Abstract
Purpose of Review.
Accelerated atherosclerosis is a significant co-morbidity and the leading cause of death for patients with systemic lupus erythematosus (SLE). It is now apparent that SLE-accelerated atherosclerosis is not driven solely by traditional cardiovascular risk factors, adding complexity to disease characterization and mechanistic understanding. In this review, we will summarize new insights into SLE-accelerated atherosclerosis evaluation, treatment, and mechanism.
Recent Findings.
Recent work highlights the need to incorporate inflammatory biomarkers into cardiovascular disease (CVD) risk assessments. This is especially true for SLE patients, where mechanisms of immune dysfunction likely drive CVD progression. There is new evidence that commonly prescribed SLE therapeutics hinder atherosclerosis development. This effect is achieved both by reducing SLE-associated inflammation and by directly improving measures of atherosclerosis, emphasizing the interconnected mechanisms of the two conditions.
Keywords: Atherosclerosis, cardiovascular disease, risk assessment, SLE-accelerated atherosclerosis, systemic lupus erythematosus
Summary.
SLE-accelerated atherosclerosis is most likely the consequence of chronic autoimmune inflammation. Therefore, diligent management of atherosclerosis requires assessment of SLE disease activity as well as traditional cardiovascular risk factors. This supports why many of the therapeutics classically used to control SLE also modulate atherosclerosis development. Greater understanding of the mechanisms underlying this condition will allow for the development of more targeted therapeutics and improved outcomes for SLE patients.
Introduction
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease affecting over 1.5 million Americans and at least 5 million individuals worldwide. Patients are predominantly female, and are two to three times more likely to be women of color. SLE is characterized by autoantibody producing B cells and dysfunctional CD4+ T cells, and can result in end organ damage to the kidneys, joints, and cardiovascular system [1]. Life expectancy for SLE patients has improved but remains lower than that of the general population [2], and the leading cause of death among SLE patients is cardiovascular disease (CVD) driven by accelerated atherosclerosis [3–7]. It is estimated that women with SLE between the ages of 35 and 44 have a 50-fold increased risk of myocardial infarction compared with age- and gender-matched controls [8]. Even if we assume that this estimate is inflated due to the fact that healthy, pre-menopausal women are typically protected from CVD development [9], post-menopausal women with SLE are still five times more likely to develop atherosclerosis than healthy, age-matched controls [10]. Statin therapy has been shown to provide less cardiovascular protection in SLE patients compared to the general population [11], suggesting the SLE-accelerated atherosclerosis is not entirely lipid dependent. In this review we highlight recent findings regarding mechanisms driving SLE-accelerated atherosclerosis as well as the best techniques to evaluate and treat this disease.
Disease Risk Assessment
Atherosclerosis is associated with a number of risk factors and the likelihood of a related adverse event (coronary heart disease, stroke, peripheral artery disease, or heart failure) occurring in the long term in the general population is often determined using composite measurements such as the Framingham Risk Score [12] or Systematic Coronary Risk Evaluation (SCORE) [13]. The Framingham Risk Score is a 10-year predictive measurement that uses sex, age, total and high-density lipoprotein (HDL) cholesterol levels, systolic blood pressure, hypertension, smoking, and diabetes status to determine an individual’s CVD risk [12]. SCORE utilizes many of the same metrics, but also incorporates geography as a variable to consider whether an individual’s country as a whole has rising or falling CVD rates [13]. In SLE patients these scores may inaccurately predict an individual’s true risk of CVD [14,15]. This is likely due to several factors; the lack of consideration of inflammatory variables that are increased in SLE, the heterogeneity of the disease, and our incomplete understanding of disease mechanism. Therefore, improved methods for predicting risk SLE are required to ensure appropriate management of CVD and other life-threatening co-morbidities.
Techniques for Improved Risk Assessment.
One strategy for adjusting the Framingham risk score in SLE has been to simply multiply the traditional score by a factor of two [16]. A similar technique was successfully applied to rheumatoid arthritis patients, who also experience increased rates of CVD related death [17,18]. Unfortunately, given the heterogeneous nature of SLE, a blanket correction factor may oversimplify the contribution of autoimmune factors to CVD risk. Therefore, it became necessary to develop an SLE-centered cardiovascular risk score which uses traditional cardiovascular risk factors like age, gender, systolic blood pressure, cholesterol, smoking, and diabetes, but also SLE-specific risk factors. Variables such as time since SLE diagnosis, corticosteroid and hydroxychloroquine use, C3 and C4 levels, anti-double stranded DNA (dsDNA) titer, proteinuria, disease activity score (SELENA-SLEDAI score), estimated glomerular filtration rate, and history of lupus anticoagulant and anticardiolipin were evaluated. In the subjects of this study, only disease activity score, C3 level, and lupus anticoagulant titer were predictive of cardiovascular outcomes [19**]. Using the SLE-specific risk score incorporating these select variables, Petri et al. determined that some SLE patients were in appropriately assessed by the traditional Framingham risk score while others, particularly those with higher SLEDAI scores, had their ten-year risk underestimated by as much as a factor of 10 [19**]. In agreement with these findings, a separate seven-year surveillance study found no difference in the progression of subclinical atherosclerosis between the control group and SLE patients with mild, well controlled disease [20], highlighting the need to use SLE-specific criteria and disease activity level in CVD risk assessment.
Inaccurate CVD risk assessment is especially prevalent in young SLE patients (≤45 years). These patients are not likely to experience adverse cardiovascular events within 10 years (the range in which classical scoring methods are designed to predict), but still have elevated risk compared to their healthy, age-matched counterparts, including a 50-fold increase in the risk of myocardial infarction [8,21]. Clinical studies have shown that coronary artery calcification (CAC), a known predictor of CVD mortality [22], is increased in SLE patients anywhere from 2.8 to 4 times compared to controls and may be an accurate predictor of outcomes [23,24]. These studies were performed using older, mostly white cohorts (72% and 65%, respectively), perhaps not accurately representing the predominately non-white SLE patient population. Gartshteyn et al. studied SLE patients aged 18–65, who were primarily African American and Hispanic, and had no clinical CVD. They found that CAC severity increased with age in patients, and that CAC was significantly more prevalent in young SLE patients compared to age-matched controls, 32% vs 9.6%, respectively [25]. In a separate study, a related, though independent condition, aorta calcification (AC), was found to occur earlier, at a higher incidence, and in a wider range of ages than CAC in SLE patients highlighting its importance in predicting atherosclerosis development [26].
Collectively, studies indicate risk assessment for SLE-accelerated atherosclerosis needs to be distinct from that of standard atherosclerosis. By integrating more inflammatory variables into predictive equations and/or adopting regular monitoring of CAC and AC, SLE patients will experience improved care with more timely therapeutic interventions.
Biomarkers.
In addition to improved techniques to assess atherosclerosis development in SLE patients, there has also been progress in terms of new biomarkers to identify potentially high CVD risk patients. Biomarkers associated with SLE-accelerated atherosclerosis include proinflammatory HDL (piHDL), increased circulating leptin, elevated homocysteine, and the presence of soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) [27].
Soluble CD163.
A biomarker that was previously associated with increased glomerular inflammation in SLE patients is the soluble form of the scavenger receptor CD163 (sCD163). sCD163 was found at high levels in both serum and urine of SLE patients with more CD163+ macrophage glomerular infiltrate and severe inflammation (r = 0.635) as part of lupus nephritis [28]. CD163 expression on macrophages has typically been thought of as an indication of alternative activation and an anti-inflammatory phenotype [29]. However, recent findings demonstrated CD163+ macrophages actually promote atherosclerotic plaque progression in both humans and mice via a CD163/HIF1α/VEGF-A pathway [30]. The involvement of CD163+ macrophages in both SLE and atherosclerosis suggest expression of this scavenger receptor may be an underlying mechanism of SLE-accelerated atherosclerosis and that its soluble form could be a biomarker of the disease. This was tested in one study of 63 SLE patients whose carotid atherosclerotic plaque was measured by ultrasound and serum sCD163 levels monitored both at baseline of the study and during follow-up. Results indicated that serum sCD163 is elevated in SLE patients compared to controls, and is further increased in patients with carotid plaque. Additionally, higher levels of sCD163 correlated with the development of new carotid plaque over time and positively associated with subclinical atherosclerosis in SLE independent of classic cardiovascular risk factors [31*]. These results indicate sCD163 is a promising biomarker of SLE-accelerated atherosclerosis.
FcγRIIA Polymorphism.
Fcγ receptors (FcγRs) are expressed on antigen presenting cells like macrophages, dendritic cells (DCs), and B cells, and bind to the Fc domain of IgG antibodies. Depending on whether the FcγR is activating or inhibitory, it will promote pro- or anti-inflammatory responses [32]. In mouse models, activating FcγRs are proatherogenic [33]. Functional polymorphism in the activating Fcγ receptor IIA (FcγRIIA) have previously been associated with SLE and lupus nephritis in humans [34,35]. In heparin-induced thrombocytopenia, a functional polymorphism (single amino acid substitution H131R) in FcγRIIA caused a hyperactive platelet phenotype characterized by increased expression of P-selectin and CD40 ligand, greater binding to annexin V, and increased formation of platelet-leukocyte complexes [36–40]. A similar phenotype was described in SLE where hyperactive platelets promote vascular pathogenesis by activating endothelial cells [41]. Based on these works, Clancy et al. investigated whether the H131R FcγRIIA polymorphism impacted cardiovascular outcomes in SLE patients. The H131R allelic variant was found in a similar proportion (~40%) of both the SLE patient and healthy control populations. When SLE patients were assessed by carotid ultrasound, 58% of SLE patients with the allelic variant had carotid plaque compared to only 25% of SLE patients with the ancestral variant. Yet, the FcγRIIA allelic variant did not associate with carotid plaque in heathy control subjects [42*]. This suggests the H131R functional polymorphism confers CVD risk specifically in the context of SLE and could potentially acts as a genetic CVD biomarker in patients.
Appropriate CVD risk assessment for the SLE patient population has long been needed. The techniques and biomarkers outlined above have the potential to improve CVD risk detection in patients, hopefully resulting in prompt therapeutic intervention and prevention of devastating cardiovascular outcomes.
Mechanism and Treatment
Treatments for SLE are limited in comparison to other chronic illnesses and therapeutic advancement has been relatively slow, in part because of lack of understanding of disease mechanisms. Unfortunately, the impact of some of the most common SLE therapeutics specifically on SLE-accelerated atherosclerosis has been understudied. The following highlights recent work that has contributed to determining the impact of SLE treatments on atherosclerosis and the mechanism behind SLE-accelerated atherosclerosis. Table 1 summarizes the treatments discussed and their effects.
Table 1.
Review of SLE-Accelerated Atherosclerosis Treatments
Treatment | Effect in SLE-Accelerated Atherosclerosis | References |
---|---|---|
BAFF Inhibition | Improves SLE responder index in patients with ongoing B cell dysfunction Reduces circulating anti-dsDNA antibodies in mice Inhibits antibody and complement deposition in the kidney in mice Atheroprotective in animals with low (<5mmol/L) cholesterol and atherogenic in animals with high cholesterol |
[52,54**] |
Hydroxychloroquine | Reduces interferon-α production Lessens aortic stiffness in SLE patients Corrects lipoprotein profile (increases HDL and lowers cholesterol, LDL, VLDL, triglycerides, and cylomicrons) in SLE patients Improves glycemic control in non-diabetic women with SLE Reduces the risk of all thrombovascular events in SLE patients SLE patients with carotid plaque are less likely to be using HCQ than those with no plaque (63% vs. 82.3%) Long term HCQ treatment in ApoE−/− mice with pristane-induced SLE reduces atherosclerotic lesion size, anti-dsDNA antibodies, total leukocyte numbers, macrophages, and DCs, and increases lymohocytes HDL from SLE patients is more functional following 12 weeks of treatment with HCQ. |
[58–67*,70*] |
Low-dose IL-2 | IL-2 treatment reduces renal inflammation and activation of kidney-infiltrating CD4+ T cells in a mouse model of lupus nephritis Targeted IL-2 treatment in ApoE−/− mice reduces the size of pre-established atherosclerotic lesions Currently under study in clinical trials to determine its effectiveness in SLE and atherosclerosis |
[96*,97–99] |
Mycophenolate | Treatment with MMF for 12 weeks improves HDL function in SLE patients Treatment with MMF for 12 weeks improves CVD biomarkers like sTWEAK in SLE patients Reduces atherosclerosis in Ldlr−/− B6.Sle1.2.3 bone marrow chimeras and limits recruitment of CD4+ T cells to atherosclerotic lesions |
[70*,71] |
VLDL = very low-density lipoprotein
In SLE, B cells are hyperactive leading to the production of autoantibodies, inappropriate T cell activation and DC recruitment, and inhibition of regulatory T cells (Tregs) [43]. In the context of atherosclerosis, B cells were initially thought to be protective [44,45], but recent work has demonstrated B2 B cells and production of IgG are proatherogenic [46,47]. The cytokine B-cell activating factor (BAFF) is required for the maturation and survival of B2 B cells and plays a major role in SLE [48–51]. Benlysta®, a B-cell-depleting monoclonal antibody that targets BAFF, was the first new SLE therapy in 50 years [52]. Therefore, inhibition of BAFF signaling seemed an attractive target for treating SLE-accelerated atherosclerosis. To test this, Saidoune et al. crossed atherosclerosis-susceptible ApoE−/− to Qa-1 knock-in mice (D227K) which have an amino acid mutation that hinders CD8+ Tregs and develop an SLE-like phenotype [53]. Interestingly, when ApoE−/− D227K mice were treated with a BAFF neutralizing antibody, SLE disease activity was impaired but atherosclerosis severity was only improved in animals with low cholesterol levels (<5mmol/L). This unexpected finding was the result of BAFF signaling through transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) on macrophages. Inhibiting BAFF-TACI signaling on macrophages promoted foam cell formation in high lipid environments and was largely atherogenic, overriding the atheroprotective effect of disrupting BAFF-BAFF receptor (BAFFR) on B cells in SLE [54**]. A separate study demonstrated BAFFR is also expressed on circulating endothelial progenitor cells (EPCs) [55*], a population of cells that are meant to maintain vascular homeostasis but that is reduced in SLE patients [56,57]. BAFF signaling on EPCs induced apoptosis suggesting inhibition of BAFF signaling could be beneficial in SLE-accelerated atherosclerosis [55*]. Overall, BAFF signaling seems to be a critical mechanism of SLE-accelerated atherosclerosis, but requires further study due to its complex interaction with multiple cell types.
Hydroxychloroquine (HCQ) is one of the most widely used medications for autoimmune diseases and is regularly prescribed for SLE [58]. It is known to elicit atheroprotective effects like improving lipid profiles [59–61] and glycemic control [62], lessening aortic stiffness [63], and reducing thrombotic events [64,65]. There is a documented negative correlation between HCQ treatment and atherosclerosis in SLE [66], but the mechanism has not been elucidated. By studying pristane-induced SLE in atherosclerosis-susceptible ApoE−/− mice, Liu et al. showed long term HCQ treatment reduced atherosclerosis largely through reversal of SLE-associated autoinflammation. Specifically, HCQ treatment lowered anti-dsDNA antibodies and reduced total leukocytes, macrophages, and DCs [67*]. Pristane-induced SLE is known to reduce lymphocyte numbers in the spleen by triggering their apoptosis [68]. This phenotype was also reversed by HCQ treatment in both the aorta and the spleen which authors hypothesize increased the numbers of atheroprotective Tregs [67*]. This mechanistic study provides some insight to how HCQ may also be beneficial in treating SLE-accelerated atherosclerosis.
Although typically considered to be atheroprotective due to its anti-inflammatory properties, HDL in SLE patients can become piHDL. SLE patients with carotid artery plaque are more likely to have piHDL than SLE patients without plaque [69], making it an interesting biomarker for SLE-accelerated atherosclerosis [27]. Little is known about the effects of regularly prescribed SLE disease-modifying therapies like mycophenolate (MMF), azathioprine (AZA), and HCQ on piHDL. In a recent observational study, SLE patients starting a new disease-modifying therapy (MMF, AZA, or HCQ) had plasma collected and disease activity measured at baseline, six weeks post initiation of therapy, and 12 weeks post initiation of therapy [70*]. MMF and HCQ significantly improved HDL function over the course of the 12-week treatment, though not to normal levels, while AZA had no effect on HDL function. MMF also significantly improved other biomarkers associated with plaque and intima-media thickness progression in SLE, suggesting MMF could have potential to treat both SLE associated glomerulonephritis and accelerated atherosclerosis [70*].
Findings from our own work demonstrate MMF treatment reduces atherosclerosis in Ldlr−/− B6.Sle1.2.3 bone marrow chimeric mice (which spontaneously develop SLE and are atherosclerosis prone) and inhibits CD4+ T cell activation and recruitment to atherosclerotic plaques [71]. In general, T cells are known to significantly contribute to the development of atherosclerosis [72,73], with different subsets playing unique roles. For instance, Th1 cells are widely accepted to be proatherogenic [74–76] while functional Tregs are atheroprotective [77,78]. T cells are also critical in SLE where they are long-lived, hyperactive, and produce proinflammatory cytokines like IFN-γ and IL-17 [79–82]. Much of the work from our group has shown the importance of T cells in mouse models of SLE-accelerated atherosclerosis [83–85], and our findings are in agreement with results from human studies [86,87].
Previous results from Laurence Morel’s group identified the lupus susceptibility gene, Pbx1d, as overexpressed in CD4+ T cells from SLE patients and using mouse models of SLE described its role in expanding follicular helper T cells (Tfh) and impairing Treg homeostasis [88,89]. Given the importance of CD4+ T cells in atherosclerosis, recent work explored the effect of Pbx1d transgenic CD4+ T cells in atherosclerosis in a Ldlr−/− bone marrow chimeras. Pbx1d overexpression in CD4+ T cells resulted in thicker arterial walls and larger necrotic cores within lesions, which is thought to be an indication of plaque instability and severity. Recipients of Pbx1d transgenic bone marrow had expanded Tfh cells and impaired Tregs, that was increased with western diet feeding [90**], indicating a lupus susceptibility allele can drive atherosclerosis, while at the same time dyslipidemia can enhance autoimmune phenotypes.
Impaired IL-2 signaling in T cells due to decreased IL-2 production in SLE and its contribution to disease pathogenesis is well known [91–94]. Recent findings identify a regulatory subunit (PPP2R2D) of protein phosphatase 2A (an enzyme which is increased in T cells from SLE patients and leads to decreased IL-2 production) that limits the accessibility of the IL-2 gene and other transcription factors important to IL-2 expression via chromatin remodeling. PPP2R2D is increased in T cells from SLE patients and PPP2R2D deficiency in T cells prevents autoimmunity in an imiquimod-induced lupus-like mouse model [95*]. A separate group demonstrated that treating (NZB x NZW) F1 SLE mice with IL-2 reduced renal inflammation and lessened the activity of kidney-infiltrating CD4+ T cells [96*]. Interestingly, IL-2 delivered specifically to atherosclerotic lesions through fusion to fibronectin targeting antibody reduced plaque size by activating and expanding Tregs [97]. This suggests IL-2 supplementation could be beneficial in both SLE and atherosclerosis, a hypothesis which is currently being tested in low-dose IL-2 treatment clinical trials [98,99].
In order to determine how the risk of atherosclerotic vascular events (AVE) in SLE patients has changed over time in response to new therapeutic strategies, Urowitz et al. compared two cohorts of newly identified (enrolled in the study within 12 months of diagnosis) SLE patients. Cohort 1 included patients who entered the University of Toronto Lupus Clinic between 1975 and 1987 and were then followed through 1992, while Cohort 2 included patients entering between 1999 and 2011 and were followed through 2016. Over the course of study SLE disease activity, treatment regimens, blood pressure, cholesterol, blood sugar, smoking status, and AVE were monitored. Patients in Cohort 2 had significantly less AVE than Cohort 1 over the 17-year study period. Additionally, patients in Cohort 2 had lower SLE disease burdens, smoked less and had longer periods of time with normal blood pressure, cholesterol, and glucose than patients in Cohort 1, demonstrating interventions for both SLE and CVD have improved over time [100*].
Understanding how current treatment regimens effect SLE-associated atherosclerosis is an important goal which will provide near immediate benefit to individuals living with the disease. Of equal importance, is the more long-term goal of elucidating the mechanisms which drive SLE-accelerated atherosclerosis. Though findings made in this area may not immediately translate to the clinic, they will eventually lead to more targeted therapeutics for patients. These works suggest a highly interconnected, positive feedback loop between progression of SLE and atherosclerosis, and may provide further insight as to why therapeutics designed to treat one impact the other.
Conclusion.
SLE-accelerated atherosclerosis is a complex disease in which immunological and cardiovascular mechanisms are intertwined to drive pathogenesis. While recent work has begun to elucidate this interconnected relationship, further study is needed to gain a complete mechanistic understanding of the disease and to identify potential therapeutic targets. In the meantime, the field has grown in its understanding of how to best evaluate current SLE patients for their cardiovascular risk, integrating traditional cardiovascular risk factors with information about inflammatory disease activity. This improvement in disease risk assessment will lead to better monitoring of high-risk patients as well as improved treatment regimens.
Key Points.
Accelerated atherosclerosis is the leading cause of death among SLE patients.
CVD risk assessment in SLE patients must integrate inflammatory as well as traditional cardiovascular risk factors in order to get an accurate assessment.
Many of the immune mechanisms that drive SLE also promote atherosclerosis.
Many of the existing SLE treatments hold promise for improving SLE-accelerated atherosclerosis.
Acknowledgments
Financial Support and Sponsorship
Support provided by the Veterans Association (VA Merit Award I01BX002968 to ASM) and the National Institutes of Health (NIAID R01AI153167 to ASM and NHLBI 1F31HL154569–01 to BDA).
Footnotes
Conflicts of Interest
There are no conflicts of interest.
References and Recommended Reading
- 1.Ward MM, Pyun E, Studenski S. Causes of death in systemic lupus erythematosus. Long-term followup of an inception cohort. Arthritis Rheum. 1995. October;38(10):1492–9. [DOI] [PubMed] [Google Scholar]
- 2.Urowitz MB, Gladman DD, Tom BDM, et al. Changing patterns in mortality and disease outcomes for patients with systemic lupus erythematosus. J Rheumatol. 2008;35(11):2152–8. [DOI] [PubMed] [Google Scholar]
- 3.Thomas G, Mancini J, Jourde-Chiche N, Sarlon G, et al. Mortality associated with systemic lupus erythematosus in France assessed by multiple-cause-of-death analysis. Arthritis Rheumatol. 2014;66(9):2503–11. [DOI] [PubMed] [Google Scholar]
- 4.Bartels CM, Buhr KA, Goldberg JW, et al. Mortality and cardiovascular burden of systemic lupus erythematosus in a US population-based cohort. J Rheumatol. 2014;41(4):680–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Urowitz MB, Gladman D, Ibañez D, et al. Clinical manifestations and coronary artery disease risk factors at diagnosis of systemic lupus erythematosus: Data from an international inception cohort. Lupus. 2007;16(9):731–5. [DOI] [PubMed] [Google Scholar]
- 6.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 Epidemiol. 1997;145(5):408–15. [DOI] [PubMed] [Google Scholar]
- 7.Giannelou M, Mavragani CP. Cardiovascular disease in systemic lupus erythematosus: A comprehensive update. J Autoimmun. 2017;82:1–12. [DOI] [PubMed] [Google Scholar]
- 8.Manzi S, Selzer F, Sutton-Tyrrell K, et al. Prevalence and risk factors of carotid plaque in women with systemic lupus erythematosus. Arthritis Rheum. 1999. January;42(1):51–60. [DOI] [PubMed] [Google Scholar]
- 9.Mendelsohn ME, Karas RH. Molecular and cellular basis of cardiovascular gender differences. Science. 2005;308(5728):1583–7. [DOI] [PubMed] [Google Scholar]
- 10.Urowitz MB, Bookman AAM, Koehler BE, et al. The bimodal mortality pattern of systemic lupus erythematosus. Am J Med. 1976;60(2):221–5. [DOI] [PubMed] [Google Scholar]
- 11.Petri MA, Kiani AN, Post W, et al. Lupus Atherosclerosis Prevention Study (LAPS). Ann Rheum Dis. 2011. May;70(5):760–5. [DOI] [PubMed] [Google Scholar]
- 12.D’Agostino RB, Vasan RS, Pencina MJ, et al. General cardiovascular risk profile for use in primary care: The Framingham heart study. Circulation. 2008;117(6):743–53. [DOI] [PubMed] [Google Scholar]
- 13.Conroy RM, Pyörälä K, Fitzgerald AP, et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: The SCORE project. Eur Heart J. 2003;24(11):987–1003. [DOI] [PubMed] [Google Scholar]
- 14.Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. Arthritis Rheum. 2001;44(10):2331–7. [DOI] [PubMed] [Google Scholar]
- 15.Bruce IN, Urowitz MB, Gladman DD, et al. Risk factors for coronary heart disease in women with systemic lupus erythematosus: The Toronto risk factor study. Arthritis Rheum. 2003;48(11):3159–67. [DOI] [PubMed] [Google Scholar]
- 16.Urowitz MB, Ibañez D, Su J, Gladman DD. Modified framingham risk factor score for systemic lupus erythematosus. J Rheumatol. 2016;43(5):875–9. [DOI] [PubMed] [Google Scholar]
- 17.Agca R, Heslinga SC, Rollefstad S, et al. EULAR recommendations for cardiovascular disease risk management in patients with rheumatoid arthritis and other forms of inflammatory joint disorders: 2015/2016 update. Ann Rheum Dis. 2016;76(1):17–28. [DOI] [PubMed] [Google Scholar]
- 18.Wahlin B, Innala L, Magnusson S, et al. Performance of the expanded cardiovascular risk prediction score for rheumatoid arthritis is not superior to the ACC/AHA risk calculator. J Rheumatol. 2019;46(2):130–7. [DOI] [PubMed] [Google Scholar]
- 19.Petri MA, Barr E, Magder LS. Development of a systemic lupus erythematosus cardiovascular risk equation. Lupus Sci Med. 2019. October;6(1):e000346.** This study describes a SLE-specific cardiovascular risk equation that combines SLE inflammatory variables like disease activity score, C3 concentration, and lupus anticoagulant titer with traditional cardiovascular risk criteria. The development of this SLE-specific score corrects the underestimation of risk typically associated with the use of Framingham or SCORE predictions on SLE patients.
- 20.Ajeganova S, Gustafsson T, Lindberg L, et al. Similar progression of carotid intima–media thickness in 7-year surveillance of patients with mild SLE and controls, but this progression is still promoted by dyslipidaemia, lower HDL levels, hypertension, history of lupus nephritis and a higher prednisolone usage in patients. Lupus Sci Med. 2020. January 9;7(1):e000362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bonow RO. Should coronary calcium screening be used in cardiovascular prevention strategies? N Engl J Med. 2009;361(10):990–7. [DOI] [PubMed] [Google Scholar]
- 22.Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. N Engl J Med. 2008;358(13):1336–45. [DOI] [PubMed] [Google Scholar]
- 23.Asanuma Y, Oeser A, Shintani AK, et al. Premature coronary-artery atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003. December;349(25):2407–15. [DOI] [PubMed] [Google Scholar]
- 24.Kiani AN, Magder LS, Post WS, et al. Coronary calcification in SLE: Comparison with the Multi-Ethnic Study of Atherosclerosis. Rheumatol (United Kingdom). 2015;54(11):1976–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gartshteyn Y, Braverman G, Mahtani S, et al. Prevalence of coronary artery calcification in young patients with SLE of predominantly Hispanic and African–American descent. Lupus Sci Med. 2019. June 29;6(1):e000330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hu L, Chen Z, Jin Y, et al. Incidence and predictors of aorta calcification in patients with systemic lupus erythematosus. Lupus. 2019. March 10;28(3):275–82. [DOI] [PubMed] [Google Scholar]
- 27.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Endo N, Tsuboi N, Furuhashi K, et al. Urinary soluble CD163 level reflects glomerular inflammation in human lupus nephritis. Nephrol Dial Transplant. 2016;31(12):2023–33. [DOI] [PubMed] [Google Scholar]
- 29.Abraham NG, Drummond G. CD163-mediated hemoglobin-heme uptake activates macrophage HO-1, providing an antiinflammatory function. Circ Res. 2006;99(9):911–4. [DOI] [PubMed] [Google Scholar]
- 30.Guo L, Akahori H, Harari E, et al. CD163+ macrophages promote angiogenesis and vascular permeability accompanied by inflammation in atherosclerosis. J Clin Invest. 2018;128(3):1106–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.David C, Divard GD, Abbas R, et al. Soluble CD163 is a biomarker for accelerated atherosclerosis in systemic lupus erythematosus patients at apparent low risk for cardiovascular disease. Scand J Rheumatol. 2020. January 2;49(1):33–7.*This work identifies a positive correlation between sCD163 and atherosclerosis progression in SLE patients. As sCD163 is already known as a biomarker of SLE severity, this work highlights it is truly a marker of SLE-accelerated atherosclerosis.
- 32.Nimmerjahn F, Ravetch JV. Fc-receptors as regulators of immunity. Adv Immunol. 2007;96(07):179–204. [DOI] [PubMed] [Google Scholar]
- 33.Hernández-Vargas P, Ortiz-Muñoz G, López-Franco O, et al. Fcγ receptor deficiency confers protection against atherosclerosis in apolipoprotein E knockout mice. Circ Res. 2006;99(11):1188–96. [DOI] [PubMed] [Google Scholar]
- 34.Manger K, Repp R, Spriewald BM, et al. Fcγ receptor IIa polymorphism in Caucasian patients with systemic lupus erythematosus: Association with clinical symptoms. Arthritis Rheum. 1998;41(7):1181–9. [DOI] [PubMed] [Google Scholar]
- 35.Brown EE, Edberg JC, Kimberly RP. Fc receptor genes and the systemic lupus erythematosus diathesis. Autoimmunity. 2007;40(8):567–81. [DOI] [PubMed] [Google Scholar]
- 36.Arepally G, McKenzie SE, Jiang X-M, et al. FcγRIIA H/R131 polymorphism, subclass-specific IgG anti-heparin/platelet factor 4 antibodies and clinical course in patients with heparin-induced thrombocytopenia and thrombosis. Blood. 1997. January 15;89(2):370–5. [PubMed] [Google Scholar]
- 37.Duffau P, Seneschal J, Nicco C, et al. Platelet CD154 potentiates interferon-α secretion by plasmacytoid dendritic cells in systemic lupus erythematosus. Sci Transl Med. 2010;2(47). [DOI] [PubMed] [Google Scholar]
- 38.Nagahama M, Nomura S, Ozaki Y, et al. Platelet activation markers and soluble adhesion molecules in patients with systemic lupus erythematosus. Autoimmunity. 2001;33(2):85–94. [DOI] [PubMed] [Google Scholar]
- 39.Lood C, Amisten S, Gullstrand B, et al. Platelet transcriptional profile and protein expression in patients with systemic lupus erythematosus: Up-regulation of the type I interferon system is strongly associated with vascular disease. Blood. 2010;116(11):1951–7. [DOI] [PubMed] [Google Scholar]
- 40.Joseph JE, Harrison P, Mackie IJ, et al. Increased circulating platelet-leucocyte complexes and platelet activation in patients with antiphospholipid syndrome, systemic lupus erythematosus and rheumatoid arthritis. Br J Haematol. 2001;115(2):451–9. [DOI] [PubMed] [Google Scholar]
- 41.Nhek S, Clancy R, Lee KA, et al. Activated platelets induce endothelial cell activation via an interleukin-1β pathway in systemic lupus erythematosus. Arterioscler Thromb Vasc Biol. 2017;37(4):707–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Clancy R, El Bannoudi H, Rasmussen SE, et al. Human low-affinity IgG receptor FcγRIIA polymorphism H131R associates with subclinical atherosclerosis and increased platelet activity in systemic lupus erythematosus. J Thromb Haemost. 2019. March;17(3):532–7.*This work highlights how genetic polymorphisms can confer risk in the context of SLE while not impacting healthy individuals. SLE patients with the H131R functional polymorphism in FcγRIIA are more likely to have carotid plaque while there is no association with allele variant in healthy controls.
- 43.Sanz I, Lee FE-H. B cells as therapeutic targets in SLE. Nat Rev Rheumatol. 2010. June;6(6):326–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Major AS, Fazio S, Linton MF. B-lymphocyte deficiency increases atherosclerosis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol. 2002;22(11):1892–8. [DOI] [PubMed] [Google Scholar]
- 45.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kyaw T, Tay C, Khan A, et al. Conventional B2 B cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. J Immunol. 2010;185(7):4410–9. [DOI] [PubMed] [Google Scholar]
- 47.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):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Vincent FB, Morand EF, Schneider P, MacKay F. The BAFF/APRIL system in SLE pathogenesis. Nat Rev Rheumatol. 2014;10(6):365–73. [DOI] [PubMed] [Google Scholar]
- 49.Mackay F, Woodcock SA, Lawton P, et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999;190(11):1697–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang J, Roschke V, Baker KP, et al. Cutting edge: A role for B lymphocyte stimulator in systemic lupus erythematosus. J Immunol. 2001;166(1):6–10. [DOI] [PubMed] [Google Scholar]
- 51.Stohl W, Metyas S, Tan SM, et al. B lymphocyte stimulator overexpression in patients with systemic lupus erythematosus: Longitudinal Observations. Arthritis Rheum. 2003;48(12):3475–86. [DOI] [PubMed] [Google Scholar]
- 52.Stohl W, Hilbert DM. The discovery and development of belimumab: the anti-BLyS–lupus connection. Nat Biotechnol. 2012. January;30(1):69–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kim HJ, Verbinnen B, Tang X, et al. Inhibition of follicular T-helper cells by CD8+ regulatory T cells is essential for self tolerance. Nature. 2010;467(7313):328–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Saidoune F, Even G, Lamri Y, et al. Effects of BAFF neutralization on atherosclerosis associated with systemic lupus erythematosus. Arthritis Rheumatol. 2020. August 11; Epub ahead of print.**Results from this study are particularly surprising given the integral role BAFF signaling plays in the activation of proatherogenic B2 B cells. Findings indicate in low-lipid conditions, BAFF inhibition is predictably atheroprotective, but in high lipid environments it is atherogenic due to a counter function in macrophages.
- 55.Spinelli FR, Barbati C, Cecarelli F, et al. B lymphocyte stimulator modulates number and function of endothelial progenitor cells in systemic lupus erythematosus. Arthritis Res Ther. 2019. December 21;21(1):245.*This includes the first description of BAFF receptor on endothelial progenitor cells which causes an increase apoptosis in response to BAFF signaling. These findings suggest the anti-BAFF SLE biologic, Benlysta ©, might also improve SLE-accelerated atherosclerosis by restoring a cell population important for vascular homeostasis.
- 56.Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275(5302):964–7. [DOI] [PubMed] [Google Scholar]
- 57.Moonen JRAJ, de Leeuw K, van Seijen XJGY, et al. Reduced number and impaired function of circulating progenitor cells in patients with systemic lupus erythematosus. Arthritis Res Ther. 2007;9(4):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Costedoat-Chalumeau N, Dunogué B, Morel N, et al. Hydroxychloroquine: A multifaceted treatment in lupus. Press Medicale. 2014;43(6P2):e167–80. [DOI] [PubMed] [Google Scholar]
- 59.Durcan L, Winegar DA, Connelly MA, et al. Longitudinal evaluation of lipoprotein variables in systemic lupus erythematosus reveals adverse changes with disease activity and prednisone and more favorable profiles with hydroxychloroquine therapy. J Rheumatol. 2016;43(4):745–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wallace DJ, Metzger AL, Stecher VJ, et al. Cholesterol-lowering effect of hydroxychloroquine in patients with rheumatic disease: Reversal of deleterious effects of steroids on lipids. Am J Med. 1990;89(3):322–6. [DOI] [PubMed] [Google Scholar]
- 61.Cairoli E, Rebella M, Danese N, et al. Hydroxychloroquine reduces low-density lipoprotein cholesterol levels in systemic lupus erythematosus: a longitudinal evaluation of the lipid-lowering effect. Lupus. 2012;21(11):1178–82. [DOI] [PubMed] [Google Scholar]
- 62.Penn SK, Kao AH, Schott LL, et al. Hydroxychloroquine and glycemia in women with rheumatoid arthritis and systemic lupus erythematosus. J Rheumatol. 2010;37(6):1136–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Selzer F, Sutton-Tyrrell K, Fitzgerald S, et al. Vascular stiffness in women with systemic lupus erythematosus. Hypertension. 2001;37(4):1075–82. [DOI] [PubMed] [Google Scholar]
- 64.Jung H, Bobba R, Su J, et al. The protective effect of antimalarial drugs on thrombovascular events in systemic lupus erythematosus. Arthritis Rheum. 2010;62(3):863–8. [DOI] [PubMed] [Google Scholar]
- 65.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Roman MJ, Shanker B-A, Davis A, et al. Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003. December;349(25):2399–406. [DOI] [PubMed] [Google Scholar]
- 67.Liu T, Shi N, Zhang S, et al. Systemic lupus erythematosus aggravates atherosclerosis by promoting IgG deposition and inflammatory cell imbalance. Lupus. 2020. March 19;29(3):273–82.*This work provides some of the first in vivo mechanistic evidence to explain the well established atheroprotective effect of HCQ treatment in SLE patients.
- 68.Calvani N, Caricchio R, Tucci M, et al. Induction of apoptosis by the hydrocarbon oil pristane: Implications for pristane-induced lupus. J Immunol. 2005;175(7):4777–82. [DOI] [PubMed] [Google Scholar]
- 69.McMahon M, Grossman J, Skaggs B, et al. Dysfunctional proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheum. 2009;60(8):2428–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.McMahon M, Skaggs B, Grossman J, et al. Comparison of PREDICTS atherosclerosis biomarker changes after initiation of new treatments in patients with SLE. Lupus Sci Med. 2019. June 27;6(1):e000321.*This study is the first to demonstrate that HCQ treatment in SLE patients improves HDL function, a finding that adds to a list of other mechanisms by which HCQ treatment is atheroprotective in SLE.
- 71.van Leuven SI, Mendez-Fernandez YV, Wilhelm AJ, et al. Mycophenolate mofetil but not atorvastatin attenuates atherosclerosis in lupus-prone LDLr−/− mice. Ann Rheum Dis. 2012. March;71(3):408–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhou X, Nicoletti A, Elhage R, Hansson GK. Transfer of CD4 + T Cells Aggravates Atherosclerosis in Immunodeficient Apolipoprotein E Knockout Mice. Circulation. 2000. December 12;102(24):2919–22. [DOI] [PubMed] [Google Scholar]
- 73.Zhou X, Robertson AKL, Rudling M, et al. Lesion development and response to immunization reveal a complex role for CD4 in atherosclerosis. Circ Res. 2005;96(4):427–34. [DOI] [PubMed] [Google Scholar]
- 74.Gupta S, Pablo AM, Jiang XC, et al. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997. June 1;99(11):2752–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Whitman SC, Ravisankar P, Elam H, Daugherty A. Exogenous interferon-γ enhances atherosclerosis in apolipoprotein E−/− mice. Am J Pathol. 2000. December;157(6):1819–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Buono C, Binder CJ, Stavrakis G, et al. T-bet deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proc Natl Acad Sci. 2005. February 1;102(5):1596–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Klingenberg R, Gerdes N, Badeau RM, et al. Depletion of FOXP3+ regulatory T cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest. 2013. March 1;123(3):1323–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.de Boer OJ, van der Meer JJ, Teeling P, et al. Low numbers of FOXP3 positive regulatory T cells are present in all developmental stages of human atherosclerotic lesions. PLoS One. 2007. January;2(8):e779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Crispín JC, Apostolidis SA, Finnell MI, Tsokos GC. Induction of PP2A Bβ, a regulator of IL-2 deprivation-induced T-cell apoptosis, is deficient in systemic lupus erythematosus. PNAS. 2011. July;108(30):12443–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Dawisha SM, Gmelig-Meyling F, Steinberg AD. Assessment of clinical parameters associated with increased frequency of mutant T cells in patients with systemic lupus erythematosus. Arthritis Rheum. 1994. February;37(2):270–7. [DOI] [PubMed] [Google Scholar]
- 81.Zhang Z, Kyttaris VC, Tsokos GC. The Role of IL-23/IL-17 Axis in Lupus Nephritis. J Immunol. 2009;183(5):3160–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Budagyan VM, Bulanova EG, Sharova NI, et al. The resistance of activated T-cells from SLE patients to apoptosis induced by human thymic stromal cells. Immunol Lett. 1998;60(1):1–5. [DOI] [PubMed] [Google Scholar]
- 83.Wilhelm AJ, Rhoads JP, Wade NS, Major AS. Dysregulated CD4+ T cells from SLE-susceptible mice are sufficient to accelerate atherosclerosis in LDLr−/− mice. Ann Rheum Dis. 2015;74(4):778–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Stanic AK, Stein CM, Morgan AC, et al. Immune dysregulation accelerates atherosclerosis and modulates plaque composition in systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2006. May;103(18):7018–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Braun N a Wade NS, Wakeland EK Major AS. Accelerated atherosclerosis is independent of feeding high fat diet in systemic lupus erythematosus-susceptible LDLr(−/−) mice. Lupus. 2008. December;17(12):1070–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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. [DOI] [PubMed] [Google Scholar]
- 87.Mengya Z, Hanyou M, Dong L, et al. Th17/Treg imbalance induced by increased incidence of atherosclerosis in patients with Systemic Lupus Erythematosus (SLE). Clin Rheumatol. 2013;32(7):1045–52. [DOI] [PubMed] [Google Scholar]
- 88.Cuda CM, Li S, Liang S, et al. Pre-B cell leukemia homeobox 1 is associated with lupus susceptibility in mice and humans. J Immunol. 2012;188(2):604–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Choi S-C, Hutchinson TE, Titov AA, et al. The lupus susceptibility gene Pbx1 regulates the balance between follicular helper T cell and regulatory T cell differentiation. J Immunol. 2016;197(2):458–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Li W, Elshikha AS, Cornaby C, et al. T cells expressing the lupus susceptibility allele Pbx1d enhance autoimmunity and atherosclerosis in dyslipidemic mice. JCI Insight. 2020. June 4;5(11): e138274.**This study provides the first evidence of a reciprocal relationship between a lupus susceptibility allele that drives autoimunity and atheroslcerosis severity. Overexpression of lupus susceptibility allele Pbx1d in CD4+ T cells increases atherosclerosis severity while atherosclerosis-related dyslipidemia promotes heightened autoimmunity.
- 91.Katsiari CG, Kyttaris VC, Juang YT, Tsokos GC. Protein phosphatase 2A is a negative regulator of IL-2 production in patients with systemic lupus erythematosus. J Clin Invest. 2005;115(11):3193–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sunahori K, Juang Y-T, Kyttaris VC, Tsokos GC. Promoter hypomethylation results in increased expression of protein phosphatase 2A in T Cells from patients with systemic lupus erythematosus. J Immunol. 2011;186(7):4508–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Crispín JC, Apostolidis SA, Finnell MI, Tsokos GC. Induction of PP2A Bβ, a regulator of IL-2 deprivation-induced T-cell apoptosis, is deficient in systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2011;108(30):12443–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Apostolidis SA, Rodríguez-Rodríguez N, Suárez-Fueyo A, et al. Phosphatase PP2A is requisite for the function of regulatory T cells. Nat Immunol. 2016;17(5):556–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Pan W, Sharabi A, Ferretti A, et al. PPP2R2D suppresses IL-2 production and Treg function. JCI Insight. 2020. October 2;5(19):138215.* This work identifies a regulatory subunit of PP2A that controls IL-2 transcription in T cells by limiting accessibility to the gene through chromatin remodeling. As PP2A negatively impacts SLE severity and cannot be systemically inhibited due to its ubiquitous expression, identification of regulatory subunits like the one described could allow for the development of more targeted therapeutics.
- 96.Rose A, von Spee-Mayer C, Kloke L, et al. IL-2 therapy diminishes renal inflammation and the activity of kidney-infiltrating CD4+ T cells in murine lupus nephritis. Cells. 2019;8(10):1234.*This study provides rationale for the currently ongoing low-dose IL-2 clinical trials in SLE by deomonstrating IL-2 administration in mice with lupus nephritis reduces disease severity by inhibiting conventional CD4+ T cell hyperactivity and expanding Tregs.
- 97.Dietrich T, Hucko T, Schneemann C, et al. Local delivery of IL-2 reduces atherosclerosis via expansion of regulatory T cells. Atherosclerosis. 2012;220(2):329–36. [DOI] [PubMed] [Google Scholar]
- 98.Humrich JY, von Spee-Mayer C, Siegert E, et al. Low-dose interleukin-2 therapy in refractory systemic lupus erythematosus: an investigator-initiated, single-centre phase 1 and 2a clinical trial. Lancet Rheumatol. 2019;1(1):e44–54. [DOI] [PubMed] [Google Scholar]
- 99.Zhao TX, Kostapanos M, Griffiths C, et al. Low-dose interleukin-2 in patients with stable ischaemic heart disease and acute coronary syndromes (LILACS): Protocol and study rationale for a randomised, double-blind, placebo-controlled, phase I/II clinical trial. BMJ Open. 2018;8(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Urowitz MB, Su J, Gladman DD. Atherosclerotic vascular events in systemic lupus erythematosus: An evolving story. J Rheumatol. 2020. January;47(1):66–71.*This work takes a broad view of SLE-accelerated atherosclerosis and evaluates whether treatment has improved over time. Results indicate premature atherosclerotic vascular events have declined in the modern era, suggesting current interventions for both SLE and CVD are more effective than prior strategies.