Buscar en
Revista Colombiana de Reumatología
Toda la web
Inicio Revista Colombiana de Reumatología Accelerated atherosclerosis and cardiovascular disease in systemic lupus erythem...
Información de la revista
Vol. 28. Núm. S1.
Systemic Lupus Erythematosus
Páginas 21-30 (Junio 2021)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
2239
Vol. 28. Núm. S1.
Systemic Lupus Erythematosus
Páginas 21-30 (Junio 2021)
Review Article
Acceso a texto completo
Accelerated atherosclerosis and cardiovascular disease in systemic lupus erythematosus
Aterosclerosis acelerada y enfermedad cardiovascular en el lupus eritematoso sistémico
Visitas
2239
Rosana Quintanaa,
Autor para correspondencia
rosanaquintana@gmail.com

Corresponding author.
, Guillermo J. Pons-Estela, Rosa Serranoa, Bernardo A. Pons-Estela, Ian N. Bruceb
a Regional Center for Autoimmune and Rheumatic Diseases, Oroño Group (GO-CREAR), Rosario, Argentina
b Centre for Epidemiology Versus Arthritis, Faculty of Biology, Medicine and Health, The University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (2)
Material adicional (1)
Suplemento especial
Este artículo forma parte de:
Vol. 28. Núm S1

Systemic Lupus Erythematosus

Más datos
Abstract

Cardiovascular disease (CVD), particularly coronary heart disease and stroke, is one of the most important causes of morbidity and mortality in patients with systemic lupus erythematosus (SLE). The increased prevalence of CVD and subclinical atherosclerosis, even after adjustment for traditional risk factors, are well established. Several associations with disease-related clinical, genetic and immunological features have been identified. The SLE-specific stratification algorithms with emphasis on composite risk-assessment scores including both traditional risk factors and novel biomarkers is recommended. The clinical complexity of accelerated atherosclerosis will most likely require an integrated approach for the identification, treatment, and intensive study into this aspect of SLE that will ultimately lead to improved cardiovascular outcomes for these patients.

Keywords:
Cardiovascular disease
Systemic lupus erythematosus
Accelerated atherosclerosis
Traditional risk factors
Lupus-specific risk factors
Antimalarial recommendations
Resumen

La enfermedad cardiovascular (ECV), en particular la enfermedad coronaria y el ictus, es una de las causas más importantes de morbimortalidad en pacientes con lupus eritematoso sistémico (LES). El aumento en la prevalencia de la ECV y de la aterosclerosis subclínica, aun después del ajuste de los factores de riesgo tradicionales, está claramente establecida. Se han identificado diversas asociaciones con características clínicas, genéticas e inmunológicas relacionadas con la enfermedad. Se recomienda el uso de los algoritmos de estratificación específicos para el LES, con énfasis en los puntajes compuestos de evaluación de riesgo, incluyendo tanto los factores de riesgo tradicionales como los nuevos biomarcadores. La complejidad clínica de la aterosclerosis acelerada muy probablemente requerirá un abordaje integral para la identificación, el tratamiento y el estudio intensivo de este aspecto del LES, que en última instancia permita obtener mejores desenlaces cardiovasculares en estos pacientes.

Palabras clave:
Enfermedad cardiovascular
Lupus eritematoso sistémico
Aterosclerosis acelerada
Factores tradicionales de riesgo
Factores de riesgo específicos del lupus
Antimaláricos
Texto completo
Introduction

Cumulative evidence over the last 30 years strongly supports the active involvement of the immune system in the development of atherosclerotic plaque as well as the interconnection between chronic autoimmune/inflammatory disorders and excessive cardiovascular burden, not easily attributed to traditional cardiovascular risk factors (CVRFs).1–3

In young women with SLE, the risk for myocardial infarction (MI) has been found to be 50 times higher compared to healthy women of similar age distribution.4,5 While traditional risk factors associated with atherosclerosis including smoking, dyslipidemia, diabetes mellitus, hypertension and increased body mass index (BMI) are present in lupus patients, standard Framingham scores do not fully explain the high rates of ischemic events so far reported. It has therefore been proposed that the atherosclerotic process is accelerated in patients with SLE due to a complex interplay of traditional and lupus-specific risk factors.6–8 SLE is known to be an independent risk factor for endothelial dysfunction.9

Epidemiology of CVD in SLE

SLE is a heterogeneous autoimmune disease, affecting women of childbearing age, with substantial morbidity and mortality. The effect of SLE on atherosclerotic disease has been recognized since the 1970s, when Urowitz et al. observed a bimodal mortality peak for lupus patients; the first was attributed to disease activity and infections and the second to CVD.10 Three decades later, progress in disease treatment had resulted in decrease of mortality due to disease activity; however, CVD and infectious complications remain the main causes of death in these patients.11 The prevalence of ischemic heart disease in SLE patients is estimated between 3.8% and 16%,12,13 conferring a 10-fold risk compared to the general population and a 50-fold risk in young women of reproductive age.1,4,14 The risk of stroke in SLE patients was also found to be increased by 2–8 fold in different studies.14,15

About 30–40% of patients with SLE show evidence of subclinical atherosclerotic lesions, depending on the diagnostic methods. The excess of carotid plaque in SLE is particularly striking in those under 55 years old and it has consistently been shown that patients with SLE have significantly higher prevalence of atherosclerotic plaque than healthy controls.16–20

In addition to macrovasculature abnormalities in SLE, there is evidence to suggest abnormal coronary microvascular function. When positron emission tomography was used, abnormal coronary flow reserve was seen even in SLE patients with normal coronary arteries.21 Abnormal stress myocardial perfusion imaging (shown by adenosine stress cardiac magnetic resonance imaging) was found in 44% of SLE patients with angina and chest pain in the absence of obstruction. Quantitative myocardial perfusion reserve index was also observed to be lower in patients with SLE as compared with controls, and the presence of SLE was a significant predictor of myocardial perfusion reserve index.22

Traditional CVD risk factors in SLEMetabolic syndrome

Metabolic syndrome (MetS) is a constellation of central obesity, insulin resistance, dyslipidemia and hypertension, that was found to be prevalent in lupus patients compared to age matched controls at a rate ranging from 15.8 to 32.4% vs. 4.2% to 10.9%, depending on the mean age of the study subjects and the definitions used.23,24 In lupus patients the presence of MetS has been associated with racial/ethnic background (Hispanic or Black African), increasing age and disease-related characteristics such as baseline renal disease, Systemic Lupus International Collaborative Clinics damage index (SDI) >1 and higher disease activity, as well as coronary atherosclerosis, arterial stiffness and inflammatory biomarkers.1

Increased waist-to-hip ratio, sedentary lifestyle and obesity are more prevalent in SLE patients compared to controls.25,26 Interestingly, increased BMI levels were found to be significantly associated with subclinical atherosclerosis in both adult and pediatric lupus populations.27

Insulin resistance is also more frequent in lupus patients compared to controls (44.1 vs. 24.8%) in association with high BMI, waist circumference, hypertension, corticosteroid treatment and SDI.28

Hypertension

The prevalence of arterial hypertension in lupus patients ranges from 33% to 74%29,30 and is a recognized risk factor for the development of CVD in SLE patients,13,31 in addition to its contribution to both plaque formation and arterial stiffening.32,33 A longitudinal study, explored the determinants of atherosclerosis progression in 187 SLE patients, identifying age and hypertension as independently associated factors with the progression of carotid intima-medial thickness (IMT) and plaque formation.34 Another study, identified that renal disease, insulin levels and SLE disease activity index (SLEDAI) were independent predictors of hypertension in SLE.29 Notably, non-obesity-related insulin levels was the main predictor of hypertension in the younger age subset (<40 years), while age and obesity were the predictors among the older group (≥40 years). In a subsequent study, examining the patterns of night-time blood pressure in female lupus patients, an adverse night-time blood pressure pattern (steady, non-dipping hypertension or nocturnal hypertension/reverse dipping) was more frequent in SLE; these patterns were independently associated with increased carotid-femoral pulse wave velocity.35

Dyslipidemia

The association of increased levels of total cholesterol, low density lipoprotein (LDL) and decreased levels of high-density lipoprotein (HDL) cholesterol with increased risk for CVD in the general population is well established and acknowledged for many years.36,37 Reported rates of dyslipidemia in SLE patients range from 36% at diagnosis, to more than 60% within a three year follow-up.38 The classical pattern, is characterized by elevated levels of very-low-density lipoprotein cholesterol (VLDL), triglycerides and low levels of HDL which can be aggravated by disease activity.39 In addition, SLE patients tend to have a more atherogenic LDL phenotype characterized by small dense LDL molecules.40 Likewise, circulating lipoprotein remnant particles and the intermediate density lipoprotein (IDL) fraction have also been strongly associated with IMT values in lupus patients, while small HDL particles have been associated with activation of the complement system, also shown to be linked to higher IMT values.1 A newly recognized proinflammatory HDL subtype (piHDLs), is also present in a high proportion of patients with SLE and is associated with carotid artery plaque and clinical CVD.41 Furthermore, levels of apolipoprotein A-I (apoA-I) are reduced in SLE patients with IgG anticardiolipin antibodies.42

Smoking

Smoking has been associated with CVD, cerebrovascular and peripheral vascular events30,43 as well as with markers of subclinical atherosclerosis. Smoking has also been identified as a risk factor for progression of coronary artery calcification after adjusting for age, gender and ethnicity.34

Hyperhomocysteinemia

Elevated homocysteine is a prothrombotic coagulation factor with toxic effects on the endothelium, increased collagen production and decreased nitric oxide availability.1

Hyperhomocysteinemia is a recognized risk factor for premature atherosclerosis and thrombotic risk in the general population because of its adverse effects on the endothelium, inhibition of nitric oxide synthesis, proliferation of smooth muscle cells and platelet activation.44,45 The increase of homocysteine levels in lupus patients compared to healthy controls ranges from 11.6 to 81.2% vs. 0.8 to 20%.46,47 Elevated homocysteine was found to be associated with subsequent development of coronary artery disease, thrombotic events and markers of subclinical atherosclerosis.13,48

Non-traditional risk factorsDisease related features

Antiphospholipid antibodies (aPLs), impaired renal function as well as low total white blood cell count, lymphopenia and renal disease have all been associated with carotid IMT and arterial stiffness.1,49,50 Carotid plaque may occur twice as commonly in SLE patients with nephritis compared to age-matched non-nephritis SLE patients and population controls. This excess risk among nephritis individuals was mainly attributed to concomitant hypertension.49 Disease duration, chronic organ damage (reflected by SDI) and disease activity were identified by numerous studies as important factors for CVD development in the setting of lupus.8,50,51 Longer lupus duration has also been independently associated with coronary artery calcification and carotid plaque formation, as well as progression. Similarly, the SDI score was found to be independently associated with increased IMT scores, carotid plaque formation, clinical CVD and arterial stiffness.4,52,53

Treatments

Long-term corticosteroid use has been associated with MI and angina, while higher cumulative doses of steroids are also associated with carotid plaque formation.4,53 Azathioprine and cyclophosphamide use has also been associated with increased rates of clinical CVD, carotid plaque and higher carotid IMT values, in addition to an independent determinant of carotid plaque.1,54 The association with immunosuppression and steroids may partially reflect unaccounted confounders due to the severity of the disease; however there is good evidence that glucocorticoids can exacerbate a number of classic risk factors including hypertension, impaired glucose tolerance and dyslipidemia.

Antimalarials (AM) are commonly used in lupus treatment and are reported to be beneficial against CVD through cholesterol lowering, reduction of thrombotic risk and possibly through dampening of type I interferon (IFN) production.55–57 In addition, AM use has been inversely associated with plaque and carotid/femoral arterial stiffness and was shown to be protective against MetS.32,54,58 Mycophenolate mofetil may also play a potentially beneficial role to prevent atherosclerosis progression.59

Autoantibodies

Anti-endothelial cell antibodies (AECA) and aPLs seem to play a significant role, although the underlying mechanisms are not fully elucidated. AECA can directly activate endothelial cells and are detected in 73% of SLE patients; however, their clinical relevance has not been clearly confirmed.60,61

APLs have been shown to activate the endothelium and inhibit annexin A5-binding, a protein shown to prevent plaque rupture to the endothelium.62 APLs have also been identified as independent predictors of cerebrovascular or peripheral vascular events and MI.4,63

The presence of anti-oxidized low-density lipoprotein (OxLDL) antibodies has been identified in up to 80% of SLE patients with antiphospholipid syndrome (APS) and are more commonly found in SLE patients with a history of CVD.64 Traditional and non-traditional risk factors for CVD in SLE are listed in Fig. 1.

Fig. 1.

Cardiovascular disease in SLE.

(0,15MB).
Pathogenic mechanisms

The mechanisms of the increased and accelerated atherosclerotic risk for SLE patients remain to be determined. It is likely that multiple mechanisms are operative, resulting from a complex interplay between traditional cardiac risk factors and SLE-driven inflammation.65 Atherosclerosis is not just a consequence of passive accumulation of lipids in the vessel wall, but also a result of inflammation. As in the pathogenesis of SLE itself, the interplay of multiple inflammatory mediators, including leukocytes, cytokines, chemokines, adhesion molecules, complement, and antibodies, results in the formation of atherosclerotic plaques.66 In response to different factors such as hemodynamic stresses or inflammatory mediators, the vascular endothelium undergoes a series of inflammatory changes that may result in AECA formation. Activated endothelial cells up-regulate leukocyte adhesion molecules such as vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and E-selectin. Chemoattractant cytokines such as monocyte chemoattractant protein1 (MCP-1), interleukin 6 (IL-6) and IL-8 are also expressed, thus inducing a cascade of pro-inflammatory, pro-atherogenic changes in the endothelium that result in migration of monocytes into the subendothelial space.67 T-cells are also recruited into the subendothelium by similar mechanisms, although in lower numbers. LDL molecules are transported into the arterial wall where they become trapped and when exposed to reactive oxygen species, become oxLDL. OxLDL can stimulate AECA formation and are also phagocytized by infiltrating monocytes/macrophages where they form foam cells around which atherosclerotic lesions are built. Monocytes and T cells infiltrate the margin of the plaque formed by foam cells. Muscle cells from the media of the artery are stimulated to grow, and ultimately encroach on the vessel lumen.68

In this context, IgG deposition may be proatherogenic by the formation of oxLDL-containing complexes and subsequent activation of macrophages and dendritic cells which can promote atherosclerosis by also inhibiting regulatory T cells (Tregs). In SLE vascular damage is accelerated and vascular repair mechanisms are less effective. SLE patients have high levels of circulating apoptotic endothelial cells, indicating increased vascular damage, and lower levels of circulating endothelial progenitor cells (EPC) that repair damaged arterial tissues.69 The production of reparative myelomonocytic circulating angiogenic cells (CAC) is also impaired. Secretion of IFNα, stimulated in part by low density granulocytes undergoing neutrophil extracellular traps (NETosis), also increased in SLE patients, induces EPC apoptosis and converts CAC to dendritic cells, thus losing their ability to repair vascular damage to endothelial cells.70

Potential biomarkers for atherosclerosis in SLEAntiphospholipid antibodies

In the LUMINA (Lupus in Minorities: Nature vs. nurture) cohort study, aPLs were an independent risk factor for cardiovascular or cerebrovascular events.43 In the Hopkins Lupus cohort, lupus anticoagulant was the only aPLs associated with MI.63 Several studies using measures of subclinical atherosclerosis failed to identify any significant associations with aPLs after adjustment for confounding factors,4,6,54 although Ahmad et al. did show an association between carotid plaque and APL, as did a follow-up study from the same cohort, suggesting a direct role in atherogenesis, as well as a role in precipitating arterial events.16,50

C-reactive protein

C-reactive protein (CRP) is not only a marker of systemic inflammation, but rather may play a direct role in the pathogenesis of atherosclerosis. In SLE subjects, however, the role of CRP as a predictor of atherosclerosis is less clear. Elevated CRP levels have been associated with cardiovascular events in the LUMINA cohort51 and high-sensitivity CRP (hs-CRP) levels were associated with cardiovascular mortality in a prospective Swedish Lupus Cohort.71 Hs-CRP has also been associated with both cross-sectional32 and longitudinal progression of carotid IMT.34 Several other studies, however, did not find an association between atherosclerosis and CRP in SLE.6,54

Pro-inflammatory HDL (piHDL)

During states of chronic inflammation, HDL can be converted from its anti-inflammatory to pro-inflammatory state.65 HDL function is abnormal in women with SLE; 45% of women with SLE, compared to 20% of rheumatoid arthritis patients and 4% of controls, had piHDL that was unable to prevent oxidation of LDL.65 HDL dysfunction has also been described in primary APS and piHDL is strongly associated with progression of carotid plaque and IMT.65,72

Paraoxonase

Serum paraoxonase 1 (PON1) has been identified as one of the important components of HDL that prevents lipid peroxidation and blocks the pro-inflammatory effects.73 Decreased levels of PON1 activity have also been associated with atherosclerosis in the general population.74 Altered levels of PON1 activity have also been seen in patients with SLE. In one study, PON1 activity was reduced in SLE and APS patients compared to controls, although there was no reduction in the total antioxidant capacity of the plasma.75 In another study of 55 SLE patients, titers of anti-apoA1 antibodies were inversely correlated to PON1 activity, and in vitro studies confirmed that apo-AI antibodies have a direct inhibitory effect on PON1 activity.76 Decreased PON1 activity has been associated with increased carotid artery IMT and abnormal flow-mediated dilation in patients with primary APS.76

Adipocytokines

Adipokine leptin is an anorectic peptide; patients with MetS have high circulating leptin levels, but they develop leptin resistance similar to insulin resistance in type II diabetes.77 Several small cohort studies have shown elevated leptin levels in adult SLE patients.78,79 Leptin levels were significantly higher in the SLE patients with carotid plaque versus those without plaque, and also weakly correlated with carotid IMT.65 In another cohort, adiponectin levels were significantly and independently associated with carotid plaque in SLE.80

Homocysteine

Homocysteine is another predictor of atherosclerosis in the general population.81 Homocysteine may play a direct role in the pathogenesis of SLE through its toxic effects on the endothelium.82 Hyperhomocysteinemia can result from advanced age, renal insufficiency, medications such as methotrexate, genetic, and/or dietary factors.65

In one cohort study of 337 SLE patients, hyperhomocysteinemia was an independent predictor of stroke and cardiovascular events.13 In several other studies, elevated levels of homocysteine in SLE correlated with progression of subclinical atherosclerosis in SLE.34,54,83,84

Vitamin D levels

Recently, low vitamin D has emerged as a potential biomarker of CVD. Patients with low vitamin D levels or with a vitamin D deficiency were found to have a high prevalence of CVRFs including dyslipidemia, hypertension, MetS, aPLs and increased hs-CRP level.85 In a prospective study of 890 patients with SLE in a large international inception cohort, multiple logistic regression analyses revealed that patients in the high quartiles of 25-hydroxyvitamin D (25-(OH) D) were less likely to present CVRFs, including hypertension and hyperlipidemia, while a non-significant trend of a decreasing hazard ratio of cardiovascular events was noted across successively higher quartiles of 25-(OH) D levels.86 A few studies have addressed the potential relationship between hypovitaminosis D and the unfavorable alterations of these biophysical cardiovascular risk markers. A study showed that patients with arterial stiffness exhibited higher levels of serum vitamin D and most of them were on vitamin D-Ca supplements. Prospective studies with a larger number of patients and follow-up are needed.87 The potential biomarkers for CVD in SLE are depicted in Table 1.

Table 1.

Potential biomarkers for cardiovascular disease in SLE.

• Antiphospholipid antibodies 
• C-reactive protein 
• Pro-inflammatory HDL 
• Paraoxonase 
• Adipocytokines 
• Homocysteine 
• Vitamin D levels 
Subclinical measures of atherosclerosis

It has been argued that the identification asymptomatic patients with significant subclinical disease is the key for targeted primary prevention of symptomatic CVD. Based on this premise, risk stratification algorithms have been developed and refined in an attempt to estimate the future risk of cardiovascular events with the highest possible predictive value. For the general population, most risk stratification tools are based on levels of well-established CVRFs such as the Framingham risk score.88 Other examples are the Reynolds score89 which includes high-sensitivity CRP, the Sheffield table system,90 and the SCORE system in Europe. These different methods are similar in their overall low sensitivity and specificity for development of CVD as they exclude various emerging, genetic and otherwise unknown risk factors. The majority of these indices fail to take into account the presence of autoimmune diseases. Indeed, several studies have shown that such population risk score systems systematically underestimate the risk of future CVD in SLE and hence should not be used for risk stratification in SLE.

The use of SLE-specific stratification algorithms has been suggested, with particular emphasis on composite risk-assessment scores, including both traditional risk factors and novel biomarkers. The Predictors of Risk for Elevated Flares, Damage Progression, and Increased Cardiovascular disease in Patients with SLE (PREDICTS) score was proposed on the basis of the presence of at least three positive biomarkers or a combination of diabetes plus at least one of the biomarkers considered.65 The QRISK algorithm has been used in the UK to calculate the likelihood of a major cardiovascular event over the following 10-year period; QRISK3 risk score included clinical variables such as chronic kidney disease, migraine, corticosteroids and SLE.91 The QRISK3 score has been shown to identify a much higher proportion of SLE patients as having a >10% 10-year risk of CVD.92

The European League Against Rheumatism (EULAR) recommendation was the calculation of the 10-year CVD risk using the Systematic Coronary Risk Evaluation (SCORE) although the actual risk is underestimated in patients with SLE.93

Other modalities have also been used to screen for subclinical atherosclerosis in SLE patients such as ultrasound assessment of the carotid that allows for the assessment of arterial wall thickness and degree of plaque. Patients with higher cumulative damage measured by the modified SDI damage score were more likely to have plaque.4

Improved ultrasound assessment of carotid plaques can be achieved using integrated backscatter analysis of the carotid-intima complex, which appears to correlate with calcium and collagen content of the vascular wall, for a non-invasive evaluation of arterial sclerosis.94 Some argue that the femoral arteries should also be scanned because in non-SLE patients, femoral plaque is also associated with increased risk of coronary disease without carotid affection.95 An alternative method to ultrasound is high-resolution computed tomography angiography, which focuses essentially on providing improved accuracy and sensitivity and allows for a ‘virtual’ histology of plaques as it has been shown to correlate with histological findings of atheromatous plaques at the carotid bifurcation.96 However, none of these imaging techniques are recommended in routine practice for CVD screening.

Management strategies for prevention and treatment of CVD outcomes

Interventions aimed at promoting health include dietary modification, exercise and smoking cessation and are generally recommended as they can contribute to improving long-term patient outcomes. Appropriate disease management focused on the remission of symptoms and signs, prevention of damage accrual and improved quality of life may also all contribute to improving CVD risk.

Lipid-lowering therapy

In patients with SLE and hyperlipidemia, mortality was lower in patients taking statin therapy; furthermore, these patients had a lower incidence of myocardial infarction and stroke.97 Interestingly, several trials have failed to find any significant changes in surrogate markers of atherosclerosis with statin therapy. In a study in adults with SLE without previous cardiovascular events, patients were randomized to atorvastatin 40mg/day or placebo; there was no statistically significant difference in progression of coronary calcification scores over two years.31 In contrast, in another study lower coronary calcium deposits measured by computed tomography scan were observed in SLE patients without CVRFs after one year of treatment with 40mg atorvastatin compared to placebo.98 In children with SLE, a further study found no benefit in the progression of carotid IMT in those randomized to atorvastatin.

Overall, these trials suggest that statin therapy should not be routinely offered to all SLE patients as standard of care. The ideal approach to targeting SLE patients for lipid-lowering therapies therefore remains unclear, but is likely to be appropriate for patients with elevated LDL and in whom lifestyle changes and non-pharmacological approaches are not successful in achieving an ideal LDL target.

Antiplatelet and anticoagulant agents

Two meta-analyses studied the effect of antithrombotic drugs (antiplatelet agents and anticoagulants) on cardiovascular risk in SLE patients.99 The main outcome was the efficacy of acetylsalicylic acid for the primary prevention of thrombotic events in patients with SLE and CVRFs. The results from both meta-analyses indicated that acetylsalicylic acid is effective for the primary prevention of thrombosis. Another study did not show any differences in the rate of thrombotic events between patients with ASA alone, or with ASA plus warfarin; however, the rate of thrombotic events in the untreated group was twice the rate in the intervention group.100 Another study assessed the effectiveness of primary (acetylsalicylic acid) and secondary prevention (acetylsalicylic acid+cumarine) of antithrombotic therapy. Three different groups were analyzed: patients with SLE and positive aPLs, patients with SLE and APS and patients with SLE and negative aPLs. The occurrence of cardiovascular events was lower in all groups.101

Acetylsalicylic acid should therefore be considered for all SLE patients, especially those with a significant cardiovascular risk (positive aPLs, hypertension, dyslipidemia), unless contraindicated.102

Specific SLE treatment

Several studies have analyzed the effectiveness of AM on cardiovascular risk in SLE. The use of AM in patients with SLE and CVRFs is useful for the primary prevention of thrombotic events as they have known positive effects on glucose levels, insulin resistance and LDL levels.103–105

The discrepancy among the studies on the effectiveness of AM in CVD prevention may be due to differences in terms of the presence/absence of a control group and disease status, variable definition of clinical outcomes and heterogeneity in the ethnic composition of the study populations and, most importantly, on the different duration of exposure of patients to AM prior to the event.102 Indeed, a time-dependent effect of AM exposure has been suggested by other studies.33,56 Strategies for prevention and treatment of CVD in SLE are depicted in Fig. 2.

Fig. 2.

Prevention and treatment of CVD in SLE.

(0,14MB).
Conflict of interest

None.

Appendix A
Supplementary material

The Spanish translation of this article is available as supplementary material:

References
[1]
M. Giannelou, C.P. Mavragani.
Cardiovascular disease in systemic lupus erythematosus: a comprehensive update.
J Autoimmun, 82 (2017), pp. 1-12
[2]
A. Gistera, G.K. Hansson.
The immunology of atherosclerosis.
Nat Rev Nephrol, 13 (2017), pp. 368-380
[3]
J.E. Salmon, M.J. Roman.
Subclinical atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus.
[4]
S. Manzi, E.N. Meilahn, J.E. Rairie, C.G. Conte, T.A. Medsger Jr., L. Jansen-McWilliams, et al.
Age-specific incidence rates of myocardial infarction and angina in women with systemic lupus erythematosus: comparison with the Framingham study.
Am J Epidemiol, 145 (1997), pp. 408-415
[5]
L.J. Catoggio, E.R. Soriano, P.M. Imamura, D. Wojdyla, S. Jacobelli, L. Massardo, et al.
Late-onset systemic lupus erythematosus in Latin Americans: a distinct subgroup?.
Lupus, 24 (2015), pp. 788-795
[6]
Y. Asanuma, A. Oeser, A.K. Shintani, E. Turner, N. Olsen, S. Fazio, et al.
Premature coronary-artery atherosclerosis in systemic lupus erythematosus.
N Engl J Med, 349 (2003), pp. 2407-2415
[7]
I.N. Bruce.
‘Not only..but also’: factors that contribute to accelerated atherosclerosis and premature coronary heart disease in systemic lupus erythematosus.
Rheumatology (Oxford), 44 (2005), pp. 1492-1502
[8]
S. Haque, S. Skeoch, C. Rakieh, H. Edlin, Y. Ahmad, P. Ho, 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, 5 (2018), pp. e000267
[9]
M. El-Magadmi, H. Bodill, Y. Ahmad, P.N. Durrington, M. Mackness, M. Walker, et al.
Systemic lupus erythematosus: an independent risk factor for endothelial dysfunction in women.
Circulation, 110 (2004), pp. 399-404
[10]
M.B. Urowitz, A.A. Bookman, B.E. Koehler, D.A. Gordon, H.A. Smythe, M.A. Ogryzlo.
The bimodal mortality pattern of systemic lupus erythematosus.
Am J Med, 60 (1976), pp. 221-225
[11]
J. Nossent, N. Cikes, E. Kiss, A. Marchesoni, V. Nassonova, M. Mosca, et al.
Current causes of death in systemic lupus erythematosus in Europe, 2000–2004: relation to disease activity and damage accrual.
Lupus, 16 (2007), pp. 309-317
[12]
D.D. Gladman, M.B. Urowitz.
Morbidity in systemic lupus erythematosus.
J Rheumatol Suppl, 14 (1987), pp. 223-226
[13]
M. Petri, S. Perez-Gutthann, D. Spence, M.C. Hochberg.
Risk factors for coronary artery disease in patients with systemic lupus erythematosus.
Am J Med, 93 (1992), pp. 513-519
[14]
J.M. Esdaile, M. Abrahamowicz, T. Grodzicky, Y. Li, C. Panaritis, R. du Berger, et al.
Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus.
[15]
J.A. Avina-Zubieta, F. To, K. Vostretsova, M. De Vera, E.C. Sayre, J.M. Esdaile.
Risk of myocardial infarction and stroke in newly diagnosed systemic lupus erythematosus: a general population-based study.
Arthritis Care Res (Hoboken), 69 (2017), pp. 849-856
[16]
Y. Ahmad, J. Shelmerdine, H. Bodill, M. Lunt, M.G. Pattrick, L.S. Teh, et al.
Subclinical atherosclerosis in systemic lupus erythematosus (SLE): the relative contribution of classic risk factors and the lupus phenotype.
Rheumatology (Oxford), 46 (2007), pp. 983-988
[17]
I.N. Bruce, R.J. Burns, D.D. Gladman, M.B. Urowitz.
Single photon emission computed tomography dual isotope myocardial perfusion imaging in women with systemic lupus erythematosus. I. Prevalence and distribution of abnormalities.
J Rheumatol, 27 (2000), pp. 2372-2377
[18]
K. Maksimowicz-McKinnon, L.S. Magder, M. Petri.
Predictors of carotid atherosclerosis in systemic lupus erythematosus.
J Rheumatol, 33 (2006), pp. 2458-2463
[19]
K. Manger, M. Kusus, C. Forster, D. Ropers, W.G. Daniel, J.R. Kalden, et al.
Factors associated with coronary artery calcification in young female patients with SLE.
Ann Rheum Dis, 62 (2003), pp. 846-850
[20]
A. Theodoridou, L. Bento, D.P. D’Cruz, M.A. Khamashta, G.R. Hughes.
Prevalence and associations of an abnormal ankle-brachial index in systemic lupus erythematosus: a pilot study.
Ann Rheum Dis, 62 (2003), pp. 1199-1203
[21]
A. Oeser, C.P. Chung, Y. Asanuma, I. Avalos, C.M. Stein.
Obesity is an independent contributor to functional capacity and inflammation in systemic lupus erythematosus.
Arthritis Rheum, 52 (2005), pp. 3651-3659
[22]
M.L. Ishimori, R. Martin, D.S. Berman, P. Goykhman, L.J. Shaw, C. Shufelt, et al.
Myocardial ischemia in the absence of obstructive coronary artery disease in systemic lupus erythematosus.
JACC Cardiovasc Imaging, 4 (2011), pp. 27-33
[23]
C.C. Mok, W.L. Poon, J.P. Lai, C.K. Wong, S.M. Chiu, C.K. Wong, et al.
Metabolic syndrome, endothelial injury, and subclinical atherosclerosis in patients with systemic lupus erythematosus.
Scand J Rheumatol, 39 (2010), pp. 42-49
[24]
J.M. Sabio, M. Zamora-Pasadas, J. Jimenez-Jaimez, F. Albadalejo, J. Vargas-Hitos, M.D. Rodriguez del Aguila, et al.
Metabolic syndrome in patients with systemic lupus erythematosus from Southern Spain.
Lupus, 17 (2008), pp. 849-859
[25]
I.N. Bruce, M.B. Urowitz, D.D. Gladman, D. Ibanez, G. Steiner.
Risk factors for coronary heart disease in women with systemic lupus erythematosus: the Toronto Risk Factor Study.
Arthritis Rheum, 48 (2003), pp. 3159-3167
[26]
P. Katz, S. Gregorich, J. Yazdany, L. Trupin, L. Julian, E. Yelin, et al.
Obesity and its measurement in a community-based sample of women with systemic lupus erythematosus.
Arthritis Care Res (Hoboken), 63 (2011), pp. 261-268
[27]
K. Sacre, B. Escoubet, M.C. Zennaro, M.P. Chauveheid, E. Gayat, T. Papo.
Overweight is a major contributor to atherosclerosis in systemic lupus erythematosus patients at apparent low risk for cardiovascular disease: a cross-sectional controlled study.
Medicine (Baltimore), 94 (2015), pp. e2177
[28]
H. Sanchez-Perez, B. Tejera-Segura, A. de Vera-Gonzalez, A. Gonzalez-Delgado, J.M. Olmos, J.L. Hernandez, et al.
Insulin resistance in systemic lupus erythematosus patients: contributing factors and relationship with subclinical atherosclerosis.
Clin Exp Rheumatol, 35 (2017), pp. 885-892
[29]
J.M. Sabio, J.A. Vargas-Hitos, N. Navarrete-Navarrete, J.D. Mediavilla, J. Jimenez-Jaimez, A. Diaz-Chamorro, et al.
Prevalence of and factors associated with hypertension in young and old women with systemic lupus erythematosus.
J Rheumatol, 38 (2011), pp. 1026-1032
[30]
K. Tselios, C. Koumaras, M.B. Urowitz, D.D. Gladman.
Do current arterial hypertension treatment guidelines apply to systemic lupus erythematosus patients? A critical appraisal.
Semin Arthritis Rheum, 43 (2014), pp. 521-525
[31]
M.A. Petri, A.N. Kiani, W. Post, L. Christopher-Stine, L.S. Magder.
Lupus Atherosclerosis Prevention Study (LAPS).
Ann Rheum Dis, 70 (2011), pp. 760-765
[32]
F. Selzer, K. Sutton-Tyrrell, S.G. Fitzgerald, J.E. Pratt, R.P. Tracy, L.H. Kuller, et al.
Comparison of risk factors for vascular disease in the carotid artery and aorta in women with systemic lupus erythematosus.
Arthritis Rheum, 50 (2004), pp. 151-159
[33]
M.G. Tektonidou, E. Kravvariti, G. Konstantonis, N. Tentolouris, P.P. Sfikakis, A. Protogerou.
Subclinical atherosclerosis in systemic lupus erythematosus: comparable risk with diabetes mellitus and rheumatoid arthritis.
Autoimmun Rev, 16 (2017), pp. 308-312
[34]
A.N. Kiani, W.S. Post, L.S. Magder, M. Petri.
Predictors of progression in atherosclerosis over 2 years in systemic lupus erythematosus.
Rheumatology (Oxford), 50 (2011), pp. 2071-2079
[35]
J.M. Sabio, J. Martinez-Bordonado, I. Sanchez-Berna, J.A. Vargas-Hitos, J.D. Mediavilla, N. Navarrete-Navarrete, et al.
Nighttime blood pressure patterns and subclinical atherosclerosis in women with systemic lupus erythematosus.
J Rheumatol, 42 (2015), pp. 2310-2317
[36]
W.P. Castelli, G.R. Cooper, J.T. Doyle, M. Garcia-Palmieri, T. Gordon, C. Hames, et al.
Distribution of triglyceride and total, LDL and HDL cholesterol in several populations: a cooperative lipoprotein phenotyping study.
J Chronic Dis, 30 (1977), pp. 147-169
[37]
T. Gordon, W.P. Castelli, M.C. Hjortland, W.B. Kannel, T.R. Dawber.
High density lipoprotein as a protective factor against coronary heart disease. The Framingham study.
Am J Med, 62 (1977), pp. 707-714
[38]
M.B. Urowitz, D. Gladman, D. Ibanez, P. Fortin, J. Sanchez-Guerrero, S. Bae, et al.
Accumulation of coronary artery disease risk factors over three years: data from an international inception cohort.
Arthritis Rheum, 59 (2008), pp. 176-180
[39]
E.F. Borba, E. Bonfa.
Dyslipoproteinemias in systemic lupus erythematosus: influence of disease, activity, and anticardiolipin antibodies.
Lupus, 6 (1997), pp. 533-539
[40]
S.O. Olusi, S. George.
Prevalence of LDL atherogenic phenotype in patients with systemic lupus erythematosus.
Vasc Health Risk Manag, 7 (2011), pp. 75-80
[41]
B.H. Hahn, J. Grossman, B.J. Ansell, B.J. Skaggs, M. McMahon.
Altered lipoprotein metabolism in chronic inflammatory states: proinflammatory high-density lipoprotein and accelerated atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis.
Arthritis Res Ther, 10 (2008), pp. 213
[42]
J. Delgado Alves, S. Kumar, D.A. Isenberg.
Cross-reactivity between anti-cardiolipin, anti-high-density lipoprotein and anti-apolipoprotein A-I IgG antibodies in patients with systemic lupus erythematosus and primary antiphospholipid syndrome.
Rheumatology (Oxford), 42 (2003), pp. 893-899
[43]
S.M. Toloza, A.G. Uribe, G. McGwin Jr., G.S. Alarcon, B.J. Fessler, H.M. Bastian, et al.
Systemic lupus erythematosus in a multiethnic US cohort (LUMINA). XXIII. Baseline predictors of vascular events.
Arthritis Rheum, 50 (2004), pp. 3947-3957
[44]
R. Clarke, L. Daly, K. Robinson, E. Naughten, S. Cahalane, B. Fowler, et al.
Hyperhomocysteinemia: an independent risk factor for vascular disease.
N Engl J Med, 324 (1991), pp. 1149-1155
[45]
M. den Heijer, H.J. Blom, W.B. Gerrits, F.R. Rosendaal, H.L. Haak, P.W. Wijermans, et al.
Is hyperhomocysteinaemia a risk factor for recurrent venous thrombosis?.
Lancet, 345 (1995), pp. 882-885
[46]
D. Bonciani, E. Antiga, V. Bonciolini, A. Verdelli, E. Del Bianco, W. Volpi, et al.
Homocysteine serum levels are increased and correlate with disease severity in patients with lupus erythematosus.
Clin Exp Rheumatol, 34 (2016), pp. 76-81
[47]
J.M. Sabio, J.A. Vargas-Hitos, J. Martinez-Bordonado, N. Navarrete-Navarrete, A. Diaz-Chamorro, C. Olvera-Porcel, et al.
Relationship between homocysteine levels and hypertension in systemic lupus erythematosus.
Arthritis Care Res (Hoboken), 66 (2014), pp. 1528-1535
[48]
J.M. Von Feldt, L.V. Scalzi, A.J. Cucchiara, S. Morthala, C. Kealey, S.D. Flagg, et al.
Homocysteine levels and disease duration independently correlate with coronary artery calcification in patients with systemic lupus erythematosus.
Arthritis Rheum, 54 (2006), pp. 2220-2227
[49]
J.T. Gustafsson, M. Herlitz Lindberg, I. Gunnarsson, S. Pettersson, K. Elvin, J. Ohrvik, et al.
Excess atherosclerosis in systemic lupus erythematosus – a matter of renal involvement: case–control study of 281 SLE patients and 281 individually matched population controls.
PLoS ONE, 12 (2017), pp. e0174572
[50]
S. Haque, C. Gordon, D. Isenberg, A. Rahman, P. Lanyon, A. Bell, et al.
Risk factors for clinical coronary heart disease in systemic lupus erythematosus: the lupus and atherosclerosis evaluation of risk (LASER) study.
J Rheumatol, 37 (2010), pp. 322-329
[51]
G.J. Pons-Estel, L.A. Gonzalez, J. Zhang, P.I. Burgos, J.D. Reveille, L.M. Vila, et al.
Predictors of cardiovascular damage in patients with systemic lupus erythematosus: data from LUMINA (LXVIII), a multiethnic US cohort.
Rheumatology (Oxford), 48 (2009), pp. 817-822
[52]
I.N. Bruce, A.G. O’Keeffe, V. Farewell, J.G. Hanly, S. Manzi, L. Su, et al.
Factors associated with damage accrual in patients with systemic lupus erythematosus: results from the Systemic Lupus International Collaborating Clinics (SLICC) Inception Cohort.
Ann Rheum Dis, 74 (2015), pp. 1706-1713
[53]
D.D. Gladman, M.B. Urowitz, P. Rahman, D. Ibanez, L.S. Tam.
Accrual of organ damage over time in patients with systemic lupus erythematosus.
J Rheumatol, 30 (2003), pp. 1955-1959
[54]
M.J. Roman, B.A. Shanker, A. Davis, M.D. Lockshin, L. Sammaritano, R. Simantov, et al.
Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus.
N Engl J Med, 349 (2003), pp. 2399-2406
[55]
M. Petri.
Hydroxychloroquine use in the Baltimore Lupus Cohort: effects on lipids, glucose and thrombosis.
Lupus, 5 (1996), pp. S16-S22
[56]
M. Petri, C. Lakatta, L. Magder, D. Goldman.
Effect of prednisone and hydroxychloroquine on coronary artery disease risk factors in systemic lupus erythematosus: a longitudinal data analysis.
Am J Med, 96 (1994), pp. 254-259
[57]
J.H. Rand, X.X. Wu, A.S. Quinn, A.W. Ashton, P.P. Chen, J.J. Hathcock, et al.
Hydroxychloroquine protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies: evidence for a novel effect for an old antimalarial drug.
Blood, 115 (2010), pp. 2292-2299
[58]
M.A. Garcia, G.S. Alarcon, G. Boggio, L. Hachuel, A.I. Marcos, J.C. Marcos, et al.
Primary cardiac disease in systemic lupus erythematosus patients: protective and risk factors – data from a multi-ethnic Latin American cohort.
Rheumatology (Oxford), 53 (2014), pp. 1431-1438
[59]
A.N. Kiani, L.S. Magder, M. Petri.
Mycophenolate mofetil (MMF) does not slow the progression of subclinical atherosclerosis in SLE over 2 years.
Rheumatol Int, 32 (2012), pp. 2701-2705
[60]
M. Dieude, J.L. Senecal, Y. Raymond.
Induction of endothelial cell apoptosis by heat-shock protein 60-reactive antibodies from anti-endothelial cell autoantibody-positive systemic lupus erythematosus patients.
Arthritis Rheum, 50 (2004), pp. 3221-3231
[61]
A. Duval, D. Helley, L. Capron, P. Youinou, Y. Renaudineau, S. Dubucquoi, et al.
Endothelial dysfunction in systemic lupus patients with low disease activity: evaluation by quantification and characterization of circulating endothelial microparticles, role of anti-endothelial cell antibodies.
Rheumatology (Oxford), 49 (2010), pp. 1049-1055
[62]
P.L. Meroni, E. Raschi, C. Testoni, M.O. Borghi.
Endothelial cell activation by antiphospholipid antibodies.
Clin Immunol, 112 (2004), pp. 169-174
[63]
M. Petri.
The lupus anticoagulant is a risk factor for myocardial infarction (but not atherosclerosis): Hopkins Lupus cohort.
Thromb Res, 114 (2004), pp. 593-595
[64]
O. Vaarala, G. Alfthan, M. Jauhiainen, M. Leirisalo-Repo, K. Aho, T. Palosuo.
Crossreaction between antibodies to oxidised low-density lipoprotein and to cardiolipin in systemic lupus erythematosus.
[65]
M. McMahon, B. Skaggs.
Pathogenesis and treatment of atherosclerosis in lupus.
Rheum Dis Clin North Am, 40 (2014), pp. 475-495
[66]
G.K. Hansson, A. Hermansson.
The immune system in atherosclerosis.
Nat Immunol, 12 (2011), pp. 204-212
[67]
B.J. Hunt.
The endothelium in atherogenesis.
Lupus, 9 (2000), pp. 189-193
[68]
K.J. Moore, I. Tabas.
Macrophages in the pathogenesis of atherosclerosis.
Cell, 145 (2011), pp. 341-355
[69]
S. Rajagopalan, E.C. Somers, R.D. Brook, C. Kehrer, D. Pfenninger, E. Lewis, et al.
Endothelial cell apoptosis in systemic lupus erythematosus: a common pathway for abnormal vascular function and thrombosis propensity.
Blood, 103 (2004), pp. 3677-3683
[70]
M.F. Denny, S. Yalavarthi, W. Zhao, S.G. Thacker, M. Anderson, A.R. Sandy, et al.
A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs.
J Immunol, 184 (2010), pp. 3284-3297
[71]
J.T. Gustafsson, J.F. Simard, I. Gunnarsson, K. Elvin, I.E. Lundberg, L.O. Hansson, et al.
Risk factors for cardiovascular mortality in patients with systemic lupus erythematosus, a prospective cohort study.
Arthritis Res Ther, 14 (2012), pp. R46
[72]
M. McMahon, J. Grossman, B. Skaggs, J. Fitzgerald, L. Sahakian, N. Ragavendra, et al.
Dysfunctional proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in women with systemic lupus erythematosus.
Arthritis Rheum, 60 (2009), pp. 2428-2437
[73]
M. Navab, S.T. Reddy, B.J. Van Lenten, A.M. Fogelman.
HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms.
Nat Rev Cardiol, 8 (2011), pp. 222-232
[74]
M. Navab, S.Y. Hama, C.J. Cooke, G.M. Anantharamaiah, M. Chaddha, L. Jin, et al.
Normal high density lipoprotein inhibits three steps in the formation of mildly oxidized low density lipoprotein: step 1.
J Lipid Res, 41 (2000), pp. 1481-1494
[75]
J. Delgado Alves, P.R. Ames, S. Donohue, L. Stanyer, J. Nourooz-Zadeh, C. Ravirajan, et al.
Antibodies to high-density lipoprotein and beta2-glycoprotein I are inversely correlated with paraoxonase activity in systemic lupus erythematosus and primary antiphospholipid syndrome.
Arthritis Rheum, 46 (2002), pp. 2686-2694
[76]
J.R. Batuca, P.R. Ames, D.A. Isenberg, J.D. Alves.
Antibodies toward high-density lipoprotein components inhibit paraoxonase activity in patients with systemic lupus erythematosus.
Ann N Y Acad Sci, 1108 (2007), pp. 137-146
[77]
G. Sweeney.
Cardiovascular effects of leptin.
Nat Rev Cardiol, 7 (2010), pp. 22-29
[78]
A. Garcia-Gonzalez, L. Gonzalez-Lopez, I.C. Valera-Gonzalez, E.G. Cardona-Munoz, M. Salazar-Paramo, M. Gonzalez-Ortiz, et al.
Serum leptin levels in women with systemic lupus erythematosus.
Rheumatol Int, 22 (2002), pp. 138-141
[79]
K.E. Sada, Y. Yamasaki, M. Maruyama, H. Sugiyama, M. Yamamura, Y. Maeshima, et al.
Altered levels of adipocytokines in association with insulin resistance in patients with systemic lupus erythematosus.
J Rheumatol, 33 (2006), pp. 1545-1552
[80]
H.R. Reynolds, J. Buyon, M. Kim, T.L. Rivera, P. Izmirly, P. Tunick, et al.
Association of plasma soluble E-selectin and adiponectin with carotid plaque in patients with systemic lupus erythematosus.
Atherosclerosis, 210 (2010), pp. 569-574
[81]
M.R. Malinow, F.J. Nieto, M. Szklo, L.E. Chambless, G. Bond.
Carotid artery intimal-medial wall thickening and plasma homocyst(e)ine in asymptomatic adults. The Atherosclerosis Risk in Communities Study.
Circulation, 87 (1993), pp. 1107-1113
[82]
R.T. Wall, J.M. Harlan, L.A. Harker, G.E. Striker.
Homocysteine-induced endothelial cell injury in vitro: a model for the study of vascular injury.
Thromb Res, 18 (1980), pp. 113-121
[83]
T.M. Refai, I.H. Al-Salem, D. Nkansa-Dwamena, M.H. Al-Salem.
Hyperhomocysteinaemia and risk of thrombosis in systemic lupus erythematosus patients.
Clin Rheumatol, 21 (2002), pp. 457-461
[84]
E. Svenungsson, K. Jensen-Urstad, M. Heimburger, A. Silveira, A. Hamsten, U. de Faire, et al.
Risk factors for cardiovascular disease in systemic lupus erythematosus.
Circulation, 104 (2001), pp. 1887-1893
[85]
A. Mak.
The impact of vitamin D on the immunopathophysiology, disease activity, and extra-musculoskeletal manifestations of systemic lupus erythematosus.
Int J Mol Sci, 19 (2018),
[86]
R.L. Ravenell, D.L. Kamen, J.D. Spence, B.W. Hollis, T.J. Fleury, M.G. Janech, et al.
Premature atherosclerosis is associated with hypovitaminosis D and angiotensin-converting enzyme inhibitor non-use in lupus patients.
Am J Med Sci, 344 (2012), pp. 268-273
[87]
S. Mellor-Pita, P. Tutor-Ureta, S. Rosado, K. Alkadi, F. Granado, C. Jimenez-Ortiz, et al.
Calcium and vitamin D supplement intake may increase arterial stiffness in systemic lupus erythematosus patients.
Clin Rheumatol, 38 (2019), pp. 1177-1186
[88]
P.W. Wilson, R.B. D’Agostino, D. Levy, A.M. Belanger, H. Silbershatz, W.B. Kannel.
Prediction of coronary heart disease using risk factor categories.
Circulation, 97 (1998), pp. 1837-1847
[89]
P.M. Ridker, N.P. Paynter, N. Rifai, J.M. Gaziano, N.R. Cook.
C-reactive protein and parental history improve global cardiovascular risk prediction: the Reynolds Risk Score for men.
Circulation, 118 (2008), pp. 2243-2251
[90]
I.U. Haq, P.R. Jackson, W.W. Yeo, L.E. Ramsay.
Sheffield risk and treatment table for cholesterol lowering for primary prevention of coronary heart disease.
Lancet, 346 (1995), pp. 1467-1471
[91]
J. Hippisley-Cox, C. Coupland, P. Brindle.
Development and validation of QRISK3 risk prediction algorithms to estimate future risk of cardiovascular disease: prospective cohort study.
BMJ, 357 (2017), pp. j2099
[92]
N. Edwards, A.W.W. Langford-Smith, B.J. Parker, I.N. Bruce, J.A. Reynolds, M.Y. Alexander, et al.
QRISK3 improves detection of cardiovascular disease risk in patients with systemic lupus erythematosus.
Lupus Sci Med, 5 (2018), pp. e000272
[93]
A. Fanouriakis, M. Kostopoulou, A. Alunno, M. Aringer, I. Bajema, J.N. Boletis, et al.
2019 update of the EULAR recommendations for the management of systemic lupus erythematosus.
Ann Rheum Dis, 78 (2019), pp. 736-745
[94]
M. Kawasaki, Y. Ito, H. Yokoyama, M. Arai, G. Takemura, A. Hara, et al.
Assessment of arterial medial characteristics in human carotid arteries using integrated backscatter ultrasound and its histological implications.
Atherosclerosis, 180 (2005), pp. 145-154
[95]
E. Smith, S. Croca, K.E. Waddington, R. Sofat, M. Griffin, A. Nicolaides, et al.
Cross-talk between iNKT cells and monocytes triggers an atheroprotective immune response in SLE patients with asymptomatic plaque.
Sci Immunol, 1 (2016),
[96]
S. Croca, A. Rahman.
Atherosclerosis in systemic lupus erythematosus.
Best Pract Res Clin Rheumatol, 31 (2017), pp. 364-372
[97]
C. Andrades, C. Fuego, S. Manrique-Arija, A. Fernandez-Nebro.
Management of cardiovascular risk in systemic lupus erythematosus: a systematic review.
Lupus, 26 (2017), pp. 1407-1419
[98]
W. Plazak, K. Gryga, H. Dziedzic, L. Tomkiewicz-Pajak, M. Konieczynska, P. Podolec, 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, 13 (2011), pp. R117
[99]
L. Arnaud, A. Mathian, H. Devilliers, A. Ruffatti, M. Tektonidou, R. Forastiero, et al.
Patient-level analysis of five international cohorts further confirms the efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies.
Autoimmun Rev, 14 (2015), pp. 192-200
[100]
M.J. Cuadrado, M.L. Bertolaccini, P.T. Seed, M.G. Tektonidou, A. Aguirre, L. Mico, et al.
Low-dose aspirin vs. low-dose aspirin plus low-intensity warfarin in thromboprophylaxis: a prospective, multicentre, randomized, open, controlled trial in patients positive for antiphospholipid antibodies (ALIWAPAS).
Rheumatology (Oxford), 53 (2014), pp. 275-284
[101]
T. Tarr, G. Lakos, H.P. Bhattoa, P. Soltesz, Y. Shoenfeld, G. Szegedi, et al.
Clinical thrombotic manifestations in SLE patients with and without antiphospholipid antibodies: a 5-year follow-up.
Clin Rev Allergy Immunol, 32 (2007), pp. 131-137
[102]
S. Fasano, D.P. Margiotta, L. Navarini, L. Pierro, I. Pantano, A. Riccardi, et al.
Primary prevention of cardiovascular disease in patients with systemic lupus erythematosus: case series and literature review.
Lupus, 26 (2017), pp. 1463-1472
[103]
R. Kaiser, C.M. Cleveland, L.A. Criswell.
Risk and protective factors for thrombosis in systemic lupus erythematosus: results from a large, multi-ethnic cohort.
Ann Rheum Dis, 68 (2009), pp. 238-241
[104]
S.K. Penn, A.H. Kao, L.L. Schott, J.R. Elliott, F.G. Toledo, L. Kuller, et al.
Hydroxychloroquine and glycemia in women with rheumatoid arthritis and systemic lupus erythematosus.
J Rheumatol, 37 (2010), pp. 1136-1142
[105]
G. Ruiz-Irastorza, M. Ramos-Casals, P. Brito-Zeron, M.A. Khamashta.
Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review.
Ann Rheum Dis, 69 (2010), pp. 20-28
Copyright © 2021. Asociación Colombiana de Reumatología
Opciones de artículo
Herramientas
Material suplementario
Quizás le interese:
10.1016/j.rcreu.2021.05.010
No mostrar más