin
against thrombotic events (pooled OR: 0.55) [197]. Given
the lack of robust evidence and the increased bleeding haz-
ard, aspirin use is recommended only for selected SLE pa-
tients with a high-risk aPL profile as defined by Tektonidou
et al. [128, 198].
7.2.3. Hydroxychloroquine (HCQ)
HCQ has been associated with a decreased clinical and
subclinical CVD hazard in a series of SLE cohorts [50, 51,
90, 109, 199]. In a recent retrospective study of SLE patients
without previous CVEs, long-term HCQ use (>5 years) was
protective against thrombosis (HR: 0.27) even after adjust-
ment for other treatments [50]. Moreover, there is increasing
evidence for a metabolic beneficial effect both in SLE and
rheumatoid arthritis patients. In a recent meta-analysis of 717
SLE patients, HCQ use was associated with a significant
decrease in total cholesterol, triglycerides, LDL-C and very-
low-density lipoprotein (VLDL) [200]. A similar effect on
lipid profile and a lower incidence of DM in HCQ users has
been recently reported in another meta-analysis of 16 studies
in patients with rheumatoid arthritis [201]. Taken together,
current evidence indicates a possible beneficial effect of
HCQ on CVD burden. However, additional trials are war-
ranted in order to validate the role of HCQ in primary and
secondary CVD prevention. An ongoing RCT
(NCT02648464) that has randomized 2500 patients with MI
to receive HCQ or placebo will compare CVE rates and de-
termine the atheroprotective role of HCQ on a wider scale.
8. RECOMMENDATIONS FOR CLINICAL PRAC-
TICE
Despite growing recognition of the CVD burden in SLE,
there is a paucity of well-designed controlled evidence to
guide management in everyday clinical practice. The 2019
update of the European League Against Rheumatism rec-
ommendations on the management of SLE underscore that
patients should be regularly assessed for both traditional and
disease-related risk factors, particularly disease activity and
duration, aPL antibodies and chronic use of glucocorticoids
[128]. Renal involvement, in particular, active nephritis typi-
cally evidenced by increased proteinuria, reduced glomerular
filtration rate, anaemia, hypalbuminaemia and hypertension,
represents a strong risk factor for accelerated atherosclerosis.
A second statement [128] in SLE recommendations under-
lines the possible use of low-dose aspirin and lipid-lowering
agents according to individual CVD risk profiles. Still, a
number of pertinent issues remain to be defined in SLE pa-
tients, such as the appropriate timing, frequency and means
for CVD risk assessment, the indications of such evaluations
and interpretation of non-invasive tools to detect subclinical
atherosclerosis, as well as optimal thresholds for traditional
atherosclerotic risk factors (e.g. LDL-C levels).
Until such data become available, one might extrapolate
from the existing recommendations for the general popula-
tion [202] while taking into consideration specific disease-
associated risk enhancers’ [203]. In this respect, screening
for traditional atherosclerotic risk factors should begin early
during the disease course since many CVD events tend to
occur within the first few years due to high inflammatory
burden. Individual 10-year risk should be calculated accord-
ing to one of the existing tools (e.g. SCORE); however, ro-
bust validation in SLE patients is missing. The risk may be
further modified depending on the presence or not of lupus-
specific (e.g. chronic exposure to glucocorticoids) or other
risk-enhancing factors (e.g. family history of premature
CVD) (Fig. 1). Pending confirmation in large prospective
studies, the majority of SLE patients should be considered as
“moderate” or “high” risk patients and thus, preventative
strategies should be applied accordingly. Similar to the gen-
eral population, these include lifestyle modifications (e.g.
tobacco cessation, physical exercise), blood pressure control,
lipid-lowering agents and in selected cases (e.g. aPL-positive
patients with moderate/high CVD risk), and/or low-dose
aspirin. Although the effectiveness of these strategies in re-
ducing CVD events has not been established for SLE pa-
tients specifically, it is reasonable to assume they are equally
efficacious as in the general population. In case of uncer-
tainty over the benefit/risk ratio of pharmacological interven-
tions, non-invasive methods of CVD assessment (e.g. coro-
nary artery calcium) may be helpful for risk stratification.
Finally, focus should be directed on inflammation control
targeting to remission or the lowest achievable disease activ-
ity, use of HCQ (unless contraindicated), along with mini-
mizing (or completely withdrawing) glucocorticoids (Fig. 1).
CONCLUSIONS
Patients with SLE are at a substantially (2- to 10-fold)
increased risk for the development of CVD events, which
may occur both early and later during the course of the dis-
ease and contribute to increased morbidity and mortality.
This risk is driven by both general and disease-specific fac-
tors such as inflammatory mediators, autoantibodies and the
deleterious effects of glucocorticoids. Accordingly, SLE
10 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
patients warrant a regular assessment of their CV risk and
continuous consideration of general and disease-specific
risk-lowering strategies. Importantly, the collaboration be-
tween different disciplines (rheumatologists, internists, car-
diologists, nephrologists and general practitioners) is desir-
able to fine-tune risk stratification and optimize pharmacol-
ogical treatments. We anticipate the emergence of new data
pertaining to the indications and interpretation of non-
invasive tools and biomarkers for CVD risk assessment, as
well as the efficacy of risk-lowering strategies tailored to
SLE.
LIST OF ABBREVIATIONS
Anti-β2-GPI = Anti-β2-Glycoprotein I
aPL = Antiphospholipid
CAC = Coronary Artery Calcium
CHD = Coronary Heart Disease
CV = Cardiovascular
CVD = Cardiovascular Disease
Fig. (1). Cardiovascular risk assessment, stratification and preventative measures in patients with SLE. A1C, glycosylated haemoglobin
A1C; CKD, chronic kidney disease; CVD, cardiovascular disease; FRS, Framingham Risk Score; GC, glucocorticoids; HCQ, hydroxy-
chloroquine; hsCRP, high-sensitivity C-reactive protein; LDA, low disease activity; LDL-C, Low-density lipoprotein cholesterol; LPa, lipo-
protein-a; OGTT, oral-glucose tolerance test; PG, plasma glucose; SCORE, Systematic COronary Risk Evaluation; T2DM, Type 2 diabetes
mellitus. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 11
CVE = Cardiovascular Event
DM = Diabetes Mellitus
GC = Glucocorticoid
HDL-C = High-Density Lipoprotein Cholesterol
HR = Hazard Ratios
HCQ = Hydroxychloroquine
ICAM-1 = Intercellular Adhesion Molecule 1
IFN = Type I Interferon
MI = Myocardial Infarction
NETs = Neutrophil Extracellular Traps
NF-κB = Nuclear Factor Kappa B
oxLDL = Oxidized Low-Density-Lipoprotein
PWV = Pulse Wave Velocity
ROS = Reactive Oxygen Species
SLE = Systemic Lupus Erythematosus
TNF-α = Tumour Necrosis Factor-α
VCAM-1 = Vascular Cell Adhesion Molecule-1
VLDL = Very Low Density Lipoprotein
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
None.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Tsokos GC. Systemic lupus erythematosus. N Engl J Med 2011;
365(22): 2110-21.
http://dx.doi.org/10.1056/NEJMra1100359 PMID: 22129255
[2] Rúa-Figueroa I, Fernández Castro M, Andreu JL, et al. Sjogrenser and
Relesser Researchers and EAS-SER Group. Comorbidities in Patients
With Primary Sjögren’s Syndrome and Systemic Lupus Erythemato-
sus: A Comparative Registries-Based Study. Arthritis Care Res
(Hoboken) 2017; 69(1): 38-45.
http://dx.doi.org/10.1002/acr.23015 PMID: 27564390
[3] Barnado A, Carroll RJ, Casey C, Wheless L, Denny JC, Crofford LJ.
Phenome-wide association study identifies marked increased in burden
of comorbidities in African Americans with systemic lupus erythema-
tosus. Arthritis Res Ther 2018; 20(1): 69.
http://dx.doi.org/10.1186/s13075-018-1561-8 PMID: 29636090
[4] Tselios K, Gladman DD, Sheane BJ, Su J, Urowitz M. All-cause,
cause-specific and age-specific standardised mortality ratios of pa-
tients with systemic lupus erythematosus in Ontario, Canada over 43
years (1971-2013). Ann Rheum Dis 2019; 78(6): 802-6.
http://dx.doi.org/10.1136/annrheumdis-2018-214802 PMID: 30992296
[5] Hansson GK. Inflammation, atherosclerosis, and coronary artery dis-
ease. N Engl J Med 2005; 352(16): 1685-95.
http://dx.doi.org/10.1056/NEJMra043430 PMID: 15843671
[6] Ahmadi A, Argulian E, Leipsic J, Newby DE, Narula J. From Sub-
clinical Atherosclerosis to Plaque Progression and Acute Coronary
Events: JACC State-of-the-Art Review. J Am Coll Cardiol 2019;
74(12): 1608-17.
http://dx.doi.org/10.1016/j.jacc.2019.08.012 PMID: 31537271
[7] Kutom AH, Gibbs HR. Myocardial infarction due to intracoronary
thrombi without significant coronary artery disease in systemic lupus
erythematosus. Chest 1991; 100(2): 571-2.
http://dx.doi.org/10.1378/chest.100.2.571 PMID: 1864145
[8] Asanuma Y, Oeser A, Shintani AK, et al. Premature coronary-artery
atherosclerosis in systemic lupus erythematosus. N Engl J Med 2003;
349(25): 2407-15.
http://dx.doi.org/10.1056/NEJMoa035611 PMID: 14681506
[9] Roman MJ, Shanker BA, Davis A, et al. Prevalence and correlates of
accelerated atherosclerosis in systemic lupus erythematosus. N Engl J
Med 2003; 349(25): 2399-406.
http://dx.doi.org/10.1056/NEJMoa035471 PMID: 14681505
[10] Gustafsson JT, Svenungsson E. Definitions of and contributions to
cardiovascular disease in systemic lupus erythematosus. Autoimmu-
nity 2014; 47(2): 67-76.
http://dx.doi.org/10.3109/08916934.2013.856005 PMID: 24228980
[11] Lane HA, Smith JC, Davies JS. Noninvasive assessment of preclinical
atherosclerosis. Vasc Health Risk Manag 2006; 2(1): 19-30.
http://dx.doi.org/10.2147/vhrm.2006.2.1.19 PMID: 17319466
[12] Soltész P, Kerekes G, Dér H, et al. Comparative assessment of vascu-
lar function in autoimmune rheumatic diseases: considerations of pre-
vention and treatment. Autoimmun Rev 2011; 10(7): 416-25.
http://dx.doi.org/10.1016/j.autrev.2011.01.004 PMID: 21281743
[13] Matsuzawa Y, Kwon TG, Lennon RJ, Lerman LO, Lerman A. Prog-
nostic Value of Flow-Mediated Vasodilation in Brachial Artery and
Fingertip Artery for Cardiovascular Events: A Systematic Review and
Meta-Analysis. J Am Heart Assoc 2015; 4(11)e002270
http://dx.doi.org/10.1161/JAHA.115.002270 PMID: 26567372
[14] Kim HL, Kim SH. Pulse Wave Velocity in Atherosclerosis. Front
Cardiovasc Med 2019; 6: 41.
http://dx.doi.org/10.3389/fcvm.2019.00041 PMID: 31024934
[15] Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA,
Ogryzlo MA. The bimodal mortality pattern of systemic lupus erythe-
matosus. Am J Med 1976; 60(2): 221-5.
http://dx.doi.org/10.1016/0002-9343(76)90431-9 PMID: 1251849
[16] Ståhl-Hallengren C, Jönsen A, Nived O, Sturfelt G. Incidence studies
of systemic lupus erythematosus in Southern Sweden: increasing age,
decreasing frequency of renal manifestations and good prognosis. J
Rheumatol 2000; 27(3): 685-91.
PMID: 10743809
[17] Alamanos Y, Voulgari PV, Papassava M, Tsamandouraki K, Drosos
AA. Survival and mortality rates of systemic lupus erythematosus pa-
tients in northwest Greece. Study of a 21-year incidence cohort.
Rheumatology (Oxford) 2003; 42(9): 1122-3.
http://dx.doi.org/10.1093/rheumatology/keg291 PMID: 12923275
[18] Mok CC, Tse SM, Chan KL, Ho LY. Effect of the metabolic syn-
drome on organ damage and mortality in patients with systemic lupus
erythematosus: a longitudinal analysis. Clin Exp Rheumatol 2018;
36(3): 389-95.
PMID: 29148424
[19] Tselios K, Gladman DD, Su J, Ace O, Urowitz MB. Evolution of Risk
Factors for Atherosclerotic Cardiovascular Events in Systemic Lupus
Erythematosus: A Longterm Prospective Study. J Rheumatol 2017;
44(12): 1841-9.
http://dx.doi.org/10.3899/jrheum.161121 PMID: 29093154
[20] Bertoli AM, Vilá LM, Alarcón GS, et al. PROFILE Study Group.
Factors associated with arterial vascular events in PROFILE: a Multi-
ethnic Lupus Cohort. Lupus 2009; 18(11): 958-65.
http://dx.doi.org/10.1177/0961203309104862 PMID: 19762396
[21] Magder LS, Petri M. Incidence of and risk factors for adverse cardio-
vascular events among patients with systemic lupus erythematosus.
Am J Epidemiol 2012; 176(8): 708-19.
http://dx.doi.org/10.1093/aje/kws130 PMID: 23024137
[22] Pons-Estel GJ, González LA, Zhang J, et al. Predictors of cardiovascu-
lar damage in patients with systemic lupus erythematosus: data from
LUMINA (LXVIII), a multiethnic US cohort. Rheumatology (Oxford)
2009; 48(7): 817-22.
http://dx.doi.org/10.1093/rheumatology/kep102 PMID: 19454606
12 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
[23] Ballocca F, D’Ascenzo F, Moretti C, et al. Predictors of cardiovascu-
lar events in patients with systemic lupus erythematosus (SLE): a sys-
tematic review and meta-analysis. Eur J Prev Cardiol 2015; 22(11):
1435-41.
http://dx.doi.org/10.1177/2047487314546826 PMID: 25139772
[24] Ward MM. Premature morbidity from cardiovascular and cerebrovas-
cular diseases in women with systemic lupus erythematosus. Arthritis
Rheum 1999; 42(2): 338-46.
http://dx.doi.org/10.1002/1529-0131(199902)42:2<338::AID-
ANR17>3.0.CO;2-U PMID: 10025929
[25] Bartels CM, Buhr KA, Goldberg JW, et al. Mortality and cardiovascu-
lar burden of systemic lupus erythematosus in a US population-based
cohort. J Rheumatol 2014; 41(4): 680-7.
http://dx.doi.org/10.3899/jrheum.130874 PMID: 24532834
[26] Hak AE, Karlson EW, Feskanich D, Stampfer MJ, Costenbader KH.
Systemic lupus erythematosus and the risk of cardiovascular disease:
results from the nurses’ health study. Arthritis Rheum 2009; 61(10):
1396-402.
http://dx.doi.org/10.1002/art.24537 PMID: 19790130
[27] Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Traditional
Framingham risk factors fail to fully account for accelerated athero-
sclerosis in systemic lupus erythematosus. Arthritis Rheum 2001;
44(10): 2331-7.
http://dx.doi.org/10.1002/1529-0131(200110)44:10<2331::AID-
ART395>3.0.CO;2-I PMID: 11665973
[28] Manzi S, Meilahn EN, Rairie JE, et al. Age-specific incidence rates of
myocardial infarction and angina in women with systemic lupus
erythematosus: comparison with the Framingham Study. Am J Epide-
miol 1997; 145(5): 408-15.
http://dx.doi.org/10.1093/oxfordjournals.aje.a009122 PMID: 9048514
[29] Aviña-Zubieta JA, To F, Vostretsova K, De Vera M, Sayre EC, Es-
daile JM. Risk of Myocardial Infarction and Stroke in Newly Diag-
nosed Systemic Lupus Erythematosus: A General Population-Based
Study. Arthritis Care Res (Hoboken) 2017; 69(6): 849-56.
http://dx.doi.org/10.1002/acr.23018 PMID: 28129475
[30] Tektonidou MG, Kravvariti E, Konstantonis G, Tentolouris N, Sfika-
kis PP, Protogerou A. Subclinical atherosclerosis in Systemic Lupus
Erythematosus: Comparable risk with Diabetes Mellitus and Rheuma-
toid Arthritis. Autoimmun Rev 2017; 16(3): 308-12.
http://dx.doi.org/10.1016/j.autrev.2017.01.009 PMID: 28147263
[31] Bulkley BH, Roberts WC. The heart in systemic lupus erythematosus
and the changes induced in it by corticosteroid therapy. A study of 36
necropsy patients. Am J Med 1975; 58(2): 243-64.
http://dx.doi.org/10.1016/0002-9343(75)90575-6 PMID: 1115070
[32] Haider YS, Roberts WC. Coronary arterial disease in systemic lupus
erythematosus; quantification of degrees of narrowing in 22 necropsy
patients (21 women) aged 16 to 37 years. Am J Med 1981; 70(4): 775-
81.
http://dx.doi.org/10.1016/0002-9343(81)90532-5 PMID: 7211914
[33] de Leeuw K, Smit AJ, de Groot E, van Roon AM, Kallenberg CG, Bijl
M. Longitudinal study on premature atherosclerosis in patients with
systemic lupus erythematosus. Atherosclerosis 2009; 206(2): 546-50.
http://dx.doi.org/10.1016/j.atherosclerosis.2009.03.018 PMID:
19389674
[34] Haque S, Skeoch S, Rakieh C, et al. Progression of subclinical and
clinical cardiovascular disease in a UK SLE cohort: the role of classic
and SLE-related factors. Lupus Sci Med 2018; 5(1)e000267
http://dx.doi.org/10.1136/lupus-2018-000267 PMID: 30538814
[35] Kiani AN, Post WS, Magder LS, Petri M. Predictors of progression in
atherosclerosis over 2 years in systemic lupus erythematosus. Rheuma-
tology (Oxford) 2011; 50(11): 2071-9.
http://dx.doi.org/10.1093/rheumatology/ker285 PMID: 21875880
[36] Roman MJ, Crow MK, Lockshin MD, et al. Rate and determinants of
progression of atherosclerosis in systemic lupus erythematosus. Arthri-
tis Rheum 2007; 56(10): 3412-9.
http://dx.doi.org/10.1002/art.22924 PMID: 17907140
[37] Thompson T, Sutton-Tyrrell K, Wildman RP, et al. Progression of
carotid intima-media thickness and plaque in women with systemic lu-
pus erythematosus. Arthritis Rheum 2008; 58(3): 835-42.
http://dx.doi.org/10.1002/art.23196 PMID: 18311797
[38] Svenungsson E, Jensen-Urstad K, Heimbürger M, et al. Risk factors
for cardiovascular disease in systemic lupus erythematosus. Circula-
tion 2001; 104(16): 1887-93.
http://dx.doi.org/10.1161/hc4101.097518 PMID: 11602489
[39] Wu GC, Liu HR, Leng RX, et al. Subclinical atherosclerosis in pa-
tients with systemic lupus erythematosus: A systemic review and
meta-analysis. Autoimmun Rev 2016; 15(1): 22-37.
http://dx.doi.org/10.1016/j.autrev.2015.10.002 PMID: 26455562
[40] Kao AH, Lertratanakul A, Elliott JR, et al. Relation of carotid intima-
media thickness and plaque with incident cardiovascular events in
women with systemic lupus erythematosus. Am J Cardiol 2013;
112(7): 1025-32.
http://dx.doi.org/10.1016/j.amjcard.2013.05.040 PMID: 23827400
[41] Goldberg RJ, Urowitz MB, Ibañez D, Nikpour M, Gladman DD. Risk
factors for development of coronary artery disease in women with sys-
temic lupus erythematosus. J Rheumatol 2009; 36(11): 2454-61.
http://dx.doi.org/10.3899/jrheum.090011 PMID: 19833754
[42] Telles RW, Lanna CC, Sousa AJ, et al. Progression of carotid athero-
sclerosis in patients with systemic lupus erythematosus. Clin Rheuma-
tol 2013; 32(9): 1293-300.
http://dx.doi.org/10.1007/s10067-013-2264-9 PMID: 23620348
[43] Wang P, Mao YM, Zhao CN, et al. Increased Pulse Wave Velocity in
Systemic Lupus Erythematosus: A Meta-Analysis. Angiology 2018;
69(3): 228-35.
http://dx.doi.org/10.1177/0003319717715964 PMID: 28635303
[44] Fernández-Nebro A, Rúa-Figueroa Í, López-Longo FJ, et al. EAS-
SER (Systemic Diseases Study Group of Spanish Society of Rheuma-
tology). Cardiovascular Events in Systemic Lupus Erythematosus: A
Nationwide Study in Spain From the RELESSER Registry. Medicine
(Baltimore) 2015; 94(29)e1183
http://dx.doi.org/10.1097/MD.0000000000001183 PMID: 26200625
[45] Eder L, Gladman DD, Ibañez D, Urowitz MB. The correlation be-
tween carotid artery atherosclerosis and clinical ischemic heart disease
in lupus patients. Lupus 2014; 23(11): 1142-8.
http://dx.doi.org/10.1177/0961203314537696 PMID: 24876099
[46] Gustafsson JT, Simard JF, Gunnarsson I, et al. Risk factors for cardio-
vascular mortality in patients with systemic lupus erythematosus, a
prospective cohort study. Arthritis Res Ther 2012; 14(2): R46.
http://dx.doi.org/10.1186/ar3759 PMID: 22390680
[47] Baragetti A, Ramirez GA, Magnoni M, et al. Disease trends over time
and CD4+CCR5+ T-cells expansion predict carotid atherosclerosis de-
velopment in patients with systemic lupus erythematosus. Nutr Metab
Cardiovasc Dis 2018; 28(1): 53-63.
http://dx.doi.org/10.1016/j.numecd.2017.09.001 PMID: 29150407
[48] Karp I, Abrahamowicz M, Fortin PR, et al. Recent corticosteroid use
and recent disease activity: independent determinants of coronary
heart disease risk factors in systemic lupus erythematosus? Arthritis
Rheum 2008; 59(2): 169-75.
http://dx.doi.org/10.1002/art.23352 PMID: 18240259
[49] Barsalou J, Bradley TJ, Tyrrell PN, et al. Impact of Disease Duration
on Vascular Surrogates of Early Atherosclerosis in Childhood-Onset
Systemic Lupus Erythematosus. Arthritis Rheumatol 2016; 68(1): 237-
46.
http://dx.doi.org/10.1002/art.39423 PMID: 26361097
[50] Fasano S, Margiotta DP, Navarini L, et al. Primary prevention of
cardiovascular disease in patients with systemic lupus erythematosus:
case series and literature review. Lupus 2017; 26(14): 1463-72.
http://dx.doi.org/10.1177/0961203317722847 PMID: 28786768
[51] Iudici M, Fasano S, Gabriele Falcone L, et al. Low-dose aspirin as
primary prophylaxis for cardiovascular events in systemic lupus
erythematosus: a long-term retrospective cohort study. Rheumatology
(Oxford) 2016; 55(9): 1623-30.
http://dx.doi.org/10.1093/rheumatology/kew231 PMID: 27247433
[52] Rhoads JP, Major AS. How Oxidized Low-Density Lipoprotein Acti-
vates Inflammatory Responses. Crit Rev Immunol 2018; 38(4): 333-
42.
http://dx.doi.org/10.1615/CritRevImmunol.2018026483 PMID:
30806246
[53] Hallenbeck JM, Hansson GK, Becker KJ. Immunology of ischemic
vascular disease: plaque to attack. Trends Immunol 2005; 26(10): 550-
6.
http://dx.doi.org/10.1016/j.it.2005.08.007 PMID: 16109503
[54] Sherer Y, Shoenfeld Y. Mechanisms of disease: atherosclerosis in
autoimmune diseases. Nat Clin Pract Rheumatol 2006; 2(2): 99-106.
http://dx.doi.org/10.1038/ncprheum0092 PMID: 16932663
[55] Denny MF, Thacker S, Mehta H, et al. Interferon-alpha promotes
abnormal vasculogenesis in lupus: a potential pathway for premature
atherosclerosis. Blood 2007; 110(8): 2907-15.
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 13
http://dx.doi.org/10.1182/blood-2007-05-089086 PMID: 17638846
[56] Thacker SG, Zhao W, Smith CK, et al. Type I interferons modulate
vascular function, repair, thrombosis, and plaque progression in mur-
ine models of lupus and atherosclerosis. Arthritis Rheum 2012; 64(9):
2975-85.
http://dx.doi.org/10.1002/art.34504 PMID: 22549550
[57] Somers EC, Zhao W, Lewis EE, et al. Type I interferons are associ-
ated with subclinical markers of cardiovascular disease in a cohort of
systemic lupus erythematosus patients. PLoS One 2012; 7(5)e37000
http://dx.doi.org/10.1371/journal.pone.0037000 PMID: 22606325
[58] Lee PY, Li Y, Richards HB, et al. Type I interferon as a novel risk
factor for endothelial progenitor cell depletion and endothelial dys-
function in systemic lupus erythematosus. Arthritis Rheum 2007;
56(11): 3759-69.
http://dx.doi.org/10.1002/art.23035 PMID: 17968925
[59] Crow MK. Type I interferon in the pathogenesis of lupus. J Immunol
2014; 192(12): 5459-68.
http://dx.doi.org/10.4049/jimmunol.1002795 PMID: 24907379
[60] Goossens P, Gijbels MJ, Zernecke A, et al. Myeloid type I interferon
signaling promotes atherosclerosis by stimulating macrophage re-
cruitment to lesions. Cell Metab 2010; 12(2): 142-53.
http://dx.doi.org/10.1016/j.cmet.2010.06.008 PMID: 20674859
[61] Diao Y, Mohandas R, Lee P, et al. Effects of Long-Term Type I Inter-
feron on the Arterial Wall and Smooth Muscle Progenitor Cells Dif-
ferentiation. Arterioscler Thromb Vasc Biol 2016; 36(2): 266-73.
http://dx.doi.org/10.1161/ATVBAHA.115.306767 PMID: 26634654
[62] Tydén H, Lood C, Gullstrand B, et al. Endothelial dysfunction is asso-
ciated with activation of the type I interferon system and platelets in
patients with systemic lupus erythematosus. RMD Open 2017;
3(2)e000508
http://dx.doi.org/10.1136/rmdopen-2017-000508 PMID: 29119007
[63] Svenungsson E, Fei GZ, Jensen-Urstad K, de Faire U, Hamsten A,
Frostegard J. TNF-alpha: a link between hypertriglyceridaemia and in-
flammation in SLE patients with cardiovascular disease. Lupus 2003;
12(6): 454-61.
http://dx.doi.org/10.1191/0961203303lu412oa PMID: 12873047
[64] Rho YH, Chung CP, Oeser A, et al. Novel cardiovascular risk factors
in premature coronary atherosclerosis associated with systemic lupus
erythematosus. J Rheumatol 2008; 35(9): 1789-94.
PMID: 18634156
[65] Benagiano M, Borghi MO, Romagnoli J, et al. Interleukin-
17/Interleukin-21 and Interferon-g producing T cells specific for beta2
Glycoprotein I in atherosclerosis inflammation of systemic lupus
erythematosus patients with antiphospholipid syndrome Haema-
tologica 2018. pii: haematol.2018.209536.
[66] Shovman O, Gilburd B, Shoenfeld Y. The role of inflammatory cyto-
kines in the pathogenesis of systemic lupus erythematosus-related
atherosclerosis: a novel target for treatment? J Rheumatol 2006; 33(3):
445-7.
PMID: 16511912
[67] Asanuma Y, Chung CP, Oeser A, et al. Increased concentration of
proatherogenic inflammatory cytokines in systemic lupus erythemato-
sus: relationship to cardiovascular risk factors. J Rheumatol 2006;
33(3): 539-45.
PMID: 16463434
[68] Ahmad HM, Sarhan EM, Komber U. Higher circulating levels of
OxLDL % of LDL are associated with subclinical atherosclerosis in
female patients with systemic lupus erythematosus. Rheumatol Int
2014; 34(5): 617-23.
http://dx.doi.org/10.1007/s00296-013-2875-0 PMID: 24101173
[69] McMahon M, Grossman J, FitzGerald J, et al. Proinflammatory high-
density lipoprotein as a biomarker for atherosclerosis in patients with
systemic lupus erythematosus and rheumatoid arthritis. Arthritis
Rheum 2006; 54(8): 2541-9.
http://dx.doi.org/10.1002/art.21976 PMID: 16868975
[70] O’Neill SG, Giles I, Lambrianides A, et al. Antibodies to apolipopro-
tein A-I, high-density lipoprotein, and C-reactive protein are associ-
ated with disease activity in patients with systemic lupus erythemato-
sus. Arthritis Rheum 2010; 62(3): 845-54.
http://dx.doi.org/10.1002/art.27286 PMID: 20131231
[71] Matsuura E, Kobayashi K, Inoue K, Lopez LR, Shoenfeld Y. Oxidized
LDL/beta2-glycoprotein I complexes: new aspects in atherosclerosis.
Lupus 2005; 14(9): 736-41.
http://dx.doi.org/10.1191/0961203305lu2211oa PMID: 16218478
[72] Rosales C. Neutrophil: A Cell with Many Roles in Inflammation or
Several Cell Types? Front Physiol 2018; 9: 113.
http://dx.doi.org/10.3389/fphys.2018.00113 PMID: 29515456
[73] Gaul DS, Stein S, Matter CM. Neutrophils in cardiovascular disease.
Eur Heart J 2017; 38(22): 1702-4.
http://dx.doi.org/10.1093/eurheartj/ehx244 PMID: 30052884
[74] Knight JS, Kaplan MJ. Lupus neutrophils: ‘NET’ gain in understand-
ing lupus pathogenesis. Curr Opin Rheumatol 2012; 24(5): 441-50.
http://dx.doi.org/10.1097/BOR.0b013e3283546703 PMID: 22617827
[75] Carlucci PM, Purmalek MM, Dey AK, et al. Neutrophil subsets and
their gene signature associate with vascular inflammation and coro-
nary atherosclerosis in lupus. JCI Insight 2018; 3(8): 99276.
http://dx.doi.org/10.1172/jci.insight.99276 PMID: 29669944
[76] Denny MF, Yalavarthi S, Zhao W, et al. A distinct subset of proin-
flammatory neutrophils isolated from patients with systemic lupus
erythematosus induces vascular damage and synthesizes type I IFNs. J
Immunol 2010; 184(6): 3284-97.
http://dx.doi.org/10.4049/jimmunol.0902199 PMID: 20164424
[77] Neubert E, Meyer D, Rocca F, et al. Chromatin swelling drives neu-
trophil extracellular trap release. Nat Commun 2018; 9(1): 3767.
http://dx.doi.org/10.1038/s41467-018-06263-5 PMID: 30218080
[78] Garcia-Romo GS, Caielli S, Vega B, et al. Netting neutrophils are
major inducers of type I IFN production in pediatric systemic lupus
erythematosus. Sci Transl Med 2011; 3(73)73ra20
http://dx.doi.org/10.1126/scitranslmed.3001201 PMID: 21389264
[79] Pieterse E, Rother N, Garsen M, et al. Neutrophil Extracellular Traps
Drive Endothelial-to-Mesenchymal Transition. Arterioscler Thromb
Vasc Biol 2017; 37(7): 1371-9.
http://dx.doi.org/10.1161/ATVBAHA.117.309002 PMID: 28495931
[80] Schreiber K, Sciascia S, de Groot PG, et al. Antiphospholipid syn-
drome. Nat Rev Dis Primers 2018; 4: 17103.
http://dx.doi.org/10.1038/nrdp.2017.103 PMID: 29321641
[81] Tektonidou MG, Laskari K, Panagiotakos DB, Moutsopoulos HM.
Risk factors for thrombosis and primary thrombosis prevention in pa-
tients with systemic lupus erythematosus with or without antiphos-
pholipid antibodies. Arthritis Rheum 2009; 61(1): 29-36.
http://dx.doi.org/10.1002/art.24232 PMID: 19116963
[82] de Groot PG, Urbanus RT, Derksen RH. Pathophysiology of throm-
botic APS: where do we stand? Lupus 2012; 21(7): 704-7.
http://dx.doi.org/10.1177/0961203312438631 PMID: 22635207
[83] Zhang X, Xie Y, Zhou H, et al. Involvement of TLR4 in oxidized
LDL/β2GPI/anti-β2GPI-induced transformation of macrophages to
foam cells. J Atheroscler Thromb 2014; 21(11): 1140-51.
http://dx.doi.org/10.5551/jat.24372 PMID: 24998486
[84] Sun J, Lundström SL, Zhang B, et al. IgM antibodies against phos-
phorylcholine promote polarization of T regulatory cells from patients
with atherosclerotic plaques, systemic lupus erythematosus and
healthy donors. Atherosclerosis 2018; 268: 36-48.
http://dx.doi.org/10.1016/j.atherosclerosis.2017.11.010 PMID:
29175653
[85] Rahman M, Sing S, Golabkesh Z, et al. IgM antibodies against
malondialdehyde and phosphorylcholine are together strong protection
markers for atherosclerosis in systemic lupus erythematosus: Regula-
tion and underlying mechanisms. Clin Immunol 2016; 166-167: 27-37.
http://dx.doi.org/10.1016/j.clim.2016.04.007 PMID: 27102765
[86] Bruce IN, Urowitz MB, Gladman DD, Ibañez D, Steiner G. Risk fac-
tors for coronary heart disease in women with systemic lupus erythe-
matosus: the Toronto Risk Factor Study. Arthritis Rheum 2003;
48(11): 3159-67.
http://dx.doi.org/10.1002/art.11296 PMID: 14613278
[87] Tselios K, Sheane BJ, Gladman DD, Urowitz MB. Optimal Monitor-
ing For Coronary Heart Disease Risk in Patients with Systemic Lupus
Erythematosus: A Systematic Review. J Rheumatol 2016; 43(1): 54-
65.
http://dx.doi.org/10.3899/jrheum.150460 PMID: 26568591
[88] Schoenfeld SR, Kasturi S, Costenbader KH. The epidemiology of
atherosclerotic cardiovascular disease among patients with SLE: a sys-
tematic review. Semin Arthritis Rheum 2013; 43(1): 77-95.
http://dx.doi.org/10.1016/j.semarthrit.2012.12.002 PMID: 23422269
[89] Gustafsson J, Gunnarsson I, Börjesson O, et al. Predictors of the first
cardiovascular event in patients with systemic lupus erythematosus - a
prospective cohort study. Arthritis Res Ther 2009; 11(6): R186.
http://dx.doi.org/10.1186/ar2878 PMID: 20003285
14 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
[90] Nikpour M, Urowitz MB, Ibanez D, Harvey PJ, Gladman DD. Impor-
tance of cumulative exposure to elevated cholesterol and blood pres-
sure in development of atherosclerotic coronary artery disease in sys-
temic lupus erythematosus: a prospective proof-of-concept cohort
study. Arthritis Res Ther 2011; 13(5): R156.
http://dx.doi.org/10.1186/ar3473 PMID: 21955652
[91] Atukorala I, Weeratunga P, Kalubowila J, et al. Cardiovascular risk in
lupus nephritis: Do renal disease-related and other traditional risk fac-
tors play a role? Saudi J Kidney Dis Transpl 2015; 26(3): 526-35.
http://dx.doi.org/10.4103/1319-2442.157357 PMID: 26022023
[92] Bengtsson C, Ohman ML, Nived O, Rantapää Dahlqvist S. Cardiovas-
cular event in systemic lupus erythematosus in northern Sweden: inci-
dence and predictors in a 7-year follow-up study. Lupus 2012; 21(4):
452-9.
http://dx.doi.org/10.1177/0961203311425524 PMID: 22065097
[93] Haque S, Gordon C, Isenberg D, et al. Risk factors for clinical coro-
nary heart disease in systemic lupus erythematosus: the lupus and
atherosclerosis evaluation of risk (LASER) study. J Rheumatol 2010;
37(2): 322-9.
http://dx.doi.org/10.3899/jrheum.090306 PMID: 19955047
[94] Tselios K, Gladman DD, Su J, Urowitz MB. Does Renin-Angiotensin
System Blockade Protect Lupus Nephritis Patients From Atheroscle-
rotic Cardiovascular Events? A Case-Control Study. Arthritis Care Res
(Hoboken) 2016; 68(10): 1497-504.
http://dx.doi.org/10.1002/acr.22857 PMID: 26866934
[95] Urowitz MB, Gladman D, Ibañez D, et al. Systemic Lupus Interna-
tional Collaborating Clinics. Atherosclerotic vascular events in a mul-
tinational inception cohort of systemic lupus erythematosus. Arthritis
Care Res (Hoboken) 2010; 62(6): 881-7.
http://dx.doi.org/10.1002/acr.20122 PMID: 20535799
[96] Doria A, Shoenfeld Y, Wu R, et al. Risk factors for subclinical athero-
sclerosis in a prospective cohort of patients with systemic lupus
erythematosus. Ann Rheum Dis 2003; 62(11): 1071-7.
http://dx.doi.org/10.1136/ard.62.11.1071 PMID: 14583570
[97] Manzi S, Selzer F, Sutton-Tyrrell K, et al. Prevalence and risk factors
of carotid plaque in women with systemic lupus erythematosus. Ar-
thritis Rheum 1999; 42(1): 51-60.
http://dx.doi.org/10.1002/1529-0131(199901)42:1<51::AID-
ANR7>3.0.CO;2-D PMID: 9920014
[98] Schanberg LE, Sandborg C, Barnhart HX, et al. Atherosclerosis Pre-
vention in Pediatric Lupus Erythematosus Investigators. Premature
atherosclerosis in pediatric systemic lupus erythematosus: risk factors
for increased carotid intima-media thickness in the atherosclerosis
prevention in pediatric lupus erythematosus cohort. Arthritis Rheum
2009; 60(5): 1496-507.
http://dx.doi.org/10.1002/art.24469 PMID: 19404953
[99] Ramzy D, Tumiati LC, Tepperman E, et al. Dual immunosuppression
enhances vasomotor injury: interactive effect between endothelin-1
and nitric oxide bioavailability. J Thorac Cardiovasc Surg 2008;
135(4): 938-44.
http://dx.doi.org/10.1016/j.jtcvs.2007.09.075 PMID: 18374784
[100] Schäfer SC, Wallerath T, Closs EI, et al. Dexamethasone suppresses
eNOS and CAT-1 and induces oxidative stress in mouse resistance ar-
terioles. Am J Physiol Heart Circ Physiol 2005; 288(1): H436-44.
http://dx.doi.org/10.1152/ajpheart.00587.2004 PMID: 15598872
[101] Verhoeven F, Prati C, Maguin-Gaté K, Wendling D, Demougeot C.
Glucocorticoids and endothelial function in inflammatory diseases: fo-
cus on rheumatoid arthritis. Arthritis Res Ther 2016; 18(1): 258.
http://dx.doi.org/10.1186/s13075-016-1157-0 PMID: 27814748
[102] Boumpas DT. A novel action of glucocorticoids--NF-kappa B inhibi-
tion. Br J Rheumatol 1996; 35(8): 709-10.
http://dx.doi.org/10.1093/rheumatology/35.8.709 PMID: 8761180
[103] van de Stolpe A, Caldenhoven E, Raaijmakers JA, van der Saag PT,
Koenderman L. Glucocorticoid-mediated repression of intercellular
adhesion molecule-1 expression in human monocytic and bronchial
epithelial cell lines. Am J Respir Cell Mol Biol 1993; 8(3): 340-7.
http://dx.doi.org/10.1165/ajrcmb/8.3.340 PMID: 8095395
[104] Pamukcu B, Lip GY, Shantsila E. The nuclear factor--kappa B path-
way in atherosclerosis: a potential therapeutic target for atherothrom-
botic vascular disease. Thromb Res 2011; 128(2): 117-23.
http://dx.doi.org/10.1016/j.thromres.2011.03.025 PMID: 21636112
[105] Asai K, Funaki C, Hayashi T, et al. Dexamethasone-induced suppres-
sion of aortic atherosclerosis in cholesterol-fed rabbits. Possible
mechanisms. Arterioscler Thromb 1993; 13(6): 892-9.
http://dx.doi.org/10.1161/01.ATV.13.6.892 PMID: 8499410
[106] Sarnes E, Crofford L, Watson M, Dennis G, Kan H, Bass D. Incidence
and US costs of corticosteroid-associated adverse events: a systematic
literature review. Clin Ther 2011; 33(10): 1413-32.
http://dx.doi.org/10.1016/j.clinthera.2011.09.009 PMID: 21999885
[107] Kaiser R, Cleveland CM, Criswell LA. Risk and protective factors for
thrombosis in systemic lupus erythematosus: results from a large,
multi-ethnic cohort. Ann Rheum Dis 2009; 68(2): 238-41.
http://dx.doi.org/10.1136/ard.2008.093013 PMID: 18782792
[108] Jung H, Bobba R, Su J, et al. The protective effect of antimalarial
drugs on thrombovascular events in systemic lupus erythematosus. Ar-
thritis Rheum 2010; 62(3): 863-8.
http://dx.doi.org/10.1002/art.27289 PMID: 20131232
[109] Ruiz-Irastorza G, Egurbide MV, Pijoan JI, et al. Effect of antimalarials
on thrombosis and survival in patients with systemic lupus erythema-
tosus. Lupus 2006; 15(9): 577-83.
http://dx.doi.org/10.1177/0961203306071872 PMID: 17080912
[110] Selzer F, Sutton-Tyrrell K, Fitzgerald S, Tracy R, Kuller L, Manzi S.
Vascular stiffness in women with systemic lupus erythematosus. Hy-
pertension 2001; 37(4): 1075-82.
http://dx.doi.org/10.1161/01.HYP.37.4.1075 PMID: 11304506
[111] Durcan L, Winegar DA, Connelly MA, Otvos JD, Magder LS, Petri
M. Longitudinal Evaluation of Lipoprotein Variables in Systemic Lu-
pus Erythematosus Reveals Adverse Changes with Disease Activity
and Prednisone and More Favorable Profiles with Hydroxychloroquine
Therapy. J Rheumatol 2016; 43(4): 745-50.
http://dx.doi.org/10.3899/jrheum.150437 PMID: 26834214
[112] Rahman P, Gladman DD, Urowitz MB, Yuen K, Hallett D, Bruce IN.
The cholesterol lowering effect of antimalarial drugs is enhanced in
patients with lupus taking corticosteroid drugs. J Rheumatol 1999;
26(2): 325-30.
PMID: 9972966
[113] Virdis A, Tani C, Duranti E, et al. Early treatment with hydroxy-
chloroquine prevents the development of endothelial dysfunction in a
murine model of systemic lupus erythematosus. Arthritis Res Ther
2015; 17: 277.
http://dx.doi.org/10.1186/s13075-015-0790-3 PMID: 26444671
[114] Sacre K, Criswell LA, McCune JM. Hydroxychloroquine is associated
with impaired interferon-alpha and tumor necrosis factor-alpha pro-
duction by plasmacytoid dendritic cells in systemic lupus erythemato-
sus. Arthritis Res Ther 2012; 14(3): R155.
http://dx.doi.org/10.1186/ar3895 PMID: 22734582
[115] Frangou E, Chrysanthopoulou A, Mitsios A, et al. REDD1/autophagy
pathway promotes thromboinflammation and fibrosis in human sys-
temic lupus erythematosus (SLE) through NETs decorated with tissue
factor (TF) and interleukin-17A (IL-17A). Ann Rheum Dis 2019;
78(2): 238-48.
http://dx.doi.org/10.1136/annrheumdis-2018-213181 PMID: 30563869
[116] Teixeira V, Tam LS. Novel Insights in Systemic Lupus Erythematosus
and Atherosclerosis. Front Med (Lausanne) 2018; 4: 262.
http://dx.doi.org/10.3389/fmed.2017.00262 PMID: 29435447
[117] Espinola RG, Pierangeli SS, Gharavi AE, Harris EN. Hydroxychloro-
quine reverses platelet activation induced by human IgG antiphosphol-
ipid antibodies. Thromb Haemost 2002; 87(3): 518-22.
http://dx.doi.org/10.1055/s-0037-1613033 PMID: 11916085
[118] van Leuven SI, Mendez-Fernandez YV, Wilhelm AJ, et al. Mycophe-
nolate mofetil but not atorvastatin attenuates atherosclerosis in lupus-
prone LDLr(-/-) mice. Ann Rheum Dis 2012; 71(3): 408-14.
http://dx.doi.org/10.1136/annrheumdis-2011-200071 PMID: 21953346
[119] Romero F, Rodríguez-Iturbe B, Pons H, et al. Mycophenolate mofetil
treatment reduces cholesterol-induced atherosclerosis in the rabbit.
Atherosclerosis 2000; 152(1): 127-33.
http://dx.doi.org/10.1016/S0021-9150(99)00458-X PMID: 10996347
[120] van Leuven SI, van Wijk DF, Volger OL, et al. Mycophenolate
mofetil attenuates plaque inflammation in patients with symptomatic
carotid artery stenosis. Atherosclerosis 2010; 211(1): 231-6.
http://dx.doi.org/10.1016/j.atherosclerosis.2010.01.043 PMID:
20202636
[121] Kiani AN, Magder LS, Petri M. Mycophenolate mofetil (MMF) does
not slow the progression of subclinical atherosclerosis in SLE over 2
years. Rheumatol Int 2012; 32(9): 2701-5.
http://dx.doi.org/10.1007/s00296-011-2048-y PMID: 21792642
[122] Sazliyana S, Mohd Shahrir MS, Kong CT, Tan HJ, Hamidon BB,
Azmi MT. Implications of immunosuppressive agents in cardiovascu-
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 15
lar risks and carotid intima media thickness among lupus nephritis pa-
tients. Lupus 2011; 20(12): 1260-6.
http://dx.doi.org/10.1177/0961203311411347 PMID: 21844115
[123] Ridker PM, Everett BM, Pradhan A, et al. CIRT Investigators. Low-
Dose Methotrexate for the Prevention of Atherosclerotic Events. N
Engl J Med 2019; 380(8): 752-62.
http://dx.doi.org/10.1056/NEJMoa1809798 PMID: 30415610
[124] Micha R, Imamura F, Wyler von Ballmoos M, et al. Systematic review
and meta-analysis of methotrexate use and risk of cardiovascular dis-
ease. Am J Cardiol 2011; 108(9): 1362-70.
http://dx.doi.org/10.1016/j.amjcard.2011.06.054 PMID: 21855836
[125] Ahmad Y, Shelmerdine J, Bodill H, et al. Subclinical atherosclerosis
in systemic lupus erythematosus (SLE): the relative contribution of
classic risk factors and the lupus phenotype. Rheumatology (Oxford)
2007; 46(6): 983-8.
http://dx.doi.org/10.1093/rheumatology/kem002 PMID: 17384179
[126] Ho KT, Ahn CW, Alarcón GS, et al. Systemic lupus erythematosus in
a multiethnic cohort (LUMINA): XXVIII. Factors predictive of
thrombotic events. Rheumatology (Oxford) 2005; 44(10): 1303-7.
http://dx.doi.org/10.1093/rheumatology/kei014 PMID: 16030085
[127] Oryoji K, Kiyohara C, Horiuchi T, et al. Reduced carotid intima-
media thickness in systemic lupus erythematosus patients treated with
cyclosporine A. Mod Rheumatol 2014; 24(1): 86-92.
http://dx.doi.org/10.3109/14397595.2013.852838 PMID: 24261763
[128] Fanouriakis A, Kostopoulou M, Alunno A, et al. 2019 update of the
EULAR recommendations for the management of systemic lupus
erythematosus. Ann Rheum Dis 2019; 78(6): 736-45.
http://dx.doi.org/10.1136/annrheumdis-2019-215089 PMID: 30926722
[129] Benvenuti F, Gatto M, Larosa M, Iaccarino L, Punzi L, Doria A. Car-
diovascular risk factors, burden of disease and preventive strategies in
patients with systemic lupus erythematosus: a literature review. Expert
Opin Drug Saf 2015; 14(9): 1373-85.
http://dx.doi.org/10.1517/14740338.2015.1073259 PMID: 26212119
[130] Ait-Oufella H, Herbin O, Bouaziz JD, et al. B cell depletion reduces
the development of atherosclerosis in mice. J Exp Med 2010; 207(8):
1579-87.
http://dx.doi.org/10.1084/jem.20100155 PMID: 20603314
[131] Koulouri V, Koutsilieris M, Mavragani CP. B cells and atherosclerosis
in systemic lupus erythematosus. Expert Rev Clin Immunol 2019;
15(4): 417-29.
http://dx.doi.org/10.1080/1744666X.2019.1571411 PMID: 30652925
[132] Kyaw T, Cui P, Tay C, et al. BAFF receptor mAb treatment amelio-
rates development and progression of atherosclerosis in hyperlipi-
demic ApoE(-/-) mice. PLoS One 2013; 8(4)e60430
http://dx.doi.org/10.1371/journal.pone.0060430 PMID: 23560095
[133] Kyaw T, Tay C, Hosseini H, et al. Depletion of B2 but not B1a B cells
in BAFF receptor-deficient ApoE mice attenuates atherosclerosis by
potently ameliorating arterial inflammation. PLoS One 2012;
7(1)e29371
http://dx.doi.org/10.1371/journal.pone.0029371 PMID: 22238605
[134] Kahn P, Ramanujam M, Bethunaickan R, et al. Prevention of murine
antiphospholipid syndrome by BAFF blockade. Arthritis Rheum 2008;
58(9): 2824-34.
http://dx.doi.org/10.1002/art.23764 PMID: 18759321
[135] Sciascia S, Rubini E, Radin M, Cecchi I, Rossi D, Roccatello D. Anti-
cardiolipin and anti-beta 2 glycoprotein-I antibodies disappearance in
patients with systemic lupus erythematosus and antiphospholipid syn-
drome while on belimumab. Ann Rheum Dis 2018; 77(11): 1694-5.
http://dx.doi.org/10.1136/annrheumdis-2018-213496 PMID: 29776976
[136] Parodis I, Gomez A, Emamikia S, Chatzidionysiou K. Established
organ damage reduces belimumab efficacy in systemic lupus erythe-
matosus. Ann Rheum Dis 2019; 78(7): 1006-7.
http://dx.doi.org/10.1136/annrheumdis-2018-214880 PMID: 30610063
[137] Parodis I, Johansson P, Gomez A, Soukka S, Emamikia S, Chatzidi-
onysiou K. Predictors of low disease activity and clinical remission
following belimumab treatment in systemic lupus erythematosus.
Rheumatology (Oxford) 2019; 58(12): 2170-6.
http://dx.doi.org/10.1093/rheumatology/kez191 PMID: 31157891
[138] Parodis I, Sjöwall C, Jönsen A, et al. Smoking and pre-existing organ
damage reduce the efficacy of belimumab in systemic lupus erythema-
tosus. Autoimmun Rev 2017; 16(4): 343-51.
http://dx.doi.org/10.1016/j.autrev.2017.02.005 PMID: 28216072
[139] Casey KA, Guo X, Smith MA, et al. Type I interferon receptor block-
ade with anifrolumab corrects innate and adaptive immune perturba-
tions of SLE. Lupus Sci Med 2018; 5(1)e000286
PMID: 30538817
[140] Cheung TT, Tsoi MF, Cheung BMY. SAT0099 Effect of TNF Inhibi-
tors on Subclinical Atherosclerosis in Patients with Rheumatoid Ar-
thritis: A Meta-Analysis 2015; 742
http://dx.doi.org/10.1136/annrheumdis-2015-eular.5424
[141] Toloza SM, Uribe AG, McGwin G Jr, et al. LUMINA Study Group.
Systemic lupus erythematosus in a multiethnic US cohort (LUMINA).
XXIII. Baseline predictors of vascular events. Arthritis Rheum 2004;
50(12): 3947-57.
http://dx.doi.org/10.1002/art.20622 PMID: 15593203
[142] Koenig KF, Ribi C, Radosavac M, Zulewski H, Trendelenburg M.
Swiss SLE cohort study (SSCS). Prevalence of vascular disease in sys-
temic lupus erythematosus compared with type-1 diabetes mellitus: a
cross-sectional study of two cohorts. Lupus 2015; 24(1): 58-65.
http://dx.doi.org/10.1177/0961203314550223 PMID: 25182169
[143] Becker-Merok A, Nossent J. Prevalence, predictors and outcome of
vascular damage in systemic lupus erythematosus. Lupus 2009; 18(6):
508-15.
http://dx.doi.org/10.1177/0961203308099233 PMID: 19395452
[144] Barbhaiya M, Feldman CH, Guan H, et al. Race/Ethnicity and Cardio-
vascular Events Among Patients With Systemic Lupus Erythematosus.
Arthritis Rheumatol 2017; 69(9): 1823-31.
http://dx.doi.org/10.1002/art.40174 PMID: 28598016
[145] Ward MM. Outcomes of hospitalizations for myocardial infarctions
and cerebrovascular accidents in patients with systemic lupus erythe-
matosus. Arthritis Rheum 2004; 50(10): 3170-6.
http://dx.doi.org/10.1002/art.20556 PMID: 15476215
[146] Ando T, Adegbala O, Akintoye E, et al. Acute Myocardial Infarction
Outcomes in Systemic Lupus Erythematosus (from the Nationwide In-
patient Sample). Am J Cardiol 2019; 123(2): 227-32.
http://dx.doi.org/10.1016/j.amjcard.2018.09.043 PMID: 30424870
[147] Maksimowicz-McKinnon K, Selzer F, Manzi S, et al. Poor 1-year
outcomes after percutaneous coronary interventions in systemic lupus
erythematosus: report from the National Heart, Lung, and Blood Insti-
tute Dynamic Registry. Circ Cardiovasc Interv 2008; 1(3): 201-8.
http://dx.doi.org/10.1161/CIRCINTERVENTIONS.108.788745
PMID: 20031679
[148] Björnådal L, Yin L, Granath F, Klareskog L, Ekbom A. Cardiovascu-
lar disease a hazard despite improved prognosis in patients with sys-
temic lupus erythematosus: results from a Swedish population based
study 1964-95. J Rheumatol 2004; 31(4): 713-9.
PMID: 15088296
[149] Lin CY, Shih CC, Yeh CC, Chou WH, Chen TL, Liao CC. Increased
risk of acute myocardial infarction and mortality in patients with sys-
temic lupus erythematosus: two nationwide retrospective cohort stud-
ies. Int J Cardiol 2014; 176(3): 847-51.
http://dx.doi.org/10.1016/j.ijcard.2014.08.006 PMID: 25156834
[150] Arkema EV, Svenungsson E, Von Euler M, Sjöwall C, Simard JF.
Stroke in systemic lupus erythematosus: a Swedish population-based
cohort study. Ann Rheum Dis 2017; 76(9): 1544-9.
http://dx.doi.org/10.1136/annrheumdis-2016-210973 PMID: 28400384
[151] Mok CC, Ho LY, To CH. Annual incidence and standardized inci-
dence ratio of cerebrovascular accidents in patients with systemic lu-
pus erythematosus. Scand J Rheumatol 2009; 38(5): 362-8.
http://dx.doi.org/10.1080/03009740902776927 PMID: 19296403
[152] Holmqvist M, Simard JF, Asplund K, Arkema EV. Stroke in systemic
lupus erythematosus: a meta-analysis of population-based cohort stud-
ies. RMD Open 2015; 1(1)e000168
http://dx.doi.org/10.1136/rmdopen-2015-000168 PMID: 26719816
[153] Nikolopoulos D, Fanouriakis A, Boumpas D. Cerebrovascular Events
in Systemic Lupus Erythematosus: Diagnosis and Management.
Mediterr J Rheumatol 2019; 30(1): 7-15.
http://dx.doi.org/10.31138/mjr.30.1.7
[154] Rossides M, Simard JF, Svenungsson E, von Euler M, Arkema EV.
Mortality and Functionality after Stroke in Patients with Systemic Lu-
pus Erythematosus. J Rheumatol 2017; 44(11): 1590-6.
http://dx.doi.org/10.3899/jrheum.170241 PMID: 28916550
[155] Erdozain JG, Villar I, Nieto J, Ruiz-Irastorza G. Peripheral arterial
disease in systemic lupus erythematosus: prevalence and risk factors. J
Rheumatol 2014; 41(2): 310-7.
http://dx.doi.org/10.3899/jrheum.130817 PMID: 24429176
16 Current Vascular Pharmacology, 2020, Vol. 18, No. 00 Kostopoulou et al.
[156] June RR, Scalzi LV. Peripheral vascular disease in systemic lupus
patients. J Clin Rheumatol 2013; 19(7): 367-72.
http://dx.doi.org/10.1097/RHU.0000000000000017 PMID: 24048099
[157] Hassan AA, Habib HM, Eissa AA. Peripheral arterial disease in pa-
tients with systemic lupus erythematosus: a prospective controlled
study. Int J Rheum Dis 2013; 16(3): 319-24.
http://dx.doi.org/10.1111/1756-185x.12025 PMID: 23981754
[158] Chuang YW, Yu MC, Lin CL, Yu TM, Shu KH, Kao CH. Risk of
Peripheral Arterial Occlusive Disease in Patients With Systemic Lupus
Erythematosus: A Nationwide Population-Based Cohort Study. Medi-
cine (Baltimore) 2015; 94(46)e2121
http://dx.doi.org/10.1097/MD.0000000000002121 PMID: 26579830
[159] Chung CP, Oeser A, Avalos I, Raggi P, Stein CM. Cardiovascular risk
scores and the presence of subclinical coronary artery atherosclerosis
in women with systemic lupus erythematosus. Lupus 2006; 15(9):
562-9.
http://dx.doi.org/10.1177/0961203306071870 PMID: 17080910
[160] Sivakumaran J, Harvey P, Omar A, et al. 291 Assessment of the
QRISK2, QRISK3, SLE cardiovascular risk equation, modified
framingham and framingham risk calculators as predictors of cardio-
vascular disease events in systemic lupus erythematosus. Lupus Sci-
ence &amp. Medicine (Baltimore) 2019; 6(Suppl. 1): A211-.
[161] Hippisley-Cox J, Coupland C, Brindle P. Development and validation
of QRISK3 risk prediction algorithms to estimate future risk of car-
diovascular disease: prospective cohort study. BMJ 2017; 357: j2099.
http://dx.doi.org/10.1136/bmj.j2099 PMID: 28536104
[162] Piepoli MF, Hoes AW, Agewall S, et al. ESC Scientific Document
Group. 2016 European Guidelines on cardiovascular disease preven-
tion in clinical practice: The Sixth Joint Task Force of the European
Society of Cardiology and Other Societies on Cardiovascular Disease
Prevention in Clinical Practice (constituted by representatives of 10
societies and by invited experts)Developed with the special contribu-
tion of the European Association for Cardiovascular Prevention & Re-
habilitation (EACPR). Eur Heart J 2016; 37(29): 2315-81.
http://dx.doi.org/10.1093/eurheartj/ehw106 PMID: 27222591
[163] Urowitz MB, Ibañez D, Su J, Gladman DD. Modified Framingham
Risk Factor Score for Systemic Lupus Erythematosus. J Rheumatol
2016; 43(5): 875-9.
http://dx.doi.org/10.3899/jrheum.150983 PMID: 26879352
[164] Petri M, Magder L. 2012.https://acrabstracts.org/abstract/systemic-
lupus-erythematosus-cardiovascular-risk-equation/
[165] Hadamitzky M, Freissmuth B, Meyer T, et al. Prognostic value of
coronary computed tomographic angiography for prediction of cardiac
events in patients with suspected coronary artery disease. JACC Car-
diovasc Imaging 2009; 2(4): 404-11.
http://dx.doi.org/10.1016/j.jcmg.2008.11.015 PMID: 19580721
[166] Kiani AN, Magder LS, Post WS, et al. Coronary calcification in SLE:
comparison with the Multi-Ethnic Study of Atherosclerosis. Rheuma-
tology (Oxford) 2015; 54(11): 1976-81.
http://dx.doi.org/10.1093/rheumatology/kev198 PMID: 26106213
[167] Petri MA, Kiani AN, Post W, Christopher-Stine L, Magder LS. Lupus
Atherosclerosis Prevention Study (LAPS). Ann Rheum Dis 2011;
70(5): 760-5.
http://dx.doi.org/10.1136/ard.2010.136762 PMID: 21177297
[168] Nagata M, Kato S, Kitagawa K, et al. Diagnostic accuracy of 1.5-T
unenhanced whole-heart coronary MR angiography performed with
32-channel cardiac coils: initial single-center experience. Radiology
2011; 259(2): 384-92.
http://dx.doi.org/10.1148/radiol.11101323 PMID: 21406635
[169] Płazak W, Gryga K, Sznajd J, Pasowicz M, Musiał J, Podolec P. Myo-
cardial ischaemia in systemic lupus erythematosus: detection and
clinical relevance. Kardiol Pol 2011; 69(11): 1129-36.
PMID: 22090220
[170] Varma N, Hinojar R, D’Cruz D, et al. Coronary vessel wall contrast
enhancement imaging as a potential direct marker of coronary in-
volvement: integration of findings from CAD and SLE patients. JACC
Cardiovasc Imaging 2014; 7(8): 762-70.
http://dx.doi.org/10.1016/j.jcmg.2014.03.012 PMID: 25051945
[171] O’Leary DH, Polak JF, Kronmal RA, Manolio TA, Burke GL, Wolf-
son SK Jr. Cardiovascular Health Study Collaborative Research
Group. Carotid-artery intima and media thickness as a risk factor for
myocardial infarction and stroke in older adults. N Engl J Med 1999;
340(1): 14-22.
http://dx.doi.org/10.1056/NEJM199901073400103 PMID: 9878640
[172] Henrot P, Foret J, Barnetche T, et al. Assessment of subclinical athe-
rosclerosis in systemic lupus erythematosus: A systematic review and
meta-analysis. Joint Bone Spine 2018; 85(2): 155-63.
http://dx.doi.org/10.1016/j.jbspin.2017.12.009 PMID: 29288864
[173] Tyrrell PN, Beyene J, Feldman BM, McCrindle BW, Silverman ED,
Bradley TJ. Rheumatic disease and carotid intima-media thickness: a
systematic review and meta-analysis. Arterioscler Thromb Vasc Biol
2010; 30(5): 1014-26.
http://dx.doi.org/10.1161/ATVBAHA.109.198424 PMID: 20150560
[174] Hadi HA, Carr CS, Al Suwaidi J. Endothelial dysfunction: cardiovas-
cular risk factors, therapy, and outcome. Vasc Health Risk Manag
2005; 1(3): 183-98.
PMID: 17319104
[175] Ghiadoni L, Salvetti M, Muiesan ML, Taddei S. Evaluation of endo-
thelial function by flow mediated dilation: methodological issues and
clinical importance. High Blood Press Cardiovasc Prev 2015; 22(1):
17-22.
http://dx.doi.org/10.1007/s40292-014-0047-2 PMID: 24619864
[176] Lima DS, Sato EI, Lima VC, Miranda F Jr, Hatta FH. Brachial endo-
thelial function is impaired in patients with systemic lupus erythema-
tosus. J Rheumatol 2002; 29(2): 292-7.
PMID: 11842823
[177] Mak A, Kow NY, Schwarz H, Gong L, Tay SH, Ling LH. Endothelial
dysfunction in systemic lupus erythematosus - a case-control study and
an updated meta-analysis and meta-regression. Sci Rep 2017; 7(1):
7320.
http://dx.doi.org/10.1038/s41598-017-07574-1 PMID: 28779080
[178] Korsten P, Patschan D, Henze E, Niewold TB, Müller GA, Patschan S.
Dynamics of pulse wave velocity and vascular augmentation index in
association with endothelial progenitor cells in SLE. Lupus Sci Med
2016; 3(1)e000185
http://dx.doi.org/10.1136/lupus-2016-000185 PMID: 28176918
[179] Tso TK, Huang WN, Huang HY, Chang CK. Association of brachial-
ankle pulse wave velocity with cardiovascular risk factors in systemic
lupus erythematosus. Lupus 2005; 14(11): 878-83.
http://dx.doi.org/10.1191/0961203305lu2234oa PMID: 16335579
[180] Vázquez-Del Mercado M, Perez-Vazquez FJ, Gomez-Bañuelos E, et
al. Subclinical parameters of arterial stiffness and arteriosclerosis cor-
relate with QRISK3 in systemic lupus erythematosus. PLoS One 2018;
13(12)e0207520
http://dx.doi.org/10.1371/journal.pone.0207520 PMID: 30517121
[181] McMahon M, Skaggs BJ, Grossman JM, et al. A panel of biomarkers
is associated with increased risk of the presence and progression of
atherosclerosis in women with systemic lupus erythematosus. Arthritis
Rheumatol 2014; 66(1): 130-9.
http://dx.doi.org/10.1002/art.38204 PMID: 24449580
[182] Lewandowski LB, Kaplan MJ. Update on cardiovascular disease in
lupus. Curr Opin Rheumatol 2016; 28(5): 468-76.
http://dx.doi.org/10.1097/BOR.0000000000000307 PMID: 27227346
[183] Hughes MM, O’Neill LAJ. Metabolic regulation of NLRP3. Immunol
Rev 2018; 281(1): 88-98.
http://dx.doi.org/10.1111/imr.12608 PMID: 29247992
[184] Kahlenberg JM, Kaplan MJ. The inflammasome and lupus: another
innate immune mechanism contributing to disease pathogenesis? Curr
Opin Rheumatol 2014; 26(5): 475-81.
http://dx.doi.org/10.1097/BOR.0000000000000088 PMID: 24992143
[185] Oesterle A, Bowman MA. S100A12 and the S100/Calgranulins:
Emerging Biomarkers for Atherosclerosis and Possibly Therapeutic
Targets. Arterioscler Thromb Vasc Biol 2015; 35(12): 2496-507.
http://dx.doi.org/10.1161/ATVBAHA.115.302072 PMID: 26515415
[186] Baptista D, Mach F, Brandt KJ. Follicular regulatory T cell in athero-
sclerosis. J Leukoc Biol 2018; 104(5): 925-30.
http://dx.doi.org/10.1002/JLB.MR1117-469R PMID: 30134501
[187] Chen Y, Zhang L, Huang H, et al. Association of OX40 and OX40L
gene polymorphisms with acute coronary syndrome in a Han Chinese
population. DNA Cell Biol 2011; 30(8): 597-602.
http://dx.doi.org/10.1089/dna.2010.1201 PMID: 21476935
[188] Barta Z, Harrison MJ, Wangrangsimakul T, et al. Health-related qual-
ity of life, smoking and carotid atherosclerosis in white British women
with systemic lupus erythematosus. Lupus 2010; 19(3): 231-8.
http://dx.doi.org/10.1177/0961203309351032 PMID: 20007814
[189] dos Reis-Neto ET, da Silva AE, Monteiro CM, de Camargo LM, Sato
EI. Supervised physical exercise improves endothelial function in pa-
Cardiovascular Disease in Lupus Current Vascular Pharmacology, 2020, Vol. 18, No . 00 17
tients with systemic lupus erythematosus. Rheumatology (Oxford)
2013; 52(12): 2187-95.
http://dx.doi.org/10.1093/rheumatology/ket283 PMID: 23970541
[190] Wright SA, O’Prey FM, McHenry MT, et al. A randomised interven-
tional trial of omega-3-polyunsaturated fatty acids on endothelial func-
tion and disease activity in systemic lupus erythematosus. Ann Rheum
Dis 2008; 67(6): 841-8.
http://dx.doi.org/10.1136/ard.2007.077156 PMID: 17875549
[191] Tu H, Li Q, Xiang S, et al. Dual effects of statins therapy in systemic
lupus erythematosus and SLE-related atherosclerosis: the potential role
for regulatory T cells. Atherosclerosis 2012; 222(1): 29-33.
http://dx.doi.org/10.1016/j.atherosclerosis.2012.02.023 PMID:
22417842
[192] Norby GE, Holme I, Fellström B, et al. Assessment of Lescol in Renal
Transplantation Study Group. Effect of fluvastatin on cardiac out-
comes in kidney transplant patients with systemic lupus erythemato-
sus: a randomized placebo-controlled study. Arthritis Rheum 2009;
60(4): 1060-4.
http://dx.doi.org/10.1002/art.24379 PMID: 19333947
[193] Yu HH, Chen PC, Yang YH, et al. Statin reduces mortality and mor-
bidity in systemic lupus erythematosus patients with hyperlipidemia:
A nationwide population-based cohort study. Atherosclerosis 2015;
243(1): 11-8.
http://dx.doi.org/10.1016/j.atherosclerosis.2015.08.030 PMID:
26342937
[194] Ardoin SP, Schanberg LE, Sandborg CI, et al. APPLE investigators.
Secondary analysis of APPLE study suggests atorvastatin may reduce
atherosclerosis progression in pubertal lupus patients with higher C re-
active protein. Ann Rheum Dis 2014; 73(3): 557-66.
http://dx.doi.org/10.1136/annrheumdis-2012-202315 PMID: 23436914
[195] Plazak W, Gryga K, Dziedzic H, et al. Influence of atorvastatin on
coronary calcifications and myocardial perfusion defects in systemic
lupus erythematosus patients: a prospective, randomized, double-
masked, placebo-controlled study. Arthritis Res Ther 2011; 13(4):
R117.
http://dx.doi.org/10.1186/ar3402 PMID: 21774822
[196] Mok CC, Wong CK, To CH, Lai JP, Lam CS. Effects of rosuvastatin
on vascular biomarkers and carotid atherosclerosis in lupus: a random-
ized, double-blind, placebo-controlled trial. Arthritis Care Res
(Hoboken) 2011; 63(6): 875-83.
http://dx.doi.org/10.1002/acr.20440 PMID: 21309005
[197] Arnaud L, Mathian A, Ruffatti A, et al. Efficacy of aspirin for the
primary prevention of thrombosis in patients with antiphospholipid an-
tibodies: an international and collaborative meta-analysis. Autoimmun
Rev 2014; 13(3): 281-91.
http://dx.doi.org/10.1016/j.autrev.2013.10.014 PMID: 24189281
[198] Tektonidou MG, Andreoli L, Limper M, et al. EULAR recommenda-
tions for the management of antiphospholipid syndrome in adults. Ann
Rheum Dis 2019; 78(10): 1296-304.
http://dx.doi.org/10.1136/annrheumdis-2019-215213 PMID: 31092409
[199] Fasano S, Pierro L, Pantano I, Iudici M, Va lentini G. Longterm Hy-
droxychloroquine Therapy and Low-dose Aspirin May Have an Addi-
tive Effectiveness in the Primary Prevention of Cardiovascular Events
in Patients with Systemic Lupus Erythematosus. J Rheumatol 2017;
44(7): 1032-8.
http://dx.doi.org/10.3899/jrheum.161351 PMID: 28507183
[200] Tao CY, Shang J, Chen T, et al. Impact of antimalarial (AM) on serum
lipids in systemic lupus erythematosus (SLE) patients: A systematic
review and meta-analysis. Medicine (Baltimore) 2019; 98(14)e15030
http://dx.doi.org/10.1097/MD.0000000000015030 PMID: 30946340
[201] Rempenault C, Combe B, Barnetche T, et al. Metabolic and cardio-
vascular benefits of hydroxychloroquine in patients with rheumatoid
arthritis: a systematic review and meta-analysis. Ann Rheum Dis
2018; 77(1): 98-103.
http://dx.doi.org/10.1136/annrheumdis-2017-211836 PMID: 28970215
[202] Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA Guide-
line on the Primary Prevention of Cardiovascular Disease: Executive
Summary: A Report of the American College of Cardiology/American
Heart Association Task Force on Clinical Practice Guidelines. J Am
Coll Cardiol 2019; 74(10): 1376-414.
http://dx.doi.org/10.1016/j.jacc.2019.03.009 PMID: 30894319
[203] Giannelou M, Mavragani CP. Cardiovascular disease in systemic
lupus erythematosus: A comprehensive update. J Autoimmun 2017;
82: 1-12.
http://dx.doi.org/10.1016/j.jaut.2017.05.008 PMID: 28606749
... The incidence ranges from 0.3 to 31.5 cases per 100,000 individuals per year and has increased during the last 40 years, probably because of recognition of milder cases [1]. SLE is an independent risk factor for cardiovascular disease, attributed both to traditional and disease-related risk factors [2]. In particular, the prevalence rate of valvular heart disease (VHD) is higher in patients with SLE than in the general population [3,4]. ...
... The novel and main findings in the present study were as follows. [1] The morbidity rate of VHD increased with advancing age, [2] a lower platelet count was significantly associated with having VHD, and [3] the use of immunosuppressant therapy might be useful to prevent VHD. ...
Article
Full-text available
Background Although the prevalence rate of valvular heart disease (VHD) is high in patients with systemic lupus erythematosus (SLE), the predictive factors of concomitant VHD have not been fully evaluated. Methods and results Among 288 patients with SLE who underwent transthoracic echocardiography at Kumamoto University Hospital from 2016 to 2021, 241 patients with sufficient echocardiographic data were retrospectively analysed. Among them, 22 (9 %) had VHD (10 had mitral regurgitation, 3 had aortic regurgitation, 6 had tricuspid regurgitation, 1 had mitral regurgitation and tricuspid regurgitation, and 2 had a prosthetic cardiac valve). After excluding the two patients with a prosthetic cardiac valve, we divided the remaining patients into two groups: the VHD group and non-VHD group. Multivariate logistic regression analysis revealed that age and the platelet count were significantly and independently associated with having VHD (age: odds ratio, 1.06; 95 % confidence interval, 1.02–1.10; p < 0.01) (platelet count: odds ratio, 0.99; 95 % confidence interval, 0.98–1.00; p < 0.05). After excluding 95 patients aged < 40 years, receiver operating characteristic analysis revealed that the area under the curve of the platelet count for prediction of VHD was 0.73 with an optimal cut-off value of 166.5 × 10³/µL (sensitivity: 76.6 %, specificity: 60.0 %). Among patients with a low platelet count (<166.5 × 10³/µL), the rate of having VHD was 29 % (12/41 patients). However, among those with a high platelet count (≥166.5 × 10³/µL), this rate was only 8 % (8/103 patients). Conclusion The platelet count is useful to predict concomitant VHD in middle-aged and older patients with SLE.
... The incidence of SLE in general population varied from 0.3 to 11.5 cases per 100,000 person-years and had increased in recent years with the development of diagnostic and treatment technologies and concepts, and the incidence of SLE was predominantly in middle-aged and young women, with a male to female ratio of about 1:10 [10,11] . Cardiovascular disease is one of the most common complications in patients with SLE [12] , and its development is associated with traditional risk factors as well as factors related to the disease itself, such as persistent disease activity, lupus nephritis and usages of glucocorticoids [13] . A study of Chung C P et al. found that blood NT-proBNP levels were higher than normal in patients with SLE [14] . ...
Preprint
Full-text available
Backgroud The incidence and prevalence of systemic lupus erythematosus (SLE) have increased annually over the past decade. The involvement of myocardium is one of the main reasons for the poor prognosis of patients with SLE. Identifying myocardial involvement in patients with autoimmune diseases and providing early targeted treatment can improve patient outcomes. Objectives The aim of this study is to evaluate myocardial dysfunction in patients with SLE using 99mTc-MIBI rest gated myocardial perfusion imaging (rGMPI) and to investigate factors associated with myocardial dysfunction. Method 76 patients with SLE were prospectively enrolled in the study and 46 patients without autoimmune diseases or other inflammatory diseases who had undergone 99mTc-MIBI rGMPI were selected as a control group. Results of relevant blood test indicators, echocardiography and rGMPI were recorded, and comparison was made between the two groups. Meanwhile, based on diagnostic results of rGMPI, SLE patients were divided into myocardial dysfunction group and normal myocardial function group and to analyze the influencing factors of myocardial dysfunction in SLE patients. Results The incidence of myocardial dysfunction was significantly higher in SLE patients than in controls (30.3% vs 0%, χ² = 16.131, p < 0.001). Moderate/severe disease activity, decreased myocardial perfusion and positive anti-SSA /Ro52kDa antibody were associated with impaired myocardial function in SLE patients (OR = 2.753, 5.359, 3.646; p = 0.049, 0.015, 0.014). Positive anti-SSA/Ro52kDa antibody was is independently correlated with myocardial dysfunction in SLE patients [OR (95%CI) = 3.159 (1.071–9.316), p = 0.037]. Conclusion 99mTc-MIBI rGMPI can noninvasively evaluate myocardial dysfunction in patients with SLE and provide evidence for clinical treatment decisions. Positive anti-SSA /Ro52kDa antibody was an independent risk factor for myocardial dysfunction in SLE patients.
... Hypertension, smoking, diet, and lack of physical activity are among the main risk factors. Inflammatory disorders, pathogens, pollution, and atrial diseases other than atrial fibrillation have recently been identified as risk factors and causes for stroke [48]. Traditional vascular risk factor management methods have been used to prevent stroke, transient ischemic attack and carotid revascularization, anticoagulation therapy, and other more basic treatments [49]. ...
Article
Full-text available
Although their risk factors and range of potential infections are bigger than those of older persons, younger adults have a far lower incidence of ischemic stroke. The age range of 18 to 50 accounts for less than 15% of persons who are vulnerable to stroke-related issues. Young people are still a heterogeneous population in the diagnostic process, hence it could be difficult to spot stroke mimics in them. This is the outcome of their ongoing diversity, it is well knowledge that the traditional stroke risk factors have risen. Among these risk factors which are frequently present in older persons are smoking, obesity, diabetes, dyslipidemia, and hypertension. Furthermore becoming more prevalent in younger patients who have had an acute stroke are these risk factors. Data from the most recent studies carried out in the United States and Europe show that the number of ischemic strokes among young individuals is rising. Young people having acute strokes are increasingly likely to have risk factors associated with both traditional cardiovascular disease and other cardiovascular disorders. Much debate surrounds the contribution of these traditional risk factors to strokes, if any. These challenges run the danger of aggravating the situation, ageing has been shown to have a major impact on the rising incidence, morbidity, and death rates related to stroke. A stroke is more likely to be survived by younger people than by those in the middle of their midlife. On the other hand, most survivors suffer social, psychological, and bodily effects that compel them to work or look for creche. This state has been created by the cooperation of these three factors. This makes precise prognostic forecasts for this patient population very crucial. A few investigations have concentrated on the ability of clinical, laboratory, and imaging variables to forecast the clinical course of an ischemic stroke and the likelihood of a recurrence in young patients.
... SLE can lead to an increased risk of accelerated atherosclerosis and cardiovascular (CV) events [128]. These include coronary heart disease, cerebrovascular disease and peripheral arterial disease. ...
Article
Full-text available
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the context of these systemic connective tissue diseases. Over the past few years, our understanding of the pathophysiology of atherosclerosis has changed from it being a lipid-centric to an inflammation-centric process. In this review, we examine the pathogenesis of atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis, the two most common systemic connective tissue diseases, and consider them as emblematic models of the effect of chronic inflammation on the human body. We explore the roles of the inflammasome, cells of the innate and acquired immune system, neutrophils, macrophages, lymphocytes, chemokines and soluble pro-inflammatory cytokines in rheumatoid arthritis and systemic lupus erythematosus, and the roles of certain autoantigens and autoantibodies, such as oxidized low-density lipoprotein and beta2-glycoprotein, which may play a pathogenetic role in atherosclerosis progression.
... antihypertensive medications is central; blockade of the reninangiotensin -aldosterone with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers is important for reducing proteinuria [45]. Additionally, assessment of the cardiovascular risk is essential, and the threshold for lipid-lowering agents should in general be low [46]. Non-pharmacological management is also important, including dietary interventions to restrict salt intake [47]. ...
Article
Full-text available
Introduction: Despite setbacks in clinical trials for systemic lupus erythematosus (SLE), three drugs have been approved for SLE and lupus nephritis (LN) treatment in the past decade. Several ongoing clinical trials, some viewed optimistically by the scientific community, underscore the evolving landscape. Emerging clinical data have established specific therapeutic targets in routine clinical practice for treating SLE, aiming to improve long-term outcomes. Areas covered: Research related to treatment of SLE and LN is discussed, focusing on randomized clinical trials during the last 5 years and recommendations for the management of SLE published by the European Alliance of Associations for Rheumatology (EULAR), American College of Rheumatology (ACR), Asia Pacific League of Associations for Rheumatology (APLAR), and Pan-American League of Associations of Rheumatology (PANLAR). Expert opinion: The landscape of SLE and LN treatments is evolving, as new drugs and combination treatment approaches redefine the traditional concepts of induction and maintenance treatment phases. As the therapeutic armamentarium in SLE continues to expand, the research focus is shifting from the imperative for new therapies to advancing our understanding of optimal treatment selection for individual patients, steering toward precision medicine strategies.
... Recent evidence has shown the influence of macro and micronutrients and bioactive foods compounds in alleviating the symptoms of SLE, resulting in a higher quality of life and a better prognosis for these patients 1,23,25 . In addition, the metabolic aging present in SLE and other autoimmune diseases seems to be similar to that which occurs in classic cardiovascular diseases, generating a possible association between SLE and the occurrence of accelerated atherosclerotic processes 26 . Considering this association, a healthy dietary pattern that helps control glycemic and lipid markers and maintain general health can have a positive impact on the lives of these patients 27 . ...
Article
Full-text available
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies directed against endogenous antigens causing various clinical manifestations, chronic inflammation and tissue damage. Although the pathophysiology of SLE remains unknown, it is recognized that genetic, epigenetic, environmental and neuroendocrine factors are involved in the development of the disease and its complications. A notable proportion of patients with SLE also present obesity, and this dysmetabolic profile can cause renal, musculoskeletal and/or respiratory deterioration, fatigue, various pathophysiological alterations and functional deterioration. In this context, precision nutrition emerges as a promising tool in the inflammatory control of SLE, especially in patients with associated obesity. Various studies demonstrate the beneficial influence of balanced dietary patterns in macronutrients with foods rich in fiber, vitamins, minerals, antioxidants and polyphenols on the inflammatory control of SLE and the most diverse pathologies, highlighting the Mediterranean diet and plant-based diets. Finally, the intestinal microbiota may play a relevant role in this clinical scenario, since dysbiosis is associated with inflammatory processes and immune deregulation. It is believed that precision nutrition can modulate inflammatory profiles and immune dysfunctions to ensure better quality of life and metabolic well-being of SLE patients with the support of precision omics technologies.
... 6 When present, coronary artery disease is directly linked to chronic SLE-related inflammation and impacts prognosis, highlighting the need for cardiac surveillance and risk factor modification, even in patients with optimal control of SLE activity. [7][8][9] In addition, recent data from patients with connective tissue disease suggest that diastolic dysfunction is more frequent than in the general population and is associated with poor survival. 10 Other markers of cardiac involvement, such as reductions in global longitudinal strain (GLS) or right ventricular systolic function, may also indicate a poorer prognosis. ...
Article
Full-text available
Background Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. Cardiac involvement in SLE is rare but plays an important prognostic role. The degree of cardiac involvement according to SLE subsets defined by non-cardiac manifestations is unknown. The objective of this study was to identify differences in transthoracic echocardiography (TTE) parameters associated with different SLE subgroups. Methods One hundred eighty-one patients who fulfilled the 2019 American College of Rheumatology/EULAR classification criteria for SLE and underwent baseline TTE were included in this cross-sectional study. We defined four subsets of SLE based on the predominant clinical manifestations. A multivariate multinomial regression analysis was performed to determine whether TTE parameters differed between groups. Results Four clinical subsets were defined according to non-cardiac clinical manifestations: group A (n=37 patients) showed features of mixed connective tissue disease, group B (n=76 patients) had primarily cutaneous involvement, group C (n=18) exhibited prominent serositis and group D (n=50) had severe, multi-organ involvement, including notable renal disease. Forty TTE parameters were assessed between groups. Per multivariate multinomial regression analysis, there were statistically significant differences in early diastolic tricuspid annular velocity (RV-Ea, p<0.0001), RV S’ wave (p=0.0031) and RV end-diastolic diameter (p=0.0419) between the groups. Group B (primarily cutaneous involvement) had the lowest degree of RV dysfunction. Conclusion When defining clinical phenotypes of SLE based on organ involvement, we found four distinct subgroups which showed notable differences in RV function on TTE. Risk-stratifying patients by clinical phenotype could help better tailor cardiac follow-up in this population.
... Despite improvement in survival rates in the last 70 years [2], the prognosis remains highly impacted by infections and cardiovascular events, which are major causes of mortality [3]. Apart from the traditional risk factors, steroids and aPLs increase the adverse cardiovascular profile in SLE [4][5][6]. APLs are present in 30-40% of SLE patients [7], of whom about one-third develop the clinical features, notably increased risk of blood clots and miscarriages typical of the clinically overt APS [8,9]. Patients tend to have either the clotting or obstetric clinical features rather than both [10,11]. ...
Article
Full-text available
aPLs are a major determinant of the increased cardiovascular risk in patients with SLE. They adversely affect clinical manifestations, damage accrual and prognosis. Apart from the antibodies included in the 2006 revised classification criteria for APS, other non-classical aPLs might help in identifying SLE patients at increased risk of thrombotic events. The best studied are IgA anti-β2-glycoprotein I, anti-domain I β2-glycoprotein I and aPS-PT. Major organ involvement includes kidney and neuropsychiatric systems. aPL/APS severely impacts pregnancy outcomes. Due to increased thrombotic risk, these patients require aggressive cardiovascular risk factor control. Primary prophylaxis is based on low-dose aspirin in high-risk patients. Warfarin is the gold-standard drug for secondary prophylaxis.
Article
To investigate whether two-dimensional speckle-tracking echocardiography (2DSTE) can be considered a criterion for early left ventricular (LV) systolic impairment in patients with systemic lupus erythematosus (SLE) and to further explore the association with each other. We included 38 patients with SLE and assessed the degree of disease activity according to the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) 2000 scoring criteria, together with 38 healthy controls who were matched by sex and age. Routine LV systolic function evaluation parameters were obtained by echocardiography as well as 2DSTE measurement of LV strain parameters to obtain global longitudinal strain (GLS) values, respectively. (I) On routine LV function parameters such as ejection fractions (EF) and left ventricular end-diastolic internal diameter (LVIDd), the SLE group and the control group did not reflect differences. In contrast, on the LV strain parameter obtained from 2DSTE measurements, the GLS values in all cardiac planes were lower in the SLE group than in the control group and showed statistically significant differences. (II) Correlation analysis showed that there was a correlation between SLEDAI and GLS, especially a meaningful correlation with GLS Avg and GLS A4C, with correlation coefficients of 0.35 and 0.47, respectively. The use of 2DSTE can detect early impaired LV systolic function in SLE patients, and GLS is progressively gaining attention as an indicator of subclinical myocardial injury and LV function in SLE patients. The correlation that exists between GLS and SLEDAI might contribute to a better assessment of cardiac involvement in SLE patients. Key Points • Cardiac involvement has become one of the major factors in the poor prognosis of SLE patients, which directly affects the mortality of SLE patients. Traditional echocardiography is difficult to detect early left ventricular function impairment, thus affecting clinicians’ judgment and diagnosis. • 2DSTE can recognize subclinical myocardial injury in SLE patients at an early stage, and its derived strain parameters may be used as an indicator to evaluate myocardial involvement and reflect disease activity in SLE patients
Article
Antecedentes: Los pacientes con lupus eritematoso sistémico (LES) tienen mayor riesgo de síndrome metabólico (SM) y enfermedad cardiovascular (ECV). El SM se evalúa de forma binaria, limitando la comprensión de la gravedad de los componentes por separado. Por lo anterior, se han desarrollado puntajes de gravedad de SM evaluándose individualmente. Este estudio tiene como objetivo determinar el pronóstico entre la gravedad del SM y el desarrollo de episodio cardiovascular mayor (ECM) en pacientes con LES. Metodología Estudio de cohorte con seguimiento de 10 años, incluyendo mujeres premenopáusicas > 18 años con LES. Se excluyeron pacientes con ECV, embarazo, enfermedad tiroidea y liposucción. La variable de interés fue ECV, y se examinaron las variables confusoras y los índices de gravedad de SM. Se estimaron las curvas de supervivencia de Kaplan-Meier y los cocientes de riesgo a través de la regresión de Cox. Resultados: Se analizaron 238 mujeres: 22 presentaron ECM y 216 no. La prevalencia de SM, evaluada según los criterios de consenso y ATP-III, fue mayor en pacientes con ECM(50 y 40,9%, respectivamente). El índice de gravedad MetSx-IMC fue más alto en el grupo de ECM. El análisis de Cox demostró un incremento en el MetSx-IMC, asociándose al aumento del riesgo de experimentar ECM en un factor de 1,107. Conclusiones: El índice de gravedad MetSx-IMC, contrario a los enfoques binarios, se recomienda para evaluar el SM como predictor de ECM en pacientes con LES. Ofrece mayor capacidad pronóstica y precisa de pacientes en riesgo de desarrollar ECM.
Article
Full-text available
Objectives: To identify predictors of low disease activity and clinical remission following belimumab treatment in SLE. Methods: SLE patients who received belimumab 10 mg/kg (N = 563) in the BLISS-52 and BLISS-76 clinical trials were surveyed. The performance of baseline factors in predicting attainment of low disease activity (defined as Lupus Low Disease Activity State) or clinical remission [defined as clinical (c)SLEDAI-2K = 0] at week 52 from treatment initiation was evaluated using logistic regression. Organ damage was assessed using the SLICC/ACR Damage Index (SDI). Results: We demonstrated a negative impact of established organ damage on attainment of Lupus Low Disease Activity State [SDI > 0; odds ratio (OR): 0.44; 95% CI 0.22, 0.90; P = 0.024] and the primary Lupus Low Disease Activity State condition, i.e. SLEDAI-2K ⩽ 4 with no renal activity, pleurisy, pericarditis or fever (SDI > 1; OR: 0.46; 95% CI 0.27, 0.77; P = 0.004); cognitive impairment/psychosis was found to mainly account for the latter association. Baseline SDI scores > 1 predicted failure to attain cSLEDAI-2K = 0 (OR: 0.53; 95% CI 0.30, 0.94; P = 0.030), with cutaneous damage mainly driving this association. Anti-dsDNA positivity increased (OR: 1.82; 95% CI 1.08, 3.06; P = 0.025) and cardiovascular damage reduced (OR: 0.13; 95% CI 0.02, 0.97; P = 0.047) the probability of attaining cSLEDAI-2K = 0 along with a daily prednisone equivalent intake restricted to ⩽7.5 mg. Conclusion: Belimumab might be expected to be more efficacious in inducing low disease activity and clinical remission in SLE patients with limited or no organ damage accrued prior to treatment initiation. Patients with positive anti-dsDNA titres might be more likely to achieve clinical remission along with limited or no CS use.
Article
Full-text available
The objective was to develop evidence-based recommendations for the management of antiphospholipid syndrome (APS) in adults. Based on evidence from a systematic literature review and expert opinion, overarching principles and recommendations were formulated and voted. High-risk antiphospholipid antibody (aPL) profile is associated with greater risk for thrombotic and obstetric APS. Risk modification includes screening for and management of cardiovascular and venous thrombosis risk factors, patient education about treatment adherence, and lifestyle counselling. Low-dose aspirin (LDA) is recommended for asymptomatic aPL carriers, patients with systemic lupus erythematosus without prior thrombotic or obstetric APS, and non-pregnant women with a history of obstetric APS only, all with high-risk aPL profiles. Patients with APS and first unprovoked venous thrombosis should receive long-term treatment with vitamin K antagonists (VKA) with a target international normalised ratio (INR) of 2–3. In patients with APS with first arterial thrombosis, treatment with VKA with INR 2–3 or INR 3–4 is recommended, considering the individual’s bleeding/thrombosis risk. Rivaroxaban should not be used in patients with APS with triple aPL positivity. For patients with recurrent arterial or venous thrombosis despite adequate treatment, addition of LDA, increase of INR target to 3–4 or switch to low molecular weight heparin may be considered. In women with prior obstetric APS, combination treatment with LDA and prophylactic dosage heparin during pregnancy is recommended. In patients with recurrent pregnancy complications, increase of heparin to therapeutic dose, addition of hydroxychloroquine or addition of low-dose prednisolone in the first trimester may be considered. These recommendations aim to guide treatment in adults with APS. High-quality evidence is limited, indicating a need for more research.
Article
Full-text available
Stroke is a major cause of morbidity, mortality and disability in systemic lupus erythematosus (SLE). Patients with SLE have a two-fold increase in the risk of stroke with younger patients (ie, less than 50 years of age) having an ever-higher risk (up to 10-fold). Although the prognosis of SLE has improved, mortality due to cerebrovascular events (CVE) remains unchanged. Cerebrovascular disease may be directly attributed to the disease per se, as a manifestation of neuropsychiatric SLE, or be the result of traditional cardiovascular risk factors accompanying the disease. Elucidation of the underlying mechanism(s) of CVE is essential as it may guide the type of therapy (ie, antithrombotic or anticoagulant therapy versus immunosuppressive). Strokes attributed to lupus usually occur early in the course of the disease and are often accompanied by evidence of activity in other organs; those related to antiphospholipid antibodies can occur at any time, in patients with either active or inactive SLE. In this review, we discuss the epidemiology, work-up, management and primary prevention of CVE in patients with lupus. In view of the effectiveness of thrombolysis, physicians need to educate lupus patients and their families for the early recognition of the signs of stroke and the need to seek prompt attention. To this end acronyms, such as FAST (Facial drooping, Arm weakness, Speech difficulties and Time to call emergency service) can be used as a mnemonic to help detect and enhance responsiveness to the needs of a person having a stroke.
Article
Full-text available
Early detection of subclinical atherosclerosis is important to reduce patients' cardiovascular risk. However, current diagnostic strategy focusing on traditional risk factors or using risk scoring is not satisfactory. Non-invasive imaging tools also have limitations such as cost, time, radiation hazard, renal toxicity, and requirement for specialized techniques or instruments. There is a close interaction between arterial stiffness and atherosclerosis. Increased luminal pressure and shear stress by arterial stiffening causes endothelial dysfunction, accelerates the formation of atheroma, and stimulates excessive collagen production and deposition in the arterial wall, leading to the progression of atherosclerosis. Pulse wave velocity (PWV), the most widely used measure of arterial stiffness, has emerged as a useful tool for the diagnosis and risk stratification of cardiovascular disease (CVD). The measurement of PWV is simple, non-invasive, and reproducible. There have been many clinical studies and meta-analyses showing the association between PWV and coronary/cerebral/carotid atherosclerosis. More importantly, longitudinal studies have shown that PWV is a significant risk factor for future CVD independent of well-known cardiovascular risk factors. The measurement of PWV may be a useful tool to select subjects at high risk of developing subclinical atherosclerosis or CVD especially in mass screening.
Article
Full-text available
Background: Dyslipidemia is a common disorder in systemic lupus erythematosus (SLE) patients. It is still inconclusive whether antimalarial drugs could affect the serum lipids in SLE patients, therefore we conducted a systematic review and meta-analysis of available data to address this issue. Methods: We comprehensively searched the databases of PubMed, EMBASE and Cochrane Library from date of inception to Sep 2018 for both randomized controlled trials (RCTs) and observational studies. Review Manager 5.3 software was used for analysis. We performed meta-analysis using random-effects model and weighted the mean difference (WMD) and its 95% confidence interval (CI). The Q test was used to assess the presence of heterogeneity and the I index was used to quantify the extent of heterogeneity. Results: In total, 8 studies met our selection criteria including 2 RCTs, 2 cohort studies, and 4 case-control studies. There were 717 patients (336 patients in CQ (chloroquine) or HCQ (hydroxychloroquine) group, and 381 patients in control group (SLE patients without the therapy of AM)). Compared with the control group, TC, TG, LDL-C, VLDL-C were associated with a significant decrease, respectively (WMD = -21.40 mg/dL, 95% CI -27.62 to -15.18, P < .00001), (WMD = -29.07 mg/dL, 95% CI -45.28 to -12.86, P = .0004), (WMD = -16.25 mg/dL, 95% CI -28.82 to -3.68, P = .01), (WMD = -6.41 mg/dL, 95% CI -12.39 to 0.44, P = .04), however the change of HDL-C did not reach statistically significance (WMD = 4.42 mg/dL, 95% CI -1.21 to 10.06, P = .12). Conclusions: CQ or HCQ can infect the serum lipids in SLE patients. However, these results should be interpreted with cautions since lacking sufficient RCTs.
Article
Full-text available
Our objective was to update the EULAR recommendations for the management of systemic lupus erythematosus (SLE), based on emerging new evidence. We performed a systematic literature review (01/2007–12/2017), followed by modified Delphi method, to form questions, elicit expert opinions and reach consensus. Treatment in SLE aims at remission or low disease activity and prevention of flares. Hydroxychloroquine is recommended in all patients with lupus, at a dose not exceeding 5 mg/kg real body weight. During chronic maintenance treatment, glucocorticoids (GC) should be minimised to less than 7.5 mg/day (prednisone equivalent) and, when possible, withdrawn. Appropriate initiation of immunomodulatory agents (methotrexate, azathioprine, mycophenolate) can expedite the tapering/discontinuation of GC. In persistently active or flaring extrarenal disease, add-on belimumab should be considered; rituximab (RTX) may be considered in organ-threatening, refractory disease. Updated specific recommendations are also provided for cutaneous, neuropsychiatric, haematological and renal disease. Patients with SLE should be assessed for their antiphospholipid antibody status, infectious and cardiovascular diseases risk profile and preventative strategies be tailored accordingly. The updated recommendations provide physicians and patients with updated consensus guidance on the management of SLE, combining evidence-base and expert-opinion.
Article
It has been believed that most acute coronary events result from the rupture of mildly stenotic plaques, based on studies in which angiographic information was available from many months to years before the event. However, serial studies in which angiographic data were available from the past as also within 1 to 3 months of myocardial infarction have clarified that nonobstructive lesions progressively enlarged relatively rapidly before the acute event occurred. Noninvasive computed tomography angiography imaging data have confirmed that lesions that did not progress voluminously over time rarely led to events, regardless of the extent of luminal stenosis or baseline high-risk plaque morphology. Therefore, plaque progression could be proposed as a necessary step between early, uncomplicated atherosclerosis and plaque rupture. On the other hand, it has been convincingly demonstrated that intensive lipid-lowering therapy (to a low-density lipoprotein cholesterol level of <70 mg/dl) halts plaque progression. Given the current ability to noninvasively detect the presence of early atherosclerosis, the importance of plaque progression in the pathogenesis of myocardial infarction, and the efficacy of maximum lipid-lowering therapy, it has been suggested that plaque progression is a modifiable step in the evolution of atherosclerotic plaque. A personalized approach based on the detection of early atherosclerosis can trigger the necessary treatment to prevent plaque progression and hence plaque instability. Therefore, this approach can redefine the traditional paradigm of primary and secondary prevention based on population-derived risk estimates and can potentially improve long-term outcomes.
Article
Background Survival in systemic lupus erythematosus (SLE) has improved substantially in the last 50 years. The aim of the present study was to assess the evolution of the all-cause, cause-specific and age-specific standardised mortality ratios (SMRs) of patients with lupus in Ontario, Canada. Patients and methods Between 1971 and 2013, 1732 patients were followed in the Toronto Lupus Clinic. Causes of death were retrieved from death certificates, autopsy reports, hospital records or the records of the family physicians. They were categorised as atherosclerotic, infectious, malignancy, active lupus and others. For the calculation of the SMR (indirect standardisation method), data from the general population of Ontario, Canada were used (Statistics Canada). Results Two hundred and forty-nine patients (205 women) died (infections 24.5%, atherosclerosis 15.7%, active lupus 13.3%, malignancy 9.6%); mean age was 53.2±16.6 years and mean disease duration 15.2±11.7 years. The all-cause SMR was substantially decreased from the 1970s (13.5, 95% CI 8.6 to 18.5) to recent years (2.2, 95% CI 1.4 to 3.1). Similar trends were observed for atherosclerosis, infections and malignancies over time. The all-cause age-specific SMR was particularly high in younger (19–39 years old) patients (SMR=12.4, 95% CI 9.7 to 15.1) as compared with individuals older than 40 years (SMR=3.1, 95% CI 2.6 to 3.6). The cause-specific SMR was also higher in younger patients, particularly for infections and malignancies. Conclusions The all-cause and cause-specific SMR significantly decreased over time, likely reflecting the advances in the management of SLE and certain comorbidities. The all-cause and cause-specific SMR was particularly high for younger patients (<40 years old).