Skip to main content
  • Review
  • Published:

Unmet medical needs in systemic lupus erythematosus

Abstract

Systemic lupus erythematosus (SLE) is an autoimmune disease of diverse manifestations, with onset usually in young women in the third to fourth decade of life. The chronic nature of this relapsing remitting disease leads to organ damage accrual over time. Mortality and morbidity are increased in patients with SLE compared with the general population. Therapeutic advances over the last few decades have led to significant improvements in patient outcomes. Five-year survival has improved to over 90% from a low of 50% in the 1950s. However, multiple aspects of the management of SLE patients are still far from optimal. Early diagnosis remains a challenge; diagnostic delays leading to delay in definitive treatment are common. Monitoring treatment remains problematic due to the paucity of sensitive biomarkers. Current treatment regimens rely heavily on corticosteroids, even though corticosteroids are well known to cause organ damage. Treatment of refractory disease manifestations such as nephritis, recalcitrant cutaneous lesions and neurological involvement require new approaches with greater efficacy. Cognitive dysfunction is common in SLE patients, but early recognition and adequate treatment are yet to be established. Premature accelerated atherosclerosis remains a leading cause of morbidity and mortality. Fatigue is one of the most disabling symptoms, and contributes to the poor quality of life in patients with SLE. Ongoing research in SLE faces many challenges, including enrollment of homogeneous patient populations, use of reliable outcome measures and a standard control arm. The current review will highlight some of the outstanding unmet challenges in the management of this complex disease.

Introduction

Systemic lupus erythematosus (SLE) is a complex autoimmune disease, predominantly affecting young women during the prime years of their life. The chronic nature of the disease, its relapsing remitting course and organ damage accrual over time frustrate both the physician and the patient. Clinical and translational research has advanced the available therapeutic options, translating into better patient outcomes. Five-year survival in patients with SLE has improved from 50% in the 1950s to over 90% currently. However, the mortality still remains high compared with the general population.

Multiple aspects of SLE remain challenging. The diverse and nonspecific presentations can lead to delay in diagnosis. Disease monitoring remains difficult due to the low sensitivity of current disease activity markers. Management of refractory disease, especially nephritis, cutaneous and neuropsychiatric manifestations, remains unsatisfactory. End-stage renal failure, scarring cutaneous lesions and neurological damage remain fearsome complications of the disease. Cardiovascular disease secondary to accelerated atherosclerosis has emerged as an important contributor to the higher morbidity and mortality in longstanding disease. Damage due to both disease and treatment, especially corticosteroid-associated damage, tends to accumulate over time. Fatigue, fibro-myalgia and depression negatively impact the quality of life (QoL). Clinical research in the field of SLE therapeutics has met with limited success in recent years. Heterogeneous patient populations, limitations of out-come measures and the lack of a uniform control group are mostly responsible for the suboptimal responses.

Although there are numerous unmet medical needs in SLE, this review will focus on some of the major out-standing issues - including early diagnosis, biomarkers in SLE, management of refractory disease, atherosclerosis in SLE, corticosteroid-associated damage, QoL in SLE, and clinical research in SLE.

Early diagnosis

SLE is a multi-system heterogeneous disease with protean manifestations. Initial nonspecific presentations can lead to diagnostic delays. The revised American College of Rheumatology (ACR) classification criteria, although not designed for diagnostic purposes, have been used by rheumatologists for almost three decades [1]. The Systemic Lupus International Collaborating Clinics (SLICC) group recently developed another set of revised criteria. These new criteria include 17 variables derived by expert consensus (SLICC committee members) and statistical analysis, using real-life patient scenarios. The final set of new criteria was then validated in another group of SLE patients and controls. The control group comprised other autoimmune diseases, which may have overlapping features with SLE.

SLICC criteria require that at least one clinical criterion and one immunologic criterion be present, with a total of four criteria, to have a classification of SLE. Under this new classification, lupus nephritis by biopsy (in the presence of SLE autoantibodies) is sufficient for classification [2]. Using expert consensus as the gold standard, the revised criteria demonstrated greater sensitivity (97% vs. 83%, P <0.0001) but less specificity (84% vs. 96%, P <0.0001) than the current ACR criteria in the validation group [2]. These criteria are clinically more relevant and will probably identify more patients with clinically defined lupus than using the current ACR criteria. How-ever, one should stress that these criteria are primarily meant for classification of patient cohorts for research and their use for diagnostic purposes has to be carried out with caution. Some patients may initially present with insufficient features to fulfill the classification criteria, termed 'incomplete lupus' or 'lupus-like disease' by some groups.

Biomarkers in systemic lupus erythematosus

SLE is the most diverse autoimmune disease, clinically and serologically. Genetic influences as well as epigenetic and environmental interactions probably play a role, perhaps dependent on the ethnic background of the individual. Classification criteria may help but currently, there is no single parameter that is sensitive or specific enough to correctly identify or subtype all SLE patients. Similarly, the disease course can be variable with either intermittent flares or chronic activity. Levels of auto-antibodies (anti-double-stranded (DNA) antibody) and complement components represent serologic disease activity. They are routinely used to monitor disease activity in most clinical settings, but their association with clinical activity has not been consistent in longitudinal studies [3].

There is an unmet need for more sensitive and reliable biomarkers that can predict susceptibility, activity, severity and disease subtype in SLE. Multiple candidate markers have been proposed, including the type 1 interferon signature, B-lymphocyte stimulator and many others [4–13]. These markers are currently only a research tool and not a single biomarker has been validated for clinical use to date. As SLE is a heterogeneous disease, a single biomarker is unlikely to be sufficient. Rather, different markers may provide information about specific disease aspects, as summarized in Table 1.

Table 1 Potential biomarkers and their proposed applications in SLE

Management of refractory disease

There is no clear definition of refractory disease in SLE, but it generally refers to patients who fail to respond to conventional treatments. In a heterogeneous disease such as SLE, the clinical situation may vary, depending on the disease manifestations and organ involvement. Although any clinical feature may become persistent and non-responsive to therapy, the most concerning features are refractory lupus nephritis, scarring cutaneous disease and neuropsychiatric lupus (NPSLE).

Lupus nephritis

Renal involvement is a major cause of mortality and morbidity in SLE. A large proportion of patients, up to 60%, develop immune complex-mediated lupus nephritis during the course of their disease. The treatment of lupus nephritis has rapidly advanced over the last few decades. Glucocorticoids and cyclophosphamide were once considered the standard of care. Although effective in the majority of patients, cyclophosphamide was associated with serious adverse effects including infections, malignancy and infertility. Lower doses of cyclophosphamide and mycophenolate emerged as effective options for induction therapy, with better safety profiles [14, 15]. Patients with different ethnic backgrounds might have differential responses to cyclophosphamide versus mycophenolate [16]. Azathioprine and mycophenolate have been shown to be effective options for maintenance therapy in randomized trials [17, 18]. Mycophenolate was superior to azathioprine in the ALMS trial, but not in a MAINTAIN trial [17, 18].

Alternate approaches have been tried for patients failing to respond to these conventional treatments. Calcineurin inhibitors, cyclosporine and recently tacrolimus have shown promising results in patients unresponsive to first-line therapies, but need further evaluation in larger controlled trials [19–21]. B cells play a central role in the pathogenesis of lupus nephritis, making them a logical therapeutic target. Rituximab, a chimeric anti-CD20 antibody, efficiently and reliably depletes CD20-positive B cells. A large number of open-label studies documented the efficacy of rituximab in refractory lupus nephritis [22]. Unfortunately, a large randomized controlled trial (LUNAR) did not show any significant differences in outcomes with rituximab compared with placebo [23]. However, this trial excluded patients with refractory disease, the very subset in which evidence of benefit was shown in open-label studies. Additionally, heavy background immunosuppression may have masked any beneficial effect of rituximab [24]. Open-label data continue to be positive and a recent systematic review concluded that evidence for rituximab efficacy in refractory lupus nephritis is strong, and another well-conducted trial may provide more answers [25, 26].

Hydroxychloroquine deserves special mention in the treatment of SLE, including lupus nephritis. Hydroxychloroquine has been shown to improve response rates, decrease flares and improve survival [27, 28]. Every SLE patient should receive hydroxychloroquine, unless in-tolerant or contraindicated. Renal protective therapies such as angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, strict control of blood pressure and serum lipids, are important adjuncts to therapeutic regimens for lupus nephritis [29, 30].

Despite aggressive immunosuppressive and supportive therapies, induction of remission may be slow or partial, relapses remain common and progression to renal damage may occur. The incidence of end-stage renal disease attributable to lupus nephritis has not declined over the decades [31, 32]. About 10 to 30% of SLE patients still progress to end-stage renal disease with associated morbidity and mortality. Multiple variables affect renal prognosis, including the disease severity, antibody associations, ethnicity, genetic background, socioeconomic status and concomitant co-morbidities [33, 34].

Cutaneous lupus

Skin involvement occurs in 70 to 85% of SLE patients and includes acute, subacute and chronic cutaneous lesions. Discoid lupus is the most common form of chronic lesion, and may be the initial presentation of SLE in up to 10% of cases. Resistant discoid lesions may cause scarring of the affected skin areas and permanent scarring alopecia. This can lead to significant disfigurement emotional distress, physical limitations and disability in a large number of patients. Although generally believed to be associated with less severe systemic disease, discoid lupus has also been associated with more damage accrual [35]. In addition, the risk of squamous cell carcinoma is increased in scarred areas.

The conventional approaches include photoprotection, corticosteroids (topical, intralesional and systemic) and antimalarial agents [36]. Other topical agents such as calcineurin inhibitors and retinoids can benefit some patients with refractory lesions. Multiple immunosuppressants including methotrexate, azathioprine, mycophenolate, cyclophosphamide, dapsone, gold and thalidonamide have been tried for refractory disease [36]. Biologic agents including TNF inhibitors, efazulimab (anti-CD11 antibody) rituximab (anti-CD20 antibody) and tocilizumab (anti-IL-6) have been used in some refractory cases [37]. Data are limited to anecdotal reports, case series and open studies. A recent Cochrane review concluded that the evidence about therapies for discoid lupus, other than topical corticosteroids and anti-malarials, was not conclusive [38]. The need for additional therapies for such a disfiguring manifestation of young people with SLE cannot be overemphasized.

Neuropsychiatric lupus

NPSLE remains one of the most challenging issues in the management of SLE patients. Affecting up to 30 to 40% of the patients, NPSLE includes diverse neurologic and psychiatric manifestations, from subtle cognitive deficits to severe psychosis, seizures and strokes. The attribution of neuropsychiatric events to SLE is often challenging in the clinical setting. The ACR committee has described case definitions for 19 NPSLE syndromes, but the specificity remains low [39, 40]. Anti-ribosomal P antibodies have been associated with NPSLE, especially psychosis [41]. Measurement of cerebral spinal fluid cytokine and chemokine levels remains a research tool [42]. Neuro-imaging can be helpful but the sensitivity and specificity remain quite low [43].

The management of NPSLE depends on the manifestation and the likely predominant mechanism. Current strategies include the use of immunosuppressive therapies when the underlying mechanisms are predominantly inflammatory. Anticoagulation and/or antiplatelet therapy should be considered when antiphospholipid antibodies are persistently positive in moderate to high titers. Non-SLE precipitation and aggravating factors should be addressed. Cognitive dysfunction merits special mention. It is present in a majority of SLE patients, even newly diagnosed patients [44, 45], and has also been associated with psychosocial factors such as depression, fatigue, anxiety and pain [45, 46]. Although antidepressants may benefit some patients with depression and cognitive dysfunction, no treatment of proven benefit exists for cognitive dysfunction in SLE [45, 47].

NPSLE management remains problematic due to the lack of specific tools for diagnosis and attribution. Treatment options are limited to glucocorticoids and a few immunosuppressants (cyclophosphamide, azathioprine, mycophenolate, rituximab), with efficacy mostly suggested by case reports and open-label studies. A European League Against Rheumatism task force recently published recommendations on the management of NPSLE; it was felt that 'there is currently no good quality evidence to guide several diagnostic, primary prevention, therapeutic and monitoring decisions in NPSLE, emphasizing the need for further research' [48].

Atherosclerosis and systemic lupus erythematosus

SLE is associated with premature and accelerated atherosclerosis, contributing significantly to the increased mortality and morbidity associated with the disease [49]. An increased frequency of conventional risk factors, such as hypertension, dyslipidemia and diabetes, has been noted in patients with SLE. Yet the excess risk cannot be fully explained by the traditional Framingham risk factors [50, 51].

Multiple putative mechanisms have been proposed but the exact pathogenesis of atherosclerosis in the setting of SLE is yet to be fully elucidated. High-density lipoprotein was reported to be significantly dysfunctional and pro-inflammatory in SLE patients, and correlated with measures of subclinical atherosclerosis [52]. Endothelial cell dysfunction in SLE leads to abnormal vascular reactivity and repair, contributing to the accelerated atherogenesis [53]. Interferon activity was independently associated with subclinical measures of atherosclerosis in a cohort study [54]. Specific subtypes of peripheral blood mononuclear cells, the low-density granulocytes, and increased Toll-like receptor signaling have been proposed to contribute to higher interferon production and vascular dysfunction in patients with SLE [55, 56]. Autoantibodies including antiphospholipid and antilipoprotein antibodies have been associated with abnormal vascular function and atherosclerosis in SLE [57].

Despite the evidence of a link between inflammation and atherosclerosis in SLE, multiple studies have failed to show any consistent association of coronary calcium scores or carotid plaques with markers of disease activity [51, 58, 59]. In patients with SLE, the proportion of noncalcified vulnerable plaque was shown to be increased compared with the calcified stable plaque, and correlated with measures of disease activity [60]. Elevated homocysteine, asymmetric dimethylarginine, leptin and high-sensitivity C-reactive protein levels have been proposed as markers of accelerated atherosclerosis in SLE in some studies [61–64].

The management of atherosclerosis in SLE is currently limited to the control of traditional risk factors. Statins are believed to have anti-inflammatory properties, in addition to their lipid-lowering effects. However, two large randomized controlled trials failed to show any beneficial effects of 20 mg atorvastatin versus placebo in adult and pediatric SLE patients without clinical cardiovascular disease [65, 66]. The use of statins in SLE patients should thus be limited to treat hyperlipidemia. Hydroxychloroquine was noted to have weak protective effects on cardiovascular risk in SLE [28]. Mycophenolate reduced the atherosclerotic burden in mice models but failed to reduce progression of subclinical atherosclerosis in a large cohort study [67, 68]. Several trials evaluating anti-interferon therapy in SLE are currently ongoing, and beneficial effects on atherosclerosis might become evident. However, no SLE treatment is currently of proven benefit to reduce the risk of or halt the progression of atherosclerosis in SLE.

Corticosteroid-associated damage in systemic lupus erythematosus

Corticosteroids are the mainstay of therapy for SLE, with proven efficacy. The harm they cause in the short term and the long term, however, is one of the major issues in SLE management. Corticosteroids increase the traditional cardiovascular risk factors, including serum lipids, blood pressure, weight and glucose. Fifteen years after the diagnosis of SLE, the majority of permanent organ damage can be attributed to the corticosteroids. Although the risk of damage is higher with high doses, there is no safe dose for chronic use. Even small doses, if continued long enough, will significantly increase the morbidity [69]. The cumulative dose of corticosteroids has significant association with cataracts and osteoporotic fractures. Both the current dose and the cumulative dose are associated with cardiovascular events [70]. When adjusted for confounding by indication due to SLE disease activity, the hazard ratio for organ damage increases dramatically with prednisone doses of 6 to 12 mg/day (hazard ratio = 1.5), 12 to 18 mg/day (hazard ratio = 1.64) and >18 mg/day (hazard ratio = 2.51) [69]. Every attempt should be made to minimize the dose and duration of corticosteroid exposure.

Quality of life in systemic lupus erythematosus

The QoL in patients with SLE is significantly lower than in healthy controls and patients with other chronic diseases [71, 72]. Disease activity and organ damage did not consistently correlate with lower QoL in SLE. Instead, the major predictors of poor QoL in SLE are nondisease-specific variables including fatigue, chronic pain and mood disturbances [71, 72].

Fatigue is a common and often crippling symptom experienced by about 85 to 92% of patients with SLE, with 50% rating it as the most disabling symptom [73]. Fatigue can significantly contribute to the poor QoL in SLE patients [72]. The pathophysiological mechanisms of SLE-related fatigue are probably multifactorial, with a predominant role being played by the psychological domains. Psychosocial factors such as mood disorders, anxiety, poor sleep quality and chronic pain syndrome have shown consistent associations with fatigue in SLE [74, 75]. Exercise programs have been shown to have a positive impact on fatigue among SLE patients [76]. However, fatigue remains an unmet need, reflected by the finding that 81% of SLE patients feel that the healthcare service did not support them enough in the management of SLE-related fatigue [77].

Fibromyalgia is a chronic pain disorder characterized by widespread generalized pain, often associated with fatigue, anxiety and sleep disturbances. The prevalence of fibromyalgia is much higher in SLE patients, compared with the general population [78]. Fibromyalgia in SLE contributes to the lower QoL and correlates with psychosomatic and affective variables but not with disease activity or damage [78–80]. The widespread pain of concomitant fibromyalgia can lead to diagnostic confusions and potential overtreatment if symptoms are mistaken for SLE disease activity.

Mood disturbances are very common in patients with SLE, with depression being the most prevalent affective symptom [81, 82]. Depression contributes to the fatigue and cognitive dysfunction, and significantly correlates with lower QoL in patients with SLE [45, 71, 83]. Although psychological effects of dealing with a chronic disease may contribute to the high prevalence of depression, disease-specific mechanisms probably play a role. Associations with specific antibodies and alterations in cerebral blood flow have been reported in depressed SLE patients [84, 85]. However, the data are not conclusive and depression in patients with SLE should be treated with conventional measures similar to the general population.

Perspective on future therapeutics and clinical research in SLE

Despite advances in therapies, the control of disease activity in SLE remains suboptimal. Flares are common and sustained disease control is generally limited to a small fraction of patients [86]. These findings suggest that, despite significant improvements in SLE treatment, conventional approaches have probably reached their maximal benefit and alternate options have to be considered. Multiple new agents with immunomodulatory effects have been investigated in recent years but limited success has been achieved. Two large randomized controlled trials evaluating rituximab for treatment of SLE failed to meet their primary endpoints, despite good efficacy data in open-label studies [23, 87]. Only belimumab showed efficacy in randomized controlled trials and received US Food and Drug Administration approval for treatment of SLE [88, 89]. These studies have raised important issues that should be addressed in future SLE research.

The heterogeneous nature of the disease makes it difficult to design clinical studies in SLE. In the absence of specific biomarkers, classification criteria are generally used to define study populations. Although these criteria encompass the breadth of the disease spectrum, patients with diverse manifestations and probably different pathogenic mechanisms will be grouped together. This limitation can be avoided by defining specific disease subgroups based on organ manifestations, such as renal disease. However, this would seriously limit the eligible patient population, however, stressing the need for multicenter collaborative projects. Limiting the inclusion in this manner may not be even a viable option if uncommon manifestations are considered. Currently, some degree of heterogeneity in study populations has to be accepted.

Another major issue in SLE research has been the choice of outcome measures. The US Food and Drug Administration draft guidance statement on SLE clinical trials suggested the use of disease activity indices to measure the efficacy of the intended intervention [90]. Several such indices have been developed and validated for use in clinical trials. Some disease activity indices - such as the SLE Disease Activity Index (SLEDAI) and its variants (SLEDAI-2K, SELENA-SLEDAI) - measure overall disease activity, while others - such as the British Isles Lupus Assessment Group Index (BILAG) - measure organ-specific activity. Physician's global assessment uses the treating physician's overall assessment to assign a numerical value to the disease activity on a visual analog scale of 0 to 3 [91]. This assessment has been used to define flares (>1 point rise) in clinical trials [91]. Physician visual analog scale rating was found to have high variability in a comparative study of outcome measures by the SLICC study group [92], but it has been successfully incorporated as part of the SLE Responder Index (SRI) [93]. The SLE Responder Index is a composite index developed to incorporate the strengths of different disease activity indices. This index provides a comprehensive definition of meaningful clinical response and has been used to define the primary end point in clinical trials [93]. The SLE Responder Index utilizes the SELENA-SLEDAI score to determine global improvement, British Isles Lupus Assessment Group Index domain scores to ensure no significant worsening in previously unaffected organ systems, and physician's global assessment to ensure that improvements in disease activity are not achieved at the expense of the patient's overall condition [93]. The SLE Responder Index has been used successfully in the belimumab phase 3 trials and has the potential to serve as an outcome measure in the future SLE therapeutic trials.

The background immunosuppression used as the standard of care in SLE trials adds another confounder to the picture. Most studies have employed diverse background treatments in both the treatment and placebo groups, to which the candidate agent is added. In addition, treatment adjustments have been either mandated or at least allowed during the studies. However, these concomitant therapies may have their own effects, masking the efficacy of the target intervention. Examples include the failed LUNAR and EXPLORER trials, in which rituximab or placebo were added to background therapy including high-dose corticosteroids and immunosuppressive drugs such as mycophenolate [23, 87]. Efficacy of rituximab over placebo was not found in these trials, despite a large body of evidence favoring rituximab in observational and cohort studies. In contrast, the belimumab trial design permitted early tapering of corticosteroids and may have contributed, at least partially, to the positive results [88, 89]. However, designing a trial in SLE where the active arm receives only the experimental agent will be difficult and unethical. There is a need to develop a clearly defined standard of care control arm, against which the newer agents can be tested.

The field of SLE clinical and therapeutic research is advancing rapidly. Large international collaborations have resulted in development of new criteria, composite outcome indices and insights into disease pathogenesis. Multiple newer biologic agents targeting specific immune system pathways and effectors are undergoing evaluation [94]. Hopefully, these efforts will lead to development of newer therapeutic agents in SLE, a dire need.

Conclusions

The management of SLE remains a challenge despite significant advances in the treatment. The new SLICC classification criteria with better sensitivity will probably help in better identification of patients, but we have not yet reached the stage where early diagnosis can be universally achieved. The field of disease biomarkers is rapidly evolving with many putative candidates. However, no biomarker has been successfully validated for use in the routine clinical setting. Instead of a single measure, a battery of markers may perhaps be developed in the future to predict different disease aspects in a heterogeneous disease such as SLE. Refractory disease manifestations and development of damage associated with disease and therapies remain outstanding issues. Premature mortality and higher morbidity from atherosclerosis in this predominantly young group of patients are a major concern. Fatigue is one of the most prevalent and disabling symptom in SLE, significantly contributing to the poor QoL. The quest for newer targeted therapies has met with limited success despite many clinical trials. Whether there is a true lack of efficacy or whether the trial designs in SLE are partly to blame is open to discussion. However, advances have been made in developing reliable outcome measures for clinical research. Future research will focus on the goals of increasing survival, limiting organ damage and improving QoL for patients with SLE.

Declarations

This article has been published as part of Arthritis Research & Therapy Volume 14 Suppl 4, 2012: New therapeutic targets in systemic lupus erythematosus. The supplement was proposed and developed by the journal. Articles were commissioned by the journal, were independently prepared by the authors and have undergone the journal's standard peer review process. Publication of the supplement has been supported by an unrestricted educational grant from UCB. Completed articles underwent a data quality check by Darwin Healthcare Communications, funded by UCB

Abbreviations

ACR:

American College of Rheumatology

IL:

interleukin

NPSLE:

neuropsychiatric lupus

QoL:

quality of life

SLE:

systemic lupus erythematosus

SLEDAI:

SLE Disease Activity Index

SLICC:

Systemic Lupus International Collaborating Clinics

TNF:

tumor necrosis factor.

References

  1. Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ: The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982, 25: 1271-1277. 10.1002/art.1780251101.

    Article  CAS  PubMed  Google Scholar 

  2. Petri M, Orbai AM, Alarcon GS, Gordon C, Merrill JT, Fortin PR, Bruce IN, Isenberg D, Wallace DJ, Nived O, Sturfelt G, Ramsey-Goldman R, Bae SC, Hanly JG, Sanchez-Guerro J, Clarke A, Aranow C, Manzi S, Urowitz M, Gladman D, Kalunian K, Costner M, Werth VP, Zoma A, Bernatsky S, Ruiz-Irastorza G, Khasmashta MA, Jacobsen S, Buyon JP: Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus. Arthritis Rheum. 2012, 64: 2677-2686. 10.1002/art.34473.

    Article  PubMed Central  PubMed  Google Scholar 

  3. Ho A, Magder LS, Barr SG, Petri M: Decreases in anti-double-stranded DNA levels are associated with concurrent flares in patients with systemic lupus erythematosus. Arthritis Rheum. 2001, 44: 2342-2349. 10.1002/1529-0131(200110)44:10<2342::AID-ART397>3.0.CO;2-8.

    Article  CAS  PubMed  Google Scholar 

  4. Liu CC, Ahearn JM: The search for lupus biomarkers. Best Pract Res Clin Rheumatol. 2009, 23: 507-523. 10.1016/j.berh.2009.01.008.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Niewold TB, Kelly JA, Kariuki SN, Franek BS, Kumar AA, Kaufman KM, Thomas K, Walker D, Kamp S, Frost JM, Wong AK, Merrill JT, Alarcon-Riquelme NE, Tikly M, Ramsey-Goldman R, Reveille JD, Petri MA, Edberg JC, Kimberly RP, Alarcon GS, Kamen DL, Gilkeson GS, Vyse TJ, James JA, Gaffney PM, Moser KL, Crow MK, Harley JB: IRF5 haplotypes demonstrate diverse serological associations which predict serum interferon alpha activity and explain the majority of the genetic association with systemic lupus erythematosus. Ann Rheum Dis. 2012, 71: 463-468. 10.1136/annrheumdis-2011-200463.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Hughes T, Adler A, Kelly JA, Kaufman KM, Williams AH, Langefeld CD, Brown EE, Alarcon GS, Kimberly RP, Edberg JC, Ramsey-Goldman R, Petri M, Boackle SA, Stevens AM, Reveille JD, Sanchez E, Martin J, Niewold TB, Vila LM, Scofield RH, Gilkeson GS, Gaff ney PM, Criswell LA, Moser KL, Merrill JT, Jacob CO, Taso BP, James JA, Vyse TJ, Alarcon-Riquelme ME, BIOLUPUS Network, Harley JB, Richardson BC, Sawalha AH: Evidence for gene-gene epistatic interactions among susceptibility loci for systemic lupus erythematosus. Arthritis Rheum. 2012, 64: 485-492. 10.1002/art.33354.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Seyhan A, Toole MA, Zhang Y, Immermann FW, Hill A, Reddy P, Masferrer J, Zhou T, Mounts W, Whitley M: Impact of baseline Interferon pathway activation on widespread gene expression changes with disease flare in lupus patients: interim report from the BOLD (Biomarkers of lupus disease) study. Ann Rheum Dis. 2012, 71 (Suppl 3): 74-

    Article  Google Scholar 

  8. Fernando MM, Freudenberg J, Lee A, Morris DL, Boteva L, Rhodes B, Gonzalez-Escribano MF, Lopez-Nevot MA, Navarra SV, Gregersen PK, Martin J, Vyse TJ: Transancestral mapping of the MHC region in systemic lupus erythematosus identifies new independent and interacting loci at MSH5, HLA-DPB1 and HLA-G. Ann Rheum Dis. 2012, 71: 777-784. 10.1136/annrheumdis-2011-200808.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK: Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005, 52: 1491-1503. 10.1002/art.21031.

    Article  CAS  PubMed  Google Scholar 

  10. Petri M, Singh S, Tesfasyone H, Dedrick R, Fry K, Lal P, Williams G, Bauer J, Gregersen P, Behrens T, Baechler E: Longitudinal expression of type I interferon responsive genes in systemic lupus erythematosus. Lupus. 2009, 18: 980-989. 10.1177/0961203309105529.

    Article  CAS  PubMed  Google Scholar 

  11. Landolt-Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J, Gladman DD, Urowitz M, Fortin PR, Wither J: Lack of association between the interferon-alpha signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann Rheum Dis. 2009, 68: 1440-1446. 10.1136/ard.2008.093146.

    Article  CAS  PubMed  Google Scholar 

  12. Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J, Recta V, Zhong J, Freimuth W: Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 2453-2459. 10.1002/art.23678.

    Article  CAS  PubMed  Google Scholar 

  13. Akhter E, Burlingame RW, Seaman AL, Magder L, Petri M: Anti-C1q antibodies have higher correlation with flares of lupus nephritis than other serum markers. Lupus. 2011, 20: 1267-1274. 10.1177/0961203311411597.

    Article  CAS  PubMed  Google Scholar 

  14. Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, Li LS, Mysler E, Sanchez-Guerrero J, Solomons N, Wofsy D, Aspreva Lupus Management Study Group: Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol. 2009, 20: 1103-1112. 10.1681/ASN.2008101028.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, de Ramon Garrido E, Danieli MG, Abramovicz D, Blockmans D, Cauli A, Direskeneli HGaleazzi M, Gül A, Levy Y, Petera P, Popovic R, Sinico RA, Cattaneo R, Font J, Depresseux G, Cosyns JP, Carvera R: The 10-year follow-up data of the Euro-Lupus Nephritis Trial comparing low-dose and high-dose intravenous cyclophosphamide. Ann Rheum Dis. 2010, 69: 61-64. 10.1136/ard.2008.102533.

    Article  CAS  PubMed  Google Scholar 

  16. Isenberg D, Appel GB, Contreras G, Dooley MA, Ginzler EM, Jayne D, Sanchez- Guerrero J, Wofsy D, Yu X, Solomons N: Influence of race/ethnicity on response to lupus nephritis treatment: the ALMS study. Rheumatology (Oxford). 2010, 49: 128-140. 10.1093/rheumatology/kep346.

    Article  Google Scholar 

  17. Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, Eitner F, Appel GB, Contreras G, Lisk L, Solomons N, ALMS Group: Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N Engl J Med. 2011, 365: 1886-1895. 10.1056/NEJMoa1014460.

    Article  CAS  PubMed  Google Scholar 

  18. Houssiau FA, D'Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, Fiehn C, de Ramon Garrido E, Gilboe IM, Tektonidou M, Blockmans D, Ravelinigien I, le Guern V, Depresseux G, Guillevin L, Cervera R, MAINTAIN Nephritis Trial Group: Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: results from the MAINTAIN Nephritis Trial. Ann Rheum Dis. 2010, 69: 2083-2089. 10.1136/ard.2010.131995.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Chen W, Liu Q, Tang X, Fu P, Liu F, Liao Y, Yang Z, Zhang J, Chen J, Lou T, Fu J, Kong Y, Liu Z, Li Z, Yu X: Outcomes of maintenance therapy with tacrolimus versus azathioprine for active lupus nephritis: a multicenter randomized clinical trial. Lupus. 2012, 21: 944-952. 10.1177/0961203312442259.

    Article  CAS  PubMed  Google Scholar 

  20. Wang S, Li X, Qu L, Wang R, Chen Y, Li Q, He X, Zhang X, Wang H, Wu J, Xu Y, Chen J: Tacrolimus versus cyclophosphamide as treatment for diffuse proliferative or membranous lupus nephritis: a non-randomized prospective cohort study. Lupus. 2012, 21: 1025-1035. 10.1177/0961203312448105.

    Article  CAS  PubMed  Google Scholar 

  21. Zavada J, Pesickova S, Rysava R, Olejarova M, Horak P, Hrncir Z, Rychlik I, Havrda M, Vitova J, Lukac J, Rovensky J, Tegzova D, Böhmova J, Zadrazil J, Hána J, Dostál C, Tesar V: Cyclosporine A or intravenous cyclophosphamide for lupus nephritis: the Cyclofa-Lune study. Lupus. 2010, 19: 1281-1289. 10.1177/0961203310371155.

    Article  CAS  PubMed  Google Scholar 

  22. Ramos-Casals M, Soto MJ, Cuadrado MJ, Khamashta MA: Rituximab in systemic lupus erythematosus: a systematic review of off -label use in 188 cases. Lupus. 2009, 18: 767-776. 10.1177/0961203309106174.

    Article  CAS  PubMed  Google Scholar 

  23. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, Maciuca R, Zhang D, Garg JP, Brunetta P, Appel G, LUNAE Investigator Group: Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012, 64: 1215-1226. 10.1002/art.34359.

    Article  CAS  PubMed  Google Scholar 

  24. Lateef A, Petri M: Biologics in the treatment of systemic lupus erythematosus. Curr Opin Rheumatol. 2010, 22: 504-509. 10.1097/BOR.0b013e32833b475e.

    Article  PubMed  Google Scholar 

  25. Weidenbusch M, Rommele C, Schrottle A, Anders HJ: Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol Dial Transplant. 2012,

    Google Scholar 

  26. Diaz-Lagares C, Croca S, Sangle S, Vital EM, Catapano F, Martinez-Berriotxoa A, Garcia-Hernandez F, Callejas-Rubio JL, Rascon J, D'Cruz D, Jayne D, Ruiz-Irastorza G, Emery P, Isenberg D, Ramos-Casals M, Khamashta MA, UK-BIOGEAS Registry: Efficacy of rituximab in 164 patients with biopsyproven lupus nephritis: pooled data from European cohorts. Autoimmun Rev. 2012, 11: 357-364. 10.1016/j.autrev.2011.10.009.

    Article  CAS  PubMed  Google Scholar 

  27. Kasitanon N, Fine DM, Haas M, Magder LS, Petri M: Hydroxychloroquine use predicts complete renal remission within 12 months among patients treated with mycophenolate mofetil therapy for membranous lupus nephritis. Lupus. 2006, 15: 366-370. 10.1191/0961203306lu2313oa.

    Article  CAS  PubMed  Google Scholar 

  28. Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, Khamashta MA: Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review. Ann Rheum Dis. 2010, 69: 20-28. 10.1136/ard.2008.101766.

    Article  CAS  PubMed  Google Scholar 

  29. Kitamura N, Matsukawa Y, Takei M, Sawada S: Antiproteinuric effect of angiotensin-converting enzyme inhibitors and an angiotensin II receptor blocker in patients with lupus nephritis. J Int Med Res. 2009, 37: 892-898. 10.1177/147323000903700335.

    Article  CAS  PubMed  Google Scholar 

  30. Duran-Barragan S, McGwin G, Vila LM, Reveille JD, Alarcon GS: Angiotensin-converting enzyme inhibitors delay the occurrence of renal involvement and are associated with a decreased risk of disease activity in patients with systemic lupus erythematosus--results from LUMINA (LIX): amultiethnic US cohort. Rheumatology (Oxford). 2008, 47: 1093-1096. 10.1093/rheumatology/ken208.

    Article  CAS  Google Scholar 

  31. Croca SC, Rodrigues T, Isenberg DA: Assessment of a lupus nephritis cohort over a 30-year period. Rheumatology (Oxford). 2011, 50: 1424-1430. 10.1093/rheumatology/ker101.

    Article  Google Scholar 

  32. Ward MM: Changes in the incidence of endstage renal disease due to lupus nephritis in the United States, 1996-2004. J Rheumatol. 2009, 36: 63-67.

    PubMed Central  PubMed  Google Scholar 

  33. Barr RG, Seliger S, Appel GB, Zuniga R, D'Agati V, Salmon J, Radhakrishnan J: Prognosis in proliferative lupus nephritis: the role of socio-economic status and race/ethnicity. Nephrol Dial Transplant. 2003, 18: 2039-2046. 10.1093/ndt/gfg345.

    Article  PubMed  Google Scholar 

  34. Costenbader KH, Desai A, Alarcon GS, Hiraki LT, Shaykevich T, Brookhart MA, Massarotti E, Lu B, Solomon DH, Winkelmayer WC: Trends in the incidence, demographics, and outcomes of end-stage renal disease due to lupus nephritis in the US from 1995 to 2006. Arthritis Rheum. 2011, 63: 1681-1688. 10.1002/art.30293.

    Article  PubMed Central  PubMed  Google Scholar 

  35. Santiago-Casas Y, Vila LM, McGwin G, Cantor RS, Petri M, Ramsey-Goldman R, Reveille JD, Kimberly RP, Alarcon GS, Brown EE: Association of discoid lupus erythematosus with clinical manifestations and damage accrual in a multiethnic lupus cohort. Arthritis Care Res (Hoboken). 2012, 64: 704-712. 10.1002/acr.21581.

    Article  Google Scholar 

  36. Kuhn A, Ruland V, Bonsmann G: Cutaneous lupus erythematosus: update of therapeutic options part I. J Am Acad Dermatol. 2011, 65: e179-e193. 10.1016/j.jaad.2010.06.018.

    Article  PubMed  Google Scholar 

  37. Kuhn A, Ruland V, Bonsmann G: Cutaneous lupus erythematosus: update of therapeutic options part II. J Am Acad Dermatol. 2011, 65: e195-e213. 10.1016/j.jaad.2010.06.017.

    Article  PubMed  Google Scholar 

  38. Jessop S, Whitelaw DA, Delamere FM: Drugs for discoid lupus erythematosus. Cochrane Database Syst Rev. 2009, 4: CD002954-

    PubMed  Google Scholar 

  39. ACR adhoc Committee on Neuropsychiatric Lupus Nomenclature: The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Arthritis Rheum. 1999, 42: 599-608.

    Article  Google Scholar 

  40. Ainiala H, Hietaharju A, Loukkola J, Peltola J, Korpela M, Metsanoja R, Auvinen A: Validity of the new American College of Rheumatology criteria for neuropsychiatric lupus syndromes: a population-based evaluation. Arthritis Rheum. 2001, 45: 419-423. 10.1002/1529-0131(200110)45:5<419::AID-ART360>3.0.CO;2-X.

    Article  CAS  PubMed  Google Scholar 

  41. Karassa FB, Afeltra A, Ambrozic A, Chang DM, De Keyser F, Doria A, Galeazzi M, Hirohata S, Hoffman IE, Inanc M, Massardo L, Mathieu A, Mok CC, Morozzi G, Sanna G, Spindler AJ, Tzioufas AG, Yoshio T, Ioannidis JP: Accuracy of antiribosomal P protein antibody testing for the diagnosis of neuropsychiatric systemic lupus erythematosus: an international meta-analysis. Arthritis Rheum. 2006, 54: 312-324. 10.1002/art.21539.

    Article  CAS  PubMed  Google Scholar 

  42. Okamoto H, Kobayashi A, Yamanaka H: Cytokines and chemokines in neuropsychiatric syndromes of systemic lupus erythematosus. J Biomed Biotechnol. 2010, 2010: 268436-

    Article  PubMed Central  PubMed  Google Scholar 

  43. Luyendijk J, Steens SC, Ouwendijk WJ, Steup-Beekman GM, Bollen EL, van der Grond J, Huizinga TW, Emmer BJ, van Buchem MA: Neuropsychiatric systemic lupus erythematosus: lessons learned from magnetic resonance imaging. Arthritis Rheum. 2011, 63: 722-732. 10.1002/art.30157.

    Article  CAS  PubMed  Google Scholar 

  44. Petri M, Naqibuddin M, Carson KA, Sampedro M, Wallace DJ, Weisman MH, Holliday SL, Padilla PA, Brey RL: Cognitive function in a systemic lupus erythematosus inception cohort. J Rheumatol. 2008, 35: 1776-1781.

    Article  PubMed  Google Scholar 

  45. Petri M, Naqibuddin M, Carson KA, Wallace DJ, Weisman MH, Holliday SL, Sampedro M, Padilla PA, Brey RL: Depression and cognitive impairment in newly diagnosed systemic lupus erythematosus. J Rheumatol. 2010, 37: 2032-2038. 10.3899/jrheum.091366.

    Article  PubMed  Google Scholar 

  46. Kozora E, Ellison MC, West S: Depression, fatigue, and pain in systemic lupus erythematosus (SLE): relationship to the American College of Rheumatology SLE neuropsychological battery. Arthritis Rheum. 2006, 55: 628-635. 10.1002/art.22101.

    Article  PubMed  Google Scholar 

  47. Petri M, Naqibuddin M, Sampedro M, Omd al R, Carson KA: Memantine in systemic lupus erythematosus: a randomized, double-blind placebo-controlled trial. Semin Arthritis Rheum. 2011, 41: 194-202. 10.1016/j.semarthrit.2011.02.005.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Bertsias GK, Ioannidis JP, Aringer M, Bollen E, Bombardieri S, Bruce IN, Cervera R, Dalakas M, Doria A, Hanly JG, Huizinga TW, Isenberg D, Kallenberg C, Piette JC, Schneider M, Scolding N, Smolen J, Stara A, Tassiulas I, Tektonidou M, Tincani A, van Buchem MA, van Vollenhoven R, Ward M, Gordon C, Boumpas DT: EULAR recommendations for the management of systemic lupus erythematosus with neuropsychiatric manifestations: report of a task force of the EULAR standing committee for clinical affairs. Ann Rheum Dis. 2010, 69: 2074-2082. 10.1136/ard.2010.130476.

    Article  CAS  PubMed  Google Scholar 

  49. Nossent J, Cikes N, Kiss E, Marchesoni A, Nassonova V, Mosca M, Olesinska M, Pokorny G, Rozman B, Schneider M, Vlachioyiannopoulos PG, Swaak A: Current causes of death in systemic lupus erythematosus in Europe, 2000-2004: relation to disease activity and damage accrual. Lupus. 2007, 16: 309-317. 10.1177/0961203307077987.

    Article  CAS  PubMed  Google Scholar 

  50. Esdaile JM, Abrahamowicz M, Grodzicky T, Li Y, Panaritis C, du Berger R, Cote R, Grover SA, Fortin PR, Clarke AE, Senécal JL: Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. Arthritis Rheum. 2001, 44: 2331-2337. 10.1002/1529-0131(200110)44:10<2331::AID-ART395>3.0.CO;2-I.

    Article  CAS  PubMed  Google Scholar 

  51. Roman MJ, Shanker BA, Davis A, Lockshin MD, Sammaritano L, Simantov R, Crow MK, Schwartz JE, Paget SA, Devereux RB, Salmon JE: Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003, 349: 2399-2406. 10.1056/NEJMoa035471.

    Article  CAS  PubMed  Google Scholar 

  52. McMahon M, Grossman J, Skaggs B, Fitzgerald J, Sahakian L, Ragavendra N, Charles-Schoeman C, Watson K, Wong WK, Volkmann E, Chen W, Gorn A, Karpouzas G, Weisman M, Wallace DJ, Hahn BH: Dysfunctional proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheum. 2009, 60: 2428-2437. 10.1002/art.24677.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  53. Wright SA, O'Prey FM, Rea DJ, Plumb RD, Gamble AJ, Leahey WJ, Devine AB, McGivern RC, Johnston DG, Finch MB, Bell AL, McVeigh GE: Microcirculatory hemodynamics and endothelial dysfunction in systemic lupus erythematosus. Arterioscler Thromb Vasc Biol. 2006, 26: 2281-2287. 10.1161/01.ATV.0000238351.82900.7f.

    Article  CAS  PubMed  Google Scholar 

  54. Somers EC, Zhao W, Lewis EE, Wang L, Wing JJ, Sundaram B, Kazerooni EA, McCune WJ, Kaplan MJ: Type I interferons are associated with subclinical markers of cardiovascular disease in a cohort of systemic lupus erythematosus patients. PLoS One. 2012, 7: e37000-10.1371/journal.pone.0037000.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  55. Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, McCune WJ, Kaplan MJ: A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol. 2010, 184: 3284-3297. 10.4049/jimmunol.0902199.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  56. Huang Q, Pope RM: Toll-like receptor signaling: a potential link among rheumatoid arthritis, systemic lupus, and atherosclerosis. J Leukoc Biol. 2010, 88: 253-262. 10.1189/jlb.0310126.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  57. Matsuura E, Lopez LR, Shoenfeld Y, Ames PR: β2-glycoprotein I and oxidative inflammation in early atherogenesis: a progression from innate to adaptive immunity?. Autoimmun Rev. 2012, 12: 241-249. 10.1016/j.autrev.2012.04.003.

    Article  CAS  PubMed  Google Scholar 

  58. Kiani AN, Magder L, Petri M: Coronary calcium in systemic lupus erythematosus is associated with traditional cardiovascular risk factors, but not with disease activity. J Rheumatol. 2008, 35: 1300-1306.

    CAS  PubMed  Google Scholar 

  59. Maksimowicz-McKinnon K, Magder LS, Petr i M: Predictors of carotid atherosclerosis in systemic lupus erythematosus. J Rheumatol. 2006, 33: 2458-2463.

    PubMed  Google Scholar 

  60. Kiani AN, Vogel-Claussen J, Magder LS, Petri M: Noncalcified coronary plaque in systemic lupus erythematosus. J Rheumatol. 2010, 37: 579-584. 10.3899/jrheum.090824.

    Article  PubMed  Google Scholar 

  61. Kiani AN, Mahoney JA, Petri M: Asymmetric dimethylarginine is a marker of poor prognosis and coronary calcium in systemic lupus erythematosus. J Rheumatol. 2007, 34: 1502-1505.

    CAS  PubMed  Google Scholar 

  62. McMahon M, Skaggs BJ, Sahakian L, Grossman J, FitzGerald J, Ragavendra N, Charles-Schoeman C, Chernishof M, Gorn A, Witztum JL, Wong WK, Weisman M, Wallace DJ, La Cava A, Hahn BH: High plasma leptin levels confer increased risk of atherosclerosis in women with systemic lupus erythematosus, and are associated with inflammatory oxidised lipids. Ann Rheum Dis. 2011, 70: 1619-1624. 10.1136/ard.2010.142737.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  63. Roman MJ, Crow MK, Lockshin MD, Devereux RB, Paget SA, Sammaritano L, Levine DM, Davis A, Salmon JE: Rate and determinants of progression of atherosclerosis in systemic lupus erythematosus. Arthritis Rheum. 2007, 56: 3412-3419. 10.1002/art.22924.

    Article  CAS  PubMed  Google Scholar 

  64. Gustafsson J, Simard JF, Gunnarsson I, Elvin K, Lundberg IE, Hansson LO, Larsson A, Svenungsson E: Risk factors for cardiovascular mortality in patients with systemic lupus erythematosus, a prospective cohort study. Arthritis Res Ther. 2012, 14: R46-10.1186/ar3759.

    Article  PubMed Central  PubMed  Google Scholar 

  65. Petri MA, Kiani AN, Post W, Christopher -Stine L, Magder LS: Lupus Atherosclerosis Prevention Study (LAPS). Ann Rheum Dis. 2011, 70: 760-765. 10.1136/ard.2010.136762.

    Article  CAS  PubMed  Google Scholar 

  66. Schanberg LE, Sandborg C, Barnhart HX, Ardoin SP, Yow E, Evans GW, Mieszkalski KL, Ilowite NT, Eberhard A, Imundo LF, Kirmua Y, von Scheven E, Silverman E, Bowyer SL, Punaro M, Singer NG, Sherry DD, McCurdy D, Klein-Gitelman M, Wallace C, Silver R, Wagner-Weiner L, Higgins GC, Brunnger HI, Jung L, Soep JB, Reed AM, Provenzale J, Thompson SD, Athesrosclerosis Prevention in Pediatric Lupus Erythematosus Investigators: Use of atorvastatin in systemic lupus erythematosus in children and adolescents. Arthritis Rheum. 2012, 64: 285-296. 10.1002/art.30645.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  67. Kiani AN, Magder LS, Petri M: Mycophenolate mofetil (MMF) does not slow the progression of subclinical atherosclerosis in SLE over 2 years. Rheumatol Int. 2012, 32: 2701-2705. 10.1007/s00296-011-2048-y.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  68. van Leuven SI, Mendez-Fernandez YV, Wilhelm AJ, Wade NS, Gabriel CL, Kastelein JJ, Stroes ES, Tak PP, Major AS: Mycophenolate mofetil but not atorvastatin attenuates atherosclerosis in lupus-prone LDLr(-/-) mice. Ann Rheum Dis. 2012, 71: 408-414. 10.1136/annrheumdis-2011-200071.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Thamer M, Hernan MA, Zhang Y, Cotter D, Petri M: Prednisone, lupus activity, and permanent organ damage. J Rheumatol. 2009, 36: 560-564. 10.3899/jrheum.080828.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  70. Magder L, Petri M: Incidence of and risk factors for adverse cardiovascular events among patients with systemic lupus erythamatosus. Am J Epidemiol. 2012, 176: 708-719. 10.1093/aje/kws130.

    Article  PubMed Central  PubMed  Google Scholar 

  71. Choi ST, Kang JI, Park IH, Lee YW, Song JS, Park YB, Lee SK: Subscale analysis of quality of life in patients with systemic lupus erythematosus: association with depression, fatigue, disease activity and damage. Clin Exp Rheumatol. 2012, 30: 664-672.

    Google Scholar 

  72. Pettersson S, Lovgren M, Eriksson L, Moberg C, Svenungsson E, Gunnarsson I, Welin Henriksson E: An exploration of patient-reported symptoms in systemic lupus erythematosus and the relationship to health-related quality of life. Scand J Rheumatol. 2012, 41: 383-390. 10.3109/03009742.2012.677857.

    Article  CAS  PubMed  Google Scholar 

  73. Zonana-Nacach A, Roseman JM, McGwin G, Friedman AW, Baethge BA, Reveille JD, Alarcon GS: Systemic lupus erythematosus in three ethnic groups. VI: factors associated with fatigue within 5 years of criteria diagnosis. LUMINA Study Group. LUpus in MInority populations: NAture vs Nurture. Lupus. 2000, 9: 101-109. 10.1191/096120300678828046.

    Article  CAS  PubMed  Google Scholar 

  74. Burgos PI, Alarcon GS, McGwin G, Crews KQ, Reveille JD, Vila LM: Disease activity and damage are not associated with increased levels of fatigue in systemic lupus erythematosus patients from a multiethnic cohort: LXVII. Arthritis Rheum. 2009, 61: 1179-1186. 10.1002/art.24649.

    Article  PubMed Central  PubMed  Google Scholar 

  75. Jump RL, Robinson ME, Armstrong AE, Barnes EV, Kilbourn KM, Richards HB: Fatigue in systemic lupus erythematosus: contributions of disease activity, pain, depression, and perceived social support. J Rheumatol. 2005, 32: 1699-1705.

    PubMed  Google Scholar 

  76. Tench CM, McCarthy J, McCurdie I, White PD, D'Cruz DP: Fatigue in systemic lupus erythematosus: a randomized controlled trial of exercise. Rheumatology (Oxford). 2003, 42: 1050-1054. 10.1093/rheumatology/keg289.

    Article  CAS  Google Scholar 

  77. Moses N, Wiggers J, Nicholas C, Cockburn J: Prevalence and correlates of perceived unmet needs of people with systemic lupus erythematosus. Patient Educ Couns. 2005, 57: 30-38. 10.1016/j.pec.2004.03.015.

    Article  PubMed  Google Scholar 

  78. Buskila D, Press J, Abu-Shakra M: Fibromyalgia in systemic lupus erythematosus: prevalence and clinical implications. Clin Rev Allergy Immunol. 2003, 25: 25-28. 10.1385/CRIAI:25:1:25.

    Article  PubMed  Google Scholar 

  79. Torrente-Segarra V, Carbonell-Abello J, Castro-Oreiro S, Manresa Dominguez JM: Association between fibromyalgia and psychiatric disorders in systemic lupus erythematosus. Clin Exp Rheumatol. 2010, 28 (6 Suppl 63): S22-S26.

    CAS  PubMed  Google Scholar 

  80. Friedman AW, Tewi MB, Ahn C, McGwin G, Fessler BJ, Bastian HM, Baethge BA, Reveille JD, Alarcon GS: Systemic lupus erythematosus in three ethnic groups: XV. Prevalence and correlates of fibromyalgia. Lupus. 2003, 12: 274-279. 10.1191/0961203303lu330oa.

    Article  CAS  PubMed  Google Scholar 

  81. Bachen EA, Chesney MA, Criswell LA: Prevalence of mood and anxiety disorders in women with systemic lupus erythematosus. Arthritis Rheum. 2009, 61: 822-829. 10.1002/art.24519.

    Article  PubMed Central  PubMed  Google Scholar 

  82. Meszaros ZS, Perl A, Faraone SV: Psychiatric symptoms in systemic lupus erythematosus: a systematic review. J Clin Psychiatry. 2012, 73: 993-1001. 10.4088/JCP.11r07425.

    Article  PubMed  Google Scholar 

  83. Moldovan I, Katsaros E, Carr FN, Cooray D, Torralba K, Shinada S, Ishimori ML, Jolly M, Wallace DJ, Weisman MH, Nicassio PM: The Patient Reported Outcomes in Lupus (PATROL) study: role of depression in health-related quality of life in a Southern California lupus cohort. Lupus. 2011, 20: 1285-1292. 10.1177/0961203311412097.

    Article  CAS  PubMed  Google Scholar 

  84. Giovacchini G, Mosca M, Manca G, Della Porta M, Neri C, Bombardieri S, Ciarmiello A, Strauss HW, Mariani G, Volterrani D: Cerebral blood flow in depressed patients with systemic lupus erythematosus. J Rheumatol. 2010, 37: 1844-1851. 10.3899/jrheum.100121.

    Article  PubMed  Google Scholar 

  85. Lapteva L, Nowak M, Yarboro CH, Takada K, Roebuck-Spencer T, Weickert T, Bleiberg J, Rosenstein D, Pao M, Patronas N, Steele S, Manzano M, van der Veen JW, Lipsky PE, Marneco S, Wesley R, Volpe B, Diamond B, Illei GG: Anti-N-methyl-D-aspartate receptor antibodies, cognitive dysfunction, and depression in systemic lupus erythematosus. Arthritis Rheum. 2006, 54: 2505-2514. 10.1002/art.22031.

    Article  CAS  PubMed  Google Scholar 

  86. Nikpour M, Urowitz MB, Ibanez D, Gladman DD: Frequency and determinants of flare and persistently active disease in systemic lupus erythematosus. Arthritis Rheum. 2009, 61: 1152-1158. 10.1002/art.24741.

    Article  PubMed  Google Scholar 

  87. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, Utset TO, Gordon C, Isenberg DA, Hsieh HJ, Zhang D, Brunetta PG: Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase ii/iii systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010, 62: 222-233. 10.1002/art.27233.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  88. Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzova D, Sanchez- Guerrero J, Schwarting A, Merrill JT, Chatham WW, Stohl W, Ginzler EM, Hough DR, Zhong ZJ, Freimuth W, van Vollenhoven RF, BLISS-76 Study Group: A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011, 63: 3918-3930. 10.1002/art.30613.

    Article  CAS  PubMed  Google Scholar 

  89. Navarra SV, Guzman RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, Li EK, Thomas M, Kim HY, Leon MG, Tanasescu C, Nasanov E, Lan JL, Pineda L, Zhong ZJ, Freimuth W, Petri MA, BLISS-52 Study Group: Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011, 377: 721-731. 10.1016/S0140-6736(10)61354-2.

    Article  CAS  PubMed  Google Scholar 

  90. Guidance for Industry, Systemic Lupus Erythematosus - Developing Medical Products for Treatment. [http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm072063.pdf]

  91. Petri M, Buyon J, Kim M: Classification and definition of major flares in SLE clinical trials. Lupus. 1999, 8: 685-691. 10.1191/096120399680411281.

    Article  CAS  PubMed  Google Scholar 

  92. Wollaston SJ, Farewell VT, Isenberg DA, Gordon C, Merrill JT, Petri MA, Kalunian KC: Defining response in systemic lupus erythematosus: a study by the Systemic Lupus International Collaborating Clinics group. J Rheumatol. 2004, 31: 2390-2394.

    PubMed  Google Scholar 

  93. Furie RA, Petri MA, Wallace DJ, Ginzler EM, Merrill JT, Stohl W, Chatham WW, Strand V, Weinstein A, Chevrier MR, Zhong ZJ, Freimuth WW: Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum. 2009, 61: 1143-1151. 10.1002/art.24698.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  94. Hahn BH: Targeted therapies in systemic lupus erythematosus: successes, failures and future. Ann Rheum Dis. 2011, 70 (Suppl 1): i64-i66. 10.1136/ard.2010.142208.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michelle Petri.

Additional information

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lateef, A., Petri, M. Unmet medical needs in systemic lupus erythematosus. Arthritis Res Ther 14 (Suppl 4), S4 (2012). https://doi.org/10.1186/ar3919

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/ar3919

Keywords