Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 1.
Published in final edited form as: J Autoimmun. 2018 Nov 16;96:1–13. doi: 10.1016/j.jaut.2018.11.001

Systemic Lupus Erythematosus: Diagnosis and Clinical Management

Andrea Fava 1, Michelle Petri 1,*
PMCID: PMC6310637  NIHMSID: NIHMS1512848  PMID: 30448290

Abstract

Systemic lupus erythematosus (SLE) is a worldwide chronic autoimmune disease which may affect every organ and tissue. Genetic predisposition, environmental triggers, and the hormonal milieu, interplay in disease development and activity. Clinical manifestations and the pattern of organ involvement are widely heterogenous, reflecting the complex mosaic of disrupted molecular pathways converging into the SLE clinical phenotype. The SLE complex pathogenesis involves multiple cellular components of the innate and immune systems, presence of autoantibodies and immunocomplexes, engagement of the complement system, dysregulation of several cytokines including type I interferons, and disruption of the clearance of nucleic acids after cell death. Use of immunomodulators and immunosuppression has altered the natural course of SLE. In addition, morbidity and mortality in SLE not only derive from direct immune mediated tissue damage but also from SLE and treatment associated complications such as accelerated coronary artery disease and increased infection risk.

Here, we review the diagnostic approach as well as the etiopathogenetic rationale and clinical evidence for the management of SLE. This includes 1) lifestyle changes such as avoidance of ultraviolet light; 2) prevention of comorbidities including coronary artery disease, osteoporosis, infections, and drug toxicities; 3) use of immunomodulators (i.e. hydroxychloroquine and vitamin D); and 4) immunosuppressants and targeted therapy. We also review new upcoming agents and regimens currently under study.

Keywords: systemic lupus erythematosus, diagnosis, management

1. Introduction

Systemic lupus erythematosus (SLE) is the quintessential autoimmune disease. A complex interaction of impaired apoptotic clearance, upregulation of innate and adaptive immune system, complement activation, immune complexes, and tissue inflammation culminates in a self-sustained autoimmune process. Multiple pathogenic mechanisms likely can converge toward the clinical phenotypes that we call SLE. In fact, while many organs and tissues may be affected by SLE, the pattern of clinical manifestations and autoimmune phenomena is heterogenous among patients and even changes over time in individual patients. For this reason, diagnosis is often difficult or delayed and relies on keen clinical expertise to combine clinical and immunological findings. Here, we review classification criteria and current and future treatments with a mechanistic and evidence-based point of view.

2. Epidemiology

Lupus is a worldwide disease with a striking predilection for women of childbearing age. In women between the age of 15 and 44 years, the female to male ratio is up to 13:1 while it is only 2:1 in children and in the elderly [13]. While present across ethnicities, it is more prevalent in non-Caucasians. While prevalence in Europe and United States is higher in people of African descent, SLE is infrequent in Africa [4,5]. In the United States, it is more common in African-Americans, who also tend to have worse outcomes. In fact, African-American women are about 3 times more likely to have lupus and suffer greater mortality compared to Caucasian-Americans [6]. The Centers for Disease Control and Prevention report an estimated prevalence of about 322,000 cases of probable or definite SLE, higher in African Americans, American Indians and Alaska Natives [3,710].

3. Pathogenesis overview

The clinical onset of SLE requires an interaction of genetic predisposition, environmental precipitants, immunological and hormonal factors. In such a permissive environment, along with proinflammatory stimuli such as type 1 interferons and other cytokines, immune tolerance to self-antigens is lost [11]. Autoimmunity then follows driven by a complex interplay of defective clearance of apoptotic waste and immune complexes along with neutrophil extracellular traps, sensing of nucleic acids, disrupted lymphocyte biology, and interferon pathways [11].

The genetic susceptibility is suggested by the 11–50% monozygotic twin concordance and increased risk in families [12]. Many genes have been associated with a predisposition to develop lupus, typically encoding for immune components including HLA, IRF5, ITGAM, STAT4, BLK and CTLA4 among others [12,13].

Many environmental triggers have been implicated in lupus. Ultraviolet light (the most recognized), drugs/supplements (echinacea, trimethoprim/sulfamethoxazole) [1416], smoking [17], infections [18,19] (Epstein-Barr virus in particular [20]), silica [21], mercury [22] and others [19,23,24]. Psychological stress has also been linked to a 50% increase risk of developing lupus [25,26].

In lupus, there is evidence for disruption of both the innate and adaptive arms of the immune system, connected in a feedback loop. T cells are defective and their aberration in lupus is complex [11,27,28]. They fail to produce enough IL-2 and there is a polarization toward Th17 over Tregs. There is an excess of double-negative T lymphocytes. T cells in SLE provide excessive help to B cells. There is evidence of an excess of autoreactive B cells but with overall B cell lymphopenia. The hyperactivation of the BLyS or BAFF pathway, a T cell independent B cell survival pathway, led to the development of a new biologic therapy, as described later.

In addition, the identification of a strong type 1 interferon “signature” in lupus recognized the pivotal role of the innate immune system [2931]. Dendritic cells are central in the production of type 1 interferon and have a role in the clearance and sensing of nucleic acids and immune complexes, known autoantigens in lupus. In fact, endogenous and external nucleic acids are a major antigenic stimulus in lupus. Autoantibodies targeting nucleic acid-bound antigens are one of the hallmarks of the disease. The major source of such antigens come from apoptosis and neutrophils extracellular traps (NETs) [3234]. Excess and impaired degradation of NETs are associated with lupus severity, lupus nephritis, anti-dsDNA antibodies and complement consumption [35,36].

Apoptosis is pivotal in the pathogenesis of lupus. Apoptotic cells such as UV-light exposed keratinocytes release blebs rich in autoantigens including Ro, La, and RNP [37,38]. In in vivo photosensitivity for example, immunocomplexes with endogenous nucleic acids can activate plasmacytoid dendritic cells via activation of TLR7 and 9 to produce conspicuous amounts of type 1 interferon [39,40]. These particles are physiologically coated by molecules such as C1q and IgM to facilitate phagocytosis and clearance in an “immunologically silent” and anti-inflammatory environment as mediated by dendritic cells [41,42]. Excessive non-cleared apoptotic waste is associated with inflammation and autoantibody production. For example, deficiency of C1q leads to early onset SLE in children [43].

The importance of this process is further substantiated by the role of DNAse1. DNAse1 is a nuclease involved in the processing of the nucleic acids exposed on apoptotic blebs. In the presence of DNAse1, apoptotic blebs tend to induce tolerance to self-DNA. When mutated or inhibited by anti-nucleic acids antibodies, the unprocessed nucleic acids trigger autoantibody production, type I interferon secretion, and provide a substrate for anti-dsDNA mediated immune complexes [35,44]. In fact, rare mutations in single genes involved in DNA damage repair, apoptosis, clearance of self-antigens, nucleic acid sensing, type 1 interferon production, and early complement components can lead to “monogenic lupus” and “interferonopathies” [45]. These observations highlighted the crucial role of the hypersensitivity to and inability to manage nucleic acids from dying cells as well as immunocomplexes, leading to the development of a “waste disposal” theory.

4. Diagnosis

The diagnosis of systemic lupus erythematosus is made based on a combination of typical clinical manifestations and positive serologies. Given the wide heterogeneity of clinical manifestations, several sets of classification criteria have been developed over time for epidemiological and research purposes. However, some, such as the SLICC classification criteria, which are more sensitive, and therefore particularly useful in early diagnosis, can be useful as a diagnostic framework to confirm clinical judgment.

4.1. Classification criteria

The 1982 ACR criteria, revised in 1997, have been widely used for more than 3 decades [46,47]. The 2012 Systemic Lupus International Collaborating Clinics (SLICC) criteria [48], were evidence-based, included a “stand alone” criterion of lupus nephritis, and required at least 1 clinical (acute cutaneous lupus, chronic cutaneous lupus, oral or nasal ulcers, synovitis, serositis, proteinuria or red blood cell casts, neurologic manifestations, hemolytic anemia, leukopenia or lymphopenia, and thrombocytopenia) and at least 1 immunologic criterion (ANA, anti-dsDNA, anti-Smith, anti-phospholipid antibodies, hypocomplementemia, and direct Coombs test) for a total of 4. The SLICC classification criteria are not limited to research and are widely used for diagnosis, as they are more sensitive and more comprehensive. The 2018 EULAR criteria, developed for research only, require an ANA of 1:80 or higher, and then weight lupus manifestations (for a score of 10) [49]. Direct comparison of the 3 sets of criteria using the SLICC validation cohort showed that they perform similarly with sensitivity and specificity of 89% and 90% for the EULAR, 83% and 96% for the ACR, and 97% and 84% for the SLICC criteria, respectively [50]. A systematic review and meta-analysis including 5236 patients and 1313 controls showed that the SLICC criteria have better sensitivity (94.6% vs 89.6%) and similar specificity (95.5% vs 98.1%) for adult SLE as compared to the 1997 ACR criteria [51].

4.2. Clinical diagnosis

This section describes key organ manifestations, including some recent developments, that are important in diagnosing lupus. Additional information and reviews of each clinical manifestation of lupus can be found elsewhere [52,53].

4.2.1. Cutaneous lupus

Skin involvement in SLE occurs in almost 90% of patients and includes lupus-specific manifestations such as acute cutaneous lupus, subacute cutaneous lupus, and chronic cutaneous lupus (discoid lupus, lupus profundus, chilblain lupus, and lupus tumidus. Non-lupus-specific manifestations include alopecia, vasculitis, livedo reticularis, periungual telangiectasias, and Raynaud’s phenomenon [54,55]. Most forms of cutaneous lupus share similar histological findings such as interface dermatitis with perivascular and periadnexial inflammation and may have immunoglobulin and complement deposition at the dermo-epidermal junction [55]. A biopsy is often key in the diagnosis of cutaneous lupus.

A key characteristic of cutaneous lupus erythematosus is the photosensitive distribution. Cutaneous lupus erythematosus must be differentiated from other more common photosensitive rashes such as polymorphous light eruption or rosacea. More than 50% of “photosensitive” biopsied rashes in SLE patients end up showing a non-lupus causation such as non-specific inflammatory reactions or polymorphous light eruption [56]. A true photosensitive rash in lupus is raised, delayed and long lasting. It typically occurs days after ultraviolet light exposure [57,58], tends to last for more than 3 weeks [56], and may be associated with systemic symptoms such as arthralgia or fatigue [59].

Alopecia is common in SLE, but can have multiple causes. True lupus alopecia or “lupus hair” is reversible and characterized by shortening of the frontal hair which is irregular and with broken hairs at 5–25mm of length [60]. Discoid lupus can lead to permanent scarring lupus. Smoking is a risk factor for SLE [17], increases the risk of discoid lupus [61,62], increases ongoing cutaneous activity [63] and impairs the benefit of hydroxychloroquine [64].

4.2.2. Musculoskeletal involvement

Arthralgia and true synovitis are very common in SLE occurring in almost 90% [65]. It most typically presents as a symmetric polyarthritis involving the metacarpophalangeal, proximal interphalangeal, and knee joints [66]. Monoarthritis should prompt evaluation for alternative causes. Erosions are rare (unless sensitive imaging is used) and are associated with anti-cyclic citrullinated peptides antibodies [67]. Periarticular involvement including tendons and joint capsule is more common than previously understood (discovered by ultrasound and MRI) and can lead to reducible deformities known as Jaccoud’s arthropathy [6870]. Articular ultrasound and magnetic resonance may help to differentiate active inflammatory disease from tenderness due to fibromyalgia and quantify the burden of disease [6870].

Importantly, fibromyalgia, fibromyalgianess (the tendency to respond to illness and psychosocial stress with fatigue and widespread pain), and depression are common in lupus patients and associate with musculoskeletal pain; these do not track with disease activity and should not be misinterpreted as such [71,72].

4.2.3. Renal disease

Renal involvement is present in about 50% of patients with lupus with a predilection for certain ethnic groups such as African-Americans (70%) [73]. Early detection and treatment are paramount since lupus nephritis is a major cause of morbidity and mortality in SLE and delayed diagnosis is a risk factor for end-stage renal disease [74,75]. Renal disease is suspected when there is proteinuria [52,75]. However, lupus nephritis (class III, IV, and V) can be present in 25% of SLE patients without clinical signs of renal disease [76]. Urine protein levels above 500 mg/24h are associated with histopathological lupus nephritis and should prompt a renal biopsy [77]. Urine protein-to-creatinine ratio is a reliable measure of proteinuria in lupus nephritis, it is easier to routinely measure, and it is highly correlated with the 24h proteinuria [78]. In addition, clinical and serological (hypocomplementemia and elevation of anti-dsDNA antibodies) activity may further suggest lupus nephritis and is associated with higher risk histological features [77,79]. Anti-C1q antibodies are strongly associated with renal involvement in SLE, may predict flares, and their absence has a negative predictive value of nearly 100% [8082].

Renal biopsy is essential to confirm the diagnosis, exclude alternative causes, evaluate for active inflammation versus irreversible damage, inform prognosis, and guide treatment. Lupus nephritis is classified based on clinicopathological correlations [83]. The Renal Pathology Society/International Society of Nephrology (or RPS/ISN) classification include 6 classes: minimal mesangial lupus nephritis, mesangial proliferative lupus nephritis, focal lupus nephritis, diffuse lupus nephritis, membranous nephropathy, and advanced sclerosing lupus nephritis [83]. Additional renal diseases associated with lupus include lupus podocytopathy [84], tubulointerstitial nephritis, vascular disease (such as thrombotic microangiopathy, vasculitis, or atherosclerosis) [85], and collapsing glomerular sclerosis [86].

4.2.4. Central nervous system disease

A wide array of neuropsychiatric manifestations has been associated with SLE. However, only a few of them are more specific for SLE and are helpful for diagnosis [87,88]. These include seizures, psychosis, mononeuritis multiplex, myelitis, peripheral or cranial neuropathy, and acute confusional state [87,88]. Importantly, these require exclusion of other known causes.

In addition to clinical evaluation, magnetic resonance imaging (MRI) and cerebral spinal fluid analysis are important diagnostic tools. Central nervous system MRI is helpful to detect chronic microvascular changes, infarcts, hemorrhages, cortical atrophy, edema, abscesses, transverse and longitudinal myelitis [89,90]. Detection of long T2 of the gray matter (suggesting edema) or gadolinium enhancement in patients with active manifestations (seizures, psychosis, coma) may suggest activity, but generally, MRI cannot distinguish active from previous disease [89,91,92]. Identification of abnormal cerebrospinal fluid IgG and oligoclonal bands may help to identify active neuropsychiatric lupus [93]. Infection, metabolic-toxic and malignancy conditions always need to be considered in the differential diagnosis.

Up to 80% of SLE patients have cognitive impairment [94]. Formal psychometric testing and a psychiatric evaluation may be helpful to detect cognitive impairment and/or functional disorders [94,95]. Surprisingly, cognitive impairment (mild to moderate) is present in the majority of patient at the time of diagnosis, and does not worsen during follow up [96]. The most important correlate of cognitive impairment is depression [97].

5. Management

5.1. Principles

The goals of treatment in lupus are 1) maintain lowest degree of activity using immunomodulators, immunosuppression as appropriate and avoiding known triggers, 2) prevent organ damage from active lupus, 3) reduce comorbidities secondary to lupus and its treatment, especially accelerated atherosclerosis, the major cause of death, and 4) address fatigue and pain, which often are not associated with active lupus. Early initiation of treatment as well as partnership with the patient towards these shared goals is essential. This translates into avoidance of known triggers of flares, the need for sun protection, maximization of immunomodulators (hydroxychloroquine and vitamin D, including monitoring for adherence), avoidance of maintenance prednisone >6mg daily, and control of active disease with immunosuppression or biologics when required. Here, we review the rationale for current and future treatments.

5.2. Immunomodulators

Immunomodulators can favorably regulate the immune system in SLE without increasing the risk of infection or malignancy.

5.2.1. Hydroxychloroquine

Hydroxychloroquine pleiotropically modulates the immune response by inhibition of B cell receptor and TLR signaling as well as intracellular TLR-3 and-7 activation, fundamental in nucleic acid sensing [98101]. It increases the lysosomal pH interfering with MHC-antigen binding, thus processing of autoantigens, as well as secretions of cytokines [102104]. Hydroxychloroquine exerts an anti-type 1 interferon effect by interfering with the STING pathway [105].

Hydroxychloroquine is the cornerstone of medical therapy in lupus. It should be used in every patient unless there is a clear contraindication. It is the only medication shown to increase survival in lupus patients [106109]. It has been shown to reduce lupus flares [110], prevent organ damage [111] including cardiovascular events [112], triple mycophenolate response in lupus nephritis [113], prevent seizures [114,115] and reduces the risk of developing neuropsychiatric lupus [116]. Hydroxychloroquine improves skin manifestations [117,118] and arthritis [119]. Hydroxychloroquine has a favorable effect on lipids [102,120122], reduces insulin resistance [123] and the risk of thrombosis [124,125], and has a favorable effect on bone density [108,126].

Hydroxychloroquine is non immunosuppressive and does not increase the risk of infection or malignancy [127,128]. Retinal toxicity is a rare complication increasing after 20 years duration of treatment [129]. Retinal screening occurs at baseline, at 5 years, and then yearly [130]. Optical coherence tomography is the preferred screening test [36]. Hyperpigmentation can occur. Very rare complications include cardiomegaly and myopathy.

5.2.2. Vitamin D

Vitamin D should be supplemented in all SLE patients with insufficiency or deficiency, for its immunomodulatory and anti-fibrotic effects. Vitamin D immunomodulatory properties are mediated by the vitamin D3 receptor (VDR) in multiple immune cells lineages including monocytes, dendritic cells, and activated T cells as well as in the skin, vasculature and other tissues [131]. In vitro, vitamin D exerts an anti-inflammatory and anti-proliferative effect by promoting a Th1 (TNF-α, IL-2, IFN-γ) to Th2 (IL-4, IL-5, IL-10, GATA3) polarization as well as Th17 (IL12, IL23, IL-6, 17) to Treg (IL-10, TGF-β, FoxP3, CTLA4) state [132]. It affects the development and function of NKT cells [133].

In addition, vitamin D may act as an anti-fibrotic agent. Vitamin D deficiency is associated with increased risk of multiorgan fibrosis including, among others, the kidneys and the lungs [134]. Importantly, patients with lupus nephritis refractory to mycophenolate have increased expression of profibrotic pathways in the affected kidneys [135] suggesting that renal tissue could be rescued by targeting such pathways. Vitamin D supplementation prevented fibrosis in animal models and inhibits pro-fibrotic pathways mediated by TGF-beta and Ras [134,136].

Vitamin D deficiency is common in patients with SLE [137]. Some VDR polymorphisms are associated with lower serum vitamin D levels and have been associated with SLE [138]. In patients with lupus, vitamin D deficiency correlates with increased disease activity and fatigue [139,140] as well as an increased risk for thrombosis, including from antiphospholipid antibodies [141,142]. Importantly, supplementation of vitamin D is associated with reduction of proteinuria, higher complement levels, and improvement in global disease activity in SLE as demonstrated in an observational cohort and in a randomized controlled study [143,144]. Supplementation should be aimed to a 25(OH) vitamin D level of 40 ng/ml [143,144]. Vitamin D supplementation is safe [143] and should be continued indefinitely. Vitamin D levels should be monitored periodically to assess adequate absorption and dosing and adherence.

5.2.3. Dehydroeipandrosterone (DHEA)

DHEA is an adrenal hormone regulated by ACTH [145]. It is an important precursor of both estrogens and androgens via peripheral conversion [146]. Women with lupus tend to have lower levels of androgens, higher estradiol, lower DHEA and DHEA-S (its metabolite), independently of corticosteroid use [147,148]. In addition, DHEA supplementation has been associated with regulation of proinflammatory cytokines (IL-2, IL-1, IL-6, TNF-a) and may reduce antibody production in mice [149152].

Many of the several randomized clinical trials in women with SLE showed a modest improvement in disease activity along with improvement in cytokine profile and bone density [153,154]. DHEA should not be used in postmenopausal women since it may increase the risk of hormone sensitive malignancies. There is no evidence to support DHEA use in men.

5.3. Corticosteroids

Corticosteroids affect all components of the immune system [155]. High dose or “pulsed” corticosteroids are important to rapidly ablate the autoimmune response in life or organ threatening manifestations such as some cases of nephritis, vasculitis, central nervous system lupus, myocarditis, or alveolitis, among others. In lupus nephritis for example, pulsed therapy (250–1000mg IV daily for 3 days) was previously recommended along with cyclophosphamide or mycophenolate for induction, but there is no consensus on an oral maintenance regimen [156,157]. The “rituxilup” protocol showed that lupus nephritis remission can be induced without any oral corticosteroids using rituximab and mycophenolate suggesting that corticosteroids might not be necessary to control even severe lupus manifestations [158]. In the recent voclosporin phase 2 clinical trial, only 25 mg of prednisone was used [159]. Importantly, any of the studied immunosuppressants agents (cyclophosphamide, azathioprine, mycophenolate, tacrolimus) is better than corticosteroids alone to prevent end stage renal disease [160].

Oral corticosteroids should be avoided as much as possible. In lupus patients, 80% of organ damage after diagnosis is directly or indirectly attributable to prednisone [161]. Doses of even 10 to 20 mgs daily increase the risk of cardiovascular events [162] and any dose above 6 mg increases later organ damage by 50% [163]. Even low doses, over time, increase cataracts, osteoporosis, fractures and coronary artery disease [164].

Intramuscular triamcinolone, or a brief 1-week methylprednisolone dose pack, is effective for management of most mild to moderate flares. Results from the FLOAT trial showed that a single intramuscular triamcinolone 100 mg injection is a faster acting and effective alternative to oral maintenance corticosteroids [165]. As it is released slowly, its effect lasts for about 1 month and has equipotency of approximately 2 mg of prednisone daily.

5.4. Cytotoxic-immunosuppressants

5.4.1. Cyclophosphamide

Cyclophosphamide is a highly toxic alkylating agent that depletes T and B cells and suppresses antibody production [166]. It was more widely used in the past for the induction and maintenance of lupus nephritis [167,168] and other severe lupus manifestations such as central nervous system lupus [169]. However, it has now been largely replaced by less toxic immunosuppressive medications such as mycophenolate, calcineurin inhibitors, and azathioprine for nephritis and rituximab for severe central nervous system lupus [170,171]. Cyclophosphamide is associated with premature ovarian failure, hemorrhagic cystitis, increased risk of bladder and other malignancies, and leukopenia along with an increased risk of infections [172].

5.4.2. Azathioprine

Azathioprine is a purine analogue. It is converted in vivo to 6-mercaptopurine followed by thioinosinic acid and 6-thioguanine which are incorporated into DNA and RNA, inhibiting their synthesis. Besides its antimetabolite role, azathioprine may have a tolerogenic effect by inhibiting CD28-mediated signal 2 in T cells [173].

Azathioprine has been commonly used in renal and extrarenal lupus since the late 1960s. In two small randomized studies, azathioprine compared to corticosteroids alone was shown to reduce mortality, rate of flares and corticosteroid use, including patients with severe renal or central nervous system disease [174,175]. In the following decades, its use in induction in lupus nephritis waned given its inferiority to cyclophosphamide [176,177]. Azathioprine was inferior to mycophenolate in the ALMS trials and equal in the MAINTAIN trial [178,179].

In extrarenal lupus, azathioprine is widely used as a corticosteroid sparing agent [180]. However, a recent non-blinded randomized controlled study showed that mycophenolate was superior to azathioprine to control disease activity and prevent flares (renal and extrarenal) while maintaining a similar side effect profile [181].

Azathioprine remains an excellent choice to control renal and extrarenal disease during pregnancy as the metabolite 6-MMP is not generated in the fetus [182,183].

5.4.3. Methotrexate

Methotrexate is an antimetabolite the interferes with DNA synthesis, repair, and replication by irreversibly binding to dihydrofolate reductase, thus reducing purine synthesis. However, the mechanism of its anti-inflammatory effects goes beyond arresting the cell cycle by folate depletion and is not completely understood [184]. For instance, co-administration of folate does not impair its efficacy, while mitigating side effects. Low-dose methotrexate has pleiotropic effects involving increased anti-inflammatory adenosine signaling, apoptosis of activated lymphocytes, reduction of circulating pro-inflammatory T-cells, reduction of adhesion molecules on endothelial and synovial cells, reactive oxygen species, and others [184].

In lupus, methotrexate has been used since the 1960s [185]. Combined evidence from 3 small randomized trials and several observational studies showed that methotrexate reduced disease activity, was corticosteroid sparing, had efficacy for joint and skin disease, and ameliorated anti-dsDNA and complement levels [186]. Methotrexate showed a modest steroid sparing activity in a randomized controlled trial [187].

5.4.4. Mycophenolate

Mycophenolate preferentially depletes guanoside nucleotides in T and B cells inhibiting proliferation. It suppresses lymphocyte and monocyte recruitment to inflamed tissue. It inhibits inducible nitric oxide synthase which may curtail nitric oxide oxidative tissue damage mediated by macrophages [188].

Mycophenolate is effective for induction and maintenance of lupus nephritis [178,179,189191]. The ALMS trial (n=140) showed that 22.5% of patients treated with mycophenolate achieved complete renal remission at 24 weeks compared to 5.8% in the cyclophosphamide group [190]. The larger ALMS lupus nephritis induction trial showed similar efficacy of mycophenolate compared to cyclophosphamide [191]. Mycophenolate had a better safety profile. This was confirmed by a recent Cochrane systematic review, although with low certainty evidence [192]. This review also confirmed that mycophenolate is superior to azathioprine in preventing nephritis relapses. Mycophenolate is also effective in extrarenal lupus in all the analyzed domains in multiple case series and in a post-hoc analysis of the ALMS trial [193,194].

5.4.5. Calcineurin inhibitors

Calcineurin inhibitors target T cells by blocking the inhibition of calcineurin [195]. This prevents translocation of transcription factors such as nuclear factor of activated T-cells (NFAT) resulting in T cells inhibition with reduction of IL-1b, IFN-γ, IL-6 and IL-10. B cell activation is also impaired along with class switching and immunoglobulin production. Furthermore, calcineurin inhibitors affect the kidneys directly by stabilizing podocytes, reducing mesangial proliferation, and improving proteinuria [196198].

Calcineurin inhibitors are commonly used to prevent transplant rejection. In lupus, tacrolimus has been used, alone or in combination with mycophenolate, more extensively than cyclosporine given its better side effects profile [199]. However, both have significant variability in plasma concentration and require monitoring.

Multiple small RCTs showed that lupus nephritis induction with calcineurin inhibitors (cyclosporine or tacrolimus) is as effective as cyclophosphamide or mycophenolate [200203]. A recent metanalysis suggested a possible superiority of tacrolimus over cyclophosphamide [204]. A larger trial comparing mycophenolate vs tacrolimus in 150 Chinese patients with active class III/IV showed similar complete response rates at 6 months (59% vs 62%, respectively) and side effects [205].

Combination of calcineurin inhibitors and mycophenolate is used to prevent graft rejection and is also effective in refractory lupus nephritis in Caucasian and African American patients [206208]. In a large RCT, 368 Chinese patients with class III/IV/V lupus nephritis were randomized to receive a combination of tacrolimus (4 mg/day) and low dose mycophenolate (1 g/day) (“multitarget”) or monthly IV cyclophosphamide (0.5–1 g/m2) [209]. After 6 months, complete remission was achieved in 45.9% vs 25.6%, and overall response in 83.5% vs 63% in the multitarget and cyclophosphamide, respectively. More patients withdrew from the multitarget arm, mostly due to pneumonias and zoster reactivation [209].

Calcineurin inhibitors have also been proven effective for maintenance therapy of lupus nephritis [210,211], as well as for pure membranous lupus nephritis with a significant antiproteinuric effect [205,212215]. They are recommended by EULAR [157].

Voclosporin is a new calcineurin inhibitor. It has greater pharmacological potency, faster elimination and less variability in blood concentration. It is non-inferior to tacrolimus in preventing kidney transplant rejection [216]. The AURA phase IIb study randomized 265 patients with active lupus nephritis to receive voclosporin or placebo in addition to mycophenolate and corticosteroids [159,217]. Both voclosporin doses showed higher complete remission rate than mycophenolate alone at 24 and 48 weeks (48 weeks complete remission 40%, 49%, and 24% in the high dose, low dose, and mycophenolate alone arm, respectively). Glomerular filtration rate decreased with voclosporin. There was an imbalance of death in the low dose voclosporin arm. A phase III trial (AURORA) is ongoing.

5.5. Biologics and small molecules

5.5.1. Available

5.5.1.1. Rituximab

Autoantibodies and immune complex formation are an immunological hallmark of lupus. B cells have been targeted in SLE for years with immunosuppressants such as cyclophosphamide and mycophenolate. Targeted therapies toward B cells that avoid broad immunosuppression are the goal.

Rituximab is an anti-CD20 monoclonal antibody that leads to peripheral B cell depletion. Several observational studies showed benefit in renal and non-renal lupus [218,219]. Two large randomized placebo-controlled trials in renal and non-renal lupus failed to meet their primary outcomes. In the EXPLORER non renal trial [220], the primary (proportion of patients achieving complete or partial remission based on BILAG score) and secondary endpoints were not met. B-cell depletion was obtained in about 90% of treated patients. Rituximab did reduce anti-dsDNA titers and improved C3 and C4 levels.

In the LUNAR renal trial [221], 144 patients with class III or IV lupus nephritis were randomized to receive rituximab (1,000 mg on day 1, 15, 168, and 182) or placebo in addition to mycophenolate and corticosteroids. The rituximab group failed to achieve the primary outcome defined as better overall (complete and partial) renal response compared to mycophenolate alone at 52 weeks (57% vs 46%, p = 0.18). Further analysis showed that rituximab did reduce proteinuria better than mycophenolate alone at 18 months and reduced the need for rescue cyclophosphamide (8/72 in the placebo group vs 0/72 for rituximab).

The disappointing results led to the consideration of the issue of reconstitution of B cells after rituximab [222]. The surge of BAFF upon depletion of B cells with rituximab may contribute to the lack of sustained response to rituximab [223225]. This provided the rationale for SynBioSe, a proof of concept phase 2A trial of rituximab followed by belimumab in patients with refractory severe lupus nephritis [226]. Eleven out of 16 patients showed a renal response, 5 with complete response. The renal response was paralleled by amelioration of immune-complex mediated NETs formation. In the CALIBRATE trial, patients with refractory lupus nephritis were treated with rituximab, cyclophosphamide and corticosteroids followed by either belimumab or placebo. This did not show a benefit of belimumab, with 24 week renal responses of 24% vs 23%. [227].

Evidence for the efficacy of rituximab comes from “rituxilup”, an oral corticosteroid-avoidance protocol to treat lupus nephritis with mycophenolate and rituximab. In this prospective observational single-center cohort study, partial or complete remission was achieved in 45 of 50 patients by a median time of 37 weeks [158].

5.5.1.2. Belimumab

B cells maturation, Ig-class switching, and antibody production are most potently driven by antigen specific T cells. Since T cells tolerance is much more strictly regulated, such T cell dependence should ensure that autoreactive naïve B cells that escaped central tolerance will be eliminated by lack of stimulation. However, T cell-independent pathways such as B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL) may bypass T cells in the selection of autoreactive B cells. BAFF exists in a soluble and membrane bound form and is produced by antigen presenting cells (dendritic cells, B-cells, monocyte/macrophages, plasmacytoid dendritic cells), neutrophils, activated T cells and endothelial cells [228]. BAFF provides essential activation and survival signals via NFkB and MAP-K pathways to B cells as mediated by 3 receptors: BAFFR, TACI and BCMA. BAFFR ligation provides the strongest signal while TACI and BCMA also bind to APRIL [229]. Importantly, patients with lupus have higher circulating BAFF and APRIL levels and the BAFF level is predictive of flares [230]. BAFF-transgenic mice have a lupus-like phenotype independent of T cells [231].

Two large phase 3 RCT, BLISS-52 and BLISS-76, assessed the SLE responder index at 52 weeks in patients with non-renal lupus [232,233]. In both trials, there was a higher response rate with the higher dose of belimumab (10 mg/kg) compared to standard of care (BLISS-52: n=867, 58% vs 44%, p=0.006; BLISS-76: n=819, 43.2% vs 33.5%, p=0.017). Belimumab also significantly reduced the number of flares. In addition, belimumab normalized anti-dsDNA in up to 17–25% (placebo 6–8%) and normalized C3 in 34–44% (vs 14–21%) and C4 in 43–46% (vs 17–19%). Belimumab decreased circulating B cells and CD20+/CD27bright short lived plasma cells were decreased by >50% and up to 43%, respectively. Pooled data analysis of both trials showed that combined hypocomplementemia and anti-dsDNA positivity was the best predictor of response to belimumab [234]. Based on these results, belimumab became the first new drug in the last five decades to be approved by the EMA and FDA (and later by NICE) for the treatment of active non-renal lupus despite standard therapy.

5.5.2. Currently studied

5.5.2.1. Anifrolumab

Type 1 interferon signaling is mediated by the type I IFN-α/β/ω receptor (IFNAR). Anifrolumab is a monoclonal antibody blocking IFNAR. In a phase 2b trial, 305 lupus patients were randomized to receive placebo or one of two dosages of anifrolumab [235]. At 24 weeks, 34% and 29% of patients receiving anifrolumab (300mg and 1000mg every 4 weeks, respectively), while only 17.6% in the placebo group, achieved the primary outcome of SRI-4 response with sustained reduction of oral corticosteroids. The effect was greater in patients with the interferon signature at baseline. Both skin and joint disease showed a favorable response. In addition, anifrolumab was associated with decreased anti-dsDNA titers and higher C3 levels. There was a mild increased risk of viral infections, including influenza and herpes zoster. However, the first phase III trial (TULIP 1) did not reach its primary end point of decreasing the SRI-4 [236]. TULIP 2 is currently under way.

5.5.2.2. Ustekinumab

There is increased Th17 activity in lupus. Serum IL-17 and IL-23 levels are higher in SLE and correlate with disease activity [28]. IL-17-producing cells are present in biopsies from lupus nephritis [28]. Double negative T cells are also a source of IL-17. STAT3, which is downstream to IL-23 stimulation, is upregulated in lupus [237], and promotes IL-17 production as well as differentiation toward Th17 and Tfh [238]. Tfh are expanded in SLE and have been implicated in the overstimulation of B cells [239]. IL-23 promotes the development of double negative T cells [240] and impairs the production of IL-2 suggesting a possible indirect effect on the production of Tregs [241].

The IL-23/Th17 axis can be disrupted by ustekinumab, a monoclonal antibody blocking IL12 and IL-23 currently approved to treat psoriasis, psoriatic arthritis and inflammatory bowel disease. In a phase 2 trial, 102 patients with SLE were randomized (3:2) to ustekinumab vs placebo. At 24 weeks, 60% of ustekinumab-treated patients achieved the primary endpoint, SLE responder index-4 (SRI-4) compared to 31% in the standard of care group (p=0.0046). Subgroup analysis showed significant improvement in skin and joint scores. Ustekinumab improved C3 and reduced anti-dsDNA levels [242].

5.5.2.3. Baricitinib

The Janus kinases (JAKs) are a family of tyrosine kinases mediating intracellular signaling of several cytokines via the JAK-STAT pathway. Inhibition of a single JAK may lead to blocking the downstream effect of several cytokines at the time. However, this is a redundant system in which a group of cytokines may signal through different JAKs and different JAKs mediate signaling from different groups. At present, baricitinib and tofacitinib are FDA approved for the treatment of rheumatoid arthritis. Baricitinib is a reversible inhibitor of JAK1 and JAK 2. These mediate signaling for type 1 interferons, IFN-γ, IL-6, IL-12, and IL-23 among others [243]. An international, multicenter, double-blind, placebo-controlled phase 2 trial assessed the efficacy of baricitinib in patients with SLE. The primary outcome was the proportion of patients achieving resolution of rash or arthritis at 24 weeks, defined by SLEDAI-2K [244]. Three-hundred and fourteen patients with inadequate control despite standard of care were included. The primary outcome was achieved in a statistically significant higher proportion of patients treated with baricitinib 4 mg daily, but not 2 mg daily, as compared to placebo (67% vs 58% vs 53%, respectively). Most of the results were driven by the baricitinib effect on arthritis as no significant difference was noted on skin scores. The frequency of flares was also lower in the 4 mg group compared to placebo (33% vs 51%). There were more serious infections in the 4 mg arm (6%) compared to the 2 mg (2%) and placebo (1%). There was 1 deep venous thrombosis (1%) detected in the 4 mg arm.

5.5.2.4. Atacicept

Atacicept is a TACI-Ig fusion protein that inhibits B cells by dual inhibition of APRIL and BLyS. In a phase 1b trial, atacicept showed a dose dependent reduction in circulating B cells and immunoglobulins [245]. In the ADDRESS II, a phase 2b trial, 306 patients were randomized to receive weekly subcutaneous atacicept (75mg or 150mg) or placebo [246]. Atacicept was associated with a trend toward better SRI-4 response at 4 weeks compared to placebo, especially in individuals with high disease activity, serologically active disease, or both. For example, in the serologically active group, 62% of patients treated with atacicept achieved SRI-4 at 24 weeks compared to 24% in the placebo arm [246].

5.6. Lifestyle

Some of lupus management is non-pharmacological [247]. Patients should avoid sun exposure using protective garments and sunscreen of at least SPF 50 (as demonstrated in a randomized clinical trial [248]).

Fibromyalgia and “fibromyalgia-ness” (tendency to respond to illness and psychosocial stress with fatigue, general increase in symptoms, and widespread pain) is increased in SLE [249,250]. Regular exercise, stretching and can help to improve fatigue, cognitive dysfunction, and pain from fibromyalgia [251253].

5.7. Prevention of comorbidities

Lupus confers a 2.4-fold increase in all-cause mortality. The number one cause of death in lupus is cardiovascular events, followed by infections, and finally by renal and respiratory complications of lupus [74].

The risk of cardiovascular events is increased 2.66 fold [162]. Therefore, aggressive management of traditional (smoking, obesity, diabetes mellitus, hypertension, dyslipidemia) and lupus (lupus activity, antiphospholipid antibodies, homocysteinemia, excessive corticosteroid use) modifiable cardiovascular risk factors is paramount to prevent early death [254].

Homocysteinemia is present in 15% of patients [255] and has an independent association with cardiovascular risk [256258], renal injury and fibrosis [259,260], and is associated with higher prevalence of myocardial infarction and thrombosis in patients with antiphospholipid antibodies [261]. Hyperhomocysteinemia is an independent risk factor for cardiovascular disease in patients with lupus [255].

Infections are common in lupus, particularly from encapsulated bacteria [262]. Typical organisms are the most common causal agents but opportunistic bacterial, mycobacterial, protozoal, fungal and viral infection are also increased [263]. Pneumonia, especially from Streptococcus pneumoniae, is common in lupus and is associated with excess mortality [74,263]. In addition to the local immunization schedule, pneumococcal and influenza vaccination should be followed.

Osteoporosis and fragility fractures are higher in lupus [264266]. Notably, fracture risk calculators may underestimate in lupus [267]. Optimal bone health should be pursued recommending smoking cessation, optimal vitamin D levels, adequate dietary calcium intake (rather than supplementation [268]), weight bearing exercise, avoidance of corticosteroids, bone density screening according to risk and treatment with DHEA (never in men) and bisphosphonate as appropriate [154,267].

6. Future perspectives and personalized medicine

The more granular understanding of the molecular basis of lupus pathogenesis has led to several new promising treatments that are undergoing late phase clinical testing. These recent phase 2 trials underlined how targeting a specific pathway may elicit dramatically different responses in patient subgroups. Precise characterization of disease phenotypes based on molecular and clinical features is crucial to design personalized treatment. The Accelerated Medicine Partnership (AMP), for example, is an ongoing effort to identify the molecular pathways, at the single cell level, involved in lupus nephritis [269]. This may help to redefine the way we classify SLE and lupus nephritis and identify precise predictors of treatment response. We expect that the understanding of the heterogeneity of autoimmunity in lupus will lead to more effective and less toxic regimens in the future.

Highlights.

  • Classification criteria are a helpful diagnostic framework to confirm clinical judgement.

  • Hydroxychloroquine is the cornerstone of medical therapy in lupus.

  • Immunosuppression and targeted therapies are paramount for renal and severe extra-renal lupus manifestations.

  • New therapies targeting the Th17, BAFF, JAK/STAT, and calcineurin pathways showed positive phase 2b trial results.

Acknowledgments

Funding: The Hopkins Lupus Cohort is funded by NIH AR 69572

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: none

References

  • [1].Petri M, Epidemiology of systemic lupus erythematosus, Best Pract. Res. Clin. Rheumatol 16 (2002) 847–858. doi: 10.1053/berh.2002.0259. [DOI] [PubMed] [Google Scholar]
  • [2].Johnson AE, Gordon C, Palmer RG, Bacon PA, The prevalence and incidence of systemic lupus erythematosus in Birmingham, England: Relationship to ethnicity and country of birth, Arthritis Rheum. 38 (1995) 551–558. doi: 10.1002/art.1780380415. [DOI] [PubMed] [Google Scholar]
  • [3].Danchenko N, Satia J, Anthony M, Epidemiology of systematic lupus erythematosus: a comparison of worldwide disease burden, Lupus. 12 (2006) 308–318. http://lup.sagepub.com.proxygw.wrlc.org/content/15/5/308.full.pdf+html. [DOI] [PubMed] [Google Scholar]
  • [4].Symmons DPM, Occasional Series: Lupus Around the World Frequency of lupus in people of African origin, Lupus. 4 (1995) 176–178. doi: 10.1177/096120339500400303. [DOI] [PubMed] [Google Scholar]
  • [5].Pons-Estel GJ, Alarcón GS, Scofield L, Reinlib L, Cooper GS, Understanding the Epidemiology and Progression of Systemic Lupus Erythematosus, Semin. Arthritis Rheum 39 (2010) 257–268. doi: 10.1016/j.semarthrit.2008.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Krishnan E, Hubert HB, Ethnicity and mortality from systemic lupus erythematosus in the US, Ann. Rheum. Dis 65 (2006) 1500–1505. doi: 10.1136/ard.2005.040907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Ferucci ED, Johnston JM, Gaddy JR, Sumner L, Posever JO, Choromanski TL, Gordon C, Lim SS, Helmick CG, Prevalence and incidence of systemic lupus erythematosus in a population-based registry of American Indian and Alaska Native people, 2007–2009, Arthritis Rheumatol. 66 (2014) 2494–2502. doi: 10.1002/art.38720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Lim SS, Bayakly AR, Helmick CG, Gordon C, Easley KA, Drenkard C, The incidence and prevalence of systemic lupus erythematosus, 2002–2004: The Georgia lupus registry, Arthritis Rheumatol. 66 (2014) 357–368. doi: 10.1002/art.38239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Somers EC, Marder W, Cagnoli P, Lewis EE, DeGuire P, Gordon C, Helmick CG, Wang L, Wing JJ, Dhar JP, Leisen J, Shaltis D, McCune WJ, Population-based incidence and prevalence of systemic lupus erythematosus: The Michigan lupus epidemiology and surveillance program, Arthritis Rheumatol. 66 (2014) 369–378. doi: 10.1002/art.38238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, Kwoh CK, Liang MH, Kremers HM, Mayes MD, Merkel PA, Pillemer SR, Reveille JD, Stone JH, Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I, Arthritis Rheum. 58 (2008) 15–25. doi: 10.1002/art.23177. [DOI] [PubMed] [Google Scholar]
  • [11].Tsokos GC, Lo MS, Reis PC, Sullivan KE, New insights into the immunopathogenesis of systemic lupus erythematosus, Nat. Rev. Rheumatol 12 (2016) 716–730. doi: 10.1038/nrrheum.2016.186. [DOI] [PubMed] [Google Scholar]
  • [12].Generali E, Ceribelli A, Stazi MA, Selmi C, Lessons learned from twins in autoimmune and chronic inflammatory diseases, J. Autoimmun 83 (2017) 51–61. doi: 10.1016/j.jaut.2017.04.005. [DOI] [PubMed] [Google Scholar]
  • [13].Ghodke-Puranik Y, Niewold TB, Immunogenetics of systemic lupus erythematosus: A comprehensive review, J. Autoimmun 64 (2015) 125–136. doi: 10.1016/j.jaut.2015.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Burger RA, Torres AR, Warren RP, Caldwell VD, Hughes BG, Echinacea-induced cytokine production by human macrophages, Int. J. Immunopharmacol 19 (1997) 371–379. doi: 10.1016/S0192-0561(97)00061-1. [DOI] [PubMed] [Google Scholar]
  • [15].Spelman K, Burns JJ, Nichols D, Winters N, Ottersberg S, Tenborg M, Modulation of cytokine expression by traditional medicines: A review of herbal immunomodulators, Altern. Med. Rev 11 (2006) 128–150. doi: 10.1016/j.cyto.2017.10.019. [DOI] [PubMed] [Google Scholar]
  • [16].Petri M, Allbritton J, Antibiotic allergy in systemic lupus erythematosus: a case-control study., J. Rheumatol 19 (1992) 265–9. http://www.ncbi.nlm.nih.gov/pubmed/1629825. [PubMed] [Google Scholar]
  • [17].Costenbader KH, Kim DJ, Peerzada J, Lockman S, Nobles-Knight D, Petri M, Karlson EW, Cigarette Smoking and the Risk of Systemic Lupus Erythematosus: A Meta-Analysis, Arthritis Rheum. 50 (2004) 849–857. doi: 10.1002/art.20049. [DOI] [PubMed] [Google Scholar]
  • [18].Blomberg J, Nived O, Pipkorn R, Bengtsson A, Erlinge D, Sturfelt G, Increased antiretroviral antibody reactivity in sera from a defined population of patients with systemic lupus erythematosus: Correlation with autoantibodies and clinical manifestations., Arthritis Rheum. 37 (1994) 57–66. http://www.ncbi.nlm.nih.gov/pubmed/7510483. [DOI] [PubMed] [Google Scholar]
  • [19].Cooper GS, Parks CG, Occupational and environmental exposures as risk factors for systemic lupus erythematosus., Curr. Rheumatol. Rep 6 (2004) 367–74. http://www.ncbi.nlm.nih.gov/pubmed/15355749. [DOI] [PubMed] [Google Scholar]
  • [20].James JA, Neas BR, Moser KL, Hall T, Bruner GR, Sestak AL, Harley JB, Systemic lupus erythematosus in adults is associated with previous Epstein-Barr virus exposure., Arthritis Rheum. 44 (2001) 1122–6. doi:. [DOI] [PubMed] [Google Scholar]
  • [21].Parks CG, Cooper GS, Nylander-French LA, Sanderson WT, Dement JM, Cohen PL, Dooley MA, Treadwell EL, St EW.Clair, G.S. Gilkeson, J.A. Hoppin, D.A. Savitz, Occupational exposure to crystalline silica and risk of systemic lupus erythematosus: A population-based, case-control study in the southeastern United States, Arthritis Rheum. 46 (2002) 1840–1850. doi: 10.1002/art.10368. [DOI] [PubMed] [Google Scholar]
  • [22].Crowe W, Allsopp PJ, Watson GE, Magee PJ, Strain JJ, Armstrong DJ, Ball E, McSorley EM, Mercury as an environmental stimulus in the development of autoimmunity – A systematic review, Autoimmun. Rev 16 (2017) 72–80. doi: 10.1016/j.autrev.2016.09.020. [DOI] [PubMed] [Google Scholar]
  • [23].Zarmbinski MA, Messner RP, Mandel JS, Anti-dsDNA antibodies in laboratory workers handling blood from patients with systemic lupus erythematosus., J. Rheumatol 19 (1992) 1380–4. http://www.ncbi.nlm.nih.gov/pubmed/1433005. [PubMed] [Google Scholar]
  • [24].Chiou S-H, Lan J-L, Lin S-L, Chen D-Y, Tsai N-Y, Kuan C-Y, Lin T-Y, Lin F-J, Lee W-M, Chang T-J, Pet dogs owned by lupus patients are at a higher risk of developing lupus, Lupus. 13 (2004) 442–449. doi: 10.1191/0961203303lu1039oa. [DOI] [PubMed] [Google Scholar]
  • [25].Song H, Fang F, Tomasson G, Arnberg FK, Mataix-Cols D, Fernández de la Cruz L, Almqvist C, Fall K, Valdimarsdóttir UA , Association of Stress-Related Disorders With Subsequent Autoimmune Disease., Jama. 319 (2018) 2388–2400. doi: 10.1001/jama.2018.7028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Roberts AL, Malspeis S, Kubzansky LD, Feldman CH, Chang S-C, Koenen KC, Costenbader KH, Association of Trauma and Posttraumatic Stress Disorder With Incident Systemic Lupus Erythematosus in a Longitudinal Cohort of Women., Arthritis Rheumatol. (Hoboken, N.J.) 69 (2017) 2162–2169. doi: 10.1002/art.40222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Crow MK, Etiology and Pathogenesis of Systemic Lupus Erythematosus, Tenth Edit, Elsevier Inc, 2017. doi: 10.1016/B978-0-323-31696-5.00079-6. [DOI] [Google Scholar]
  • [28].Katsuyama T, Tsokos GC, Moulton VR, Aberrant T cell signaling and subsets in systemic lupus erythematosus, Front. Immunol 9 (2018). doi: 10.3389/fimmu.2018.01088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, a Ortmann W, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW, Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus., Proc. Natl. Acad. Sci. U. S. A 100 (2003) 2610–5. doi: 10.1073/pnas.0337679100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V, Interferon and Granulopoiesis Signatures in Systemic Lupus Erythematosus Blood, J. Exp. Med 197 (2003) 711–723. doi: 10.1084/jem.20021553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Kirou KA, Lee C, George S, Louca K, Papagiannis IG, Peterson MGE, Ly N, Woodward RN, Fry KE, Lau AY-H, Prentice JG, Wohlgemuth JG, Crow MK, Coordinate overexpression of interferon-?-induced genes in systemic lupus erythematosus, Arthritis Rheum. 50 (2004) 3958–3967. http://doi.wiley.com/10.1002/art.20798. [DOI] [PubMed] [Google Scholar]
  • [32].Kaplan MJ, Radic M, Neutrophil Extracellular Traps: Double-Edged Swords of Innate Immunity, J. Immunol 189 (2012) 2689–2695. doi: 10.4049/jimmunol.1201719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, Banchereau J, Pascual V, Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus, Sci. Transl. Med 3 (2011). doi: 10.1126/scitranslmed.3001201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V, Bassett R, Amuro H, Fukuhara S, Ito T, Liu YJ, Gilliet M, Neutrophils Activate Plasmacytoid Dendritic Cells by Releasing Self-DNA Peptide Complexes in Systemic Lupus Erythematosus, Sci. Transl. Med 3 (2011) 73ra19–73ra19. doi: 10.1126/scitranslmed.3001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A, Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis., Proc. Natl. Acad. Sci. U. S. A 107 (2010) 9813–9818. http://eutils.ncbi.nlm.nih.gov/entrez/eutils/elink.fcgi?dbfrom=pubmed&id=20439745&retmode=ref&cmd=prlinks. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Leffler J, Ciacma K, Gullstrand B, Bengtsson AA, Martin M, Blom AM, A subset of patients with systemic lupus erythematosus fails to degrade DNA from multiple clinically relevant sources, Arthritis Res. Ther 17 (2015) 205. doi: 10.1186/s13075-015-0726-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].a Casciola-Rosen L, Anhalt G, a Rosen, Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes., J. Exp. Med 179 (1994) 1317–1330. doi: 10.1084/jem.179.4.1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Casciola-Rosen L, a Rosen M Petri, M. Schlissel, Surface blebs on apoptotic cells are sites of enhanced procoagulant activity: implications for coagulation events and antigenic spread in systemic lupus erythematosus., Proc. Natl. Acad. Sci. U. S. A 93 (1996) 1624–1629. doi: 10.1073/pnas.93.4.1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Lehmann P, Homey B, Clinic and pathophysiology of photosensitivity in lupus erythematosus, Autoimmun. Rev 8 (2009) 456–461. doi: 10.1016/j.autrev.2008.12.012. [DOI] [PubMed] [Google Scholar]
  • [40].Guiducci C, Gong M, Xu Z, Gill M, Chaussabel D, Meeker T, Chan JH, Wright T, Punaro M, Bolland S, Soumelis V, Banchereau J, Coffman RL, Pascual V, Barrat FJ, TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus, Nature. 465 (2010) 937–941. doi: 10.1038/nature09102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Paidassi H, Tacnet-Delorme P, Garlatti V, Darnault C, Ghebrehiwet B, Gaboriaud C, Arlaud GJ, Frachet P, C1q Binds Phosphatidylserine and Likely Acts as a Multiligand-Bridging Molecule in Apoptotic Cell Recognition, J. Immunol 180 (2008) 2329–2338. doi: 10.4049/jimmunol.180.4.2329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Ehrenstein MR, Notley CA, The importance of natural IgM: scavenger, protector and regulator, Nat. Rev. Immunol 10 (2010) 778–786. doi: 10.1038/nri2849. [DOI] [PubMed] [Google Scholar]
  • [43].Thompson RA, Haeney M, Reid KBM, Davies JG, White RHR, Cameron AH, A Genetic Defect of the C1q Subcomponent of Complement Associated with Childhood (Immune Complex) Nephritis, N. Engl. J. Med 303 (1980) 22–24. doi: 10.1056/NEJM198007033030107. [DOI] [PubMed] [Google Scholar]
  • [44].Sisirak V, Sally B, D’Agati V, Martinez-Ortiz W, Özçakar ZB, David J, Rashidfarrokhi A, Yeste A, Panea C, Chida ASS, Bogunovic M, Ivanov III, Quintana FJJ, Sanz I, Elkon KBB, Tekin M, Yalçınkaya F, Cardozo TJJ, Clancy RMM, Buyon JPP, Reizis B, Digestion of Chromatin in Apoptotic Cell Microparticles Prevents Autoimmunity, Cell. 166 (2016) 88–101. doi: 10.1016/j.cell.2016.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Lo MS, Monogenic Lupus, Curr. Rheumatol. Rep 18 (2016) 1–7. doi: 10.1007/s11926-016-0621-9. [DOI] [PubMed] [Google Scholar]
  • [46].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. 25 (1982) 1271–1277. doi: 10.1002/art.1780251101. [DOI] [PubMed] [Google Scholar]
  • [47].Hochberg MC, Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus., Arthritis Rheum. 40 (1997) 1725. doi:. [DOI] [PubMed] [Google Scholar]
  • [48].Petri M, Orbai A-M, Alarcón GS, et al. , Derivationand validation of the systemic lupus international collaborating clinics classification criteria for systemic lupus erythematosus, Arthritis Rheum. 64 (2012) 2677–2686. doi: 10.1002/art.34473.Derivation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Tedeschi SK, Johnson SR, Boumpas D, Daikh D, Dörner T, Jayne D, Kamen D, Lerstrøm K, Mosca M, Ramsey-Goldman R, Sinnette C, Wofsy D, Smolen JS, Naden RP, Aringer M, Costenbader KH, Developing and Refining New Candidate Criteria for Systemic Lupus Erythematosus Classification: An International Collaboration, Arthritis Care Res 70 (2018) 571–581. doi: 10.1002/acr.23317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Petri M, Goldman D, Magder LS, Validation of Proposed EULAR/Acr SLE Classification Criteria Versus SLICC SLE Classification Criteria, Arthritis Rheumatol. 70 (suppl (2018). [Google Scholar]
  • [51].Hartman EAR, van Royen-Kerkhof A, Jacobs JWG, Welsing PMJ, Fritsch-Stork RDE, Performance of the 2012 Systemic Lupus International Collaborating Clinics classification criteria versus the 1997 American College of Rheumatology classification criteria in adult and juvenile systemic lupus erythematosus. A systematic review and meta-an, Autoimmun. Rev 17 (2018) 316–322. doi: 10.1016/j.autrev.2018.01.007. [DOI] [PubMed] [Google Scholar]
  • [52].Wallace D, Hahn BH, Dubois’ Lupus Erythematosus and Related Syndromes, 8th ed., Saunders, 2012. [Google Scholar]
  • [53].Firestein GS, Budd RC, Gabriel S, McInnes IB, O’Dell J, Kelley and Firestein’s Textbook of Rheumatology, 10th ed., Elsevier, 2016. [Google Scholar]
  • [54].Hejazi EZ, Werth VP, Cutaneous Lupus Erythematosus: An Update on Pathogenesis, Diagnosis and Treatment, Am. J. Clin. Dermatol 17 (2016) 135–146. doi: 10.1007/s40257-016-0173-9. [DOI] [PubMed] [Google Scholar]
  • [55].Walling HW, Sontheimer RD, Cutaneous lupus erythematosus: issues in diagnosis and treatment., Am. J. Clin. Dermatol 10 (2009) 365–381. doi: 10.2165/11310780-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • [56].Hasan T, Nyberg F, Stephansson E, Puska P, Hakkinen M, Sarna S, Ros A-M, Ranki A, Photosensitivity in lupus erythematosus, UV photoprovocation results compared with history of photosensitivity and clinical findings, Br. J. Dermatol 136 (1997) 699–705. doi: 10.1046/j.1365-2133.1997.6591644.x. [DOI] [PubMed] [Google Scholar]
  • [57].Sanders CJG, Van Weelden H, Kazzaz GAA, Sigurdsson V, Toonstra J, Bruijnzeel-Koomen CAFM, Photosensitivity in patients with lupus erythematosus: a clinical and photobiological study of 100 patients using a prolonged phototest protocol., Br. J. Dermatol 149 (2003) 131–7. doi: 10.2165/11591790-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • [58].Kuhn A, Sonntag M, Richter-Hintz D, Oslislo C, Megahed M, Ruzicka T, Lehmann P, Phototesting in lupus erythematosus: a 15-year experience., J. Am. Acad. Dermatol 45 (2001) 86–95. doi: 10.1067/mjd.2001.114589. [DOI] [PubMed] [Google Scholar]
  • [59].Zandman-Goddard G, Solomon M, Rosman Z, Peeva E, Shoenfeld Y, Environment and lupus-related diseases, Lupus. 21 (2012) 241–250. doi: 10.1177/0961203311426568. [DOI] [PubMed] [Google Scholar]
  • [60].Alarcon Segovia D, Cetina JA, Lupus hair, Am. J. Med. Sci 267 (1974) 241–242. doi: 10.1039/c5ra13683k. [DOI] [PubMed] [Google Scholar]
  • [61].Miot HA, Miot LDB, Haddad GR, Association between discoid lupus erythematosus and cigarette smoking: A case-control study, Dermatology. 211 (2005) 118–122. doi: 10.1159/000086440. [DOI] [PubMed] [Google Scholar]
  • [62].Böckle BC, Sepp NT, Smoking is highly associated with discoid lupus erythematosus and lupus erythematosus tumidus: analysis of 405 patients, Lupus. 24 (2015) 669–674. doi: 10.1177/0961203314559630. [DOI] [PubMed] [Google Scholar]
  • [63].Piette EW, Foering KP, Chang AY, Okawa J, Ten Have TR, Feng R, Werth VP, Impact of smoking in cutaneous lupus erythematosus., Arch. Dermatol 148 (2012) 317–22. doi: 10.1001/archdermatol.2011.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Chasset F, Francès C, Barete S, Amoura Z, Arnaud L, Influence of smoking on the efficacy of antimalarials in cutaneous lupus: A meta-analysis of the literature, J. Am. Acad. Dermatol 72 (2015) 634–639. doi: 10.1016/j.jaad.2014.12.025. [DOI] [PubMed] [Google Scholar]
  • [65].Petri M, Musculoskeletal complications of systemic lupus erythematosus in the Hopkins Lupus Cohort: an update., Arthritis Care Res. 8 (1995) 137–45. http://www.ncbi.nlm.nih.gov/pubmed/7654797. [DOI] [PubMed] [Google Scholar]
  • [66].Grossman JM, Lupus arthritis, Best Pract. Res. Clin. Rheumatol 23 (2009) 495–506. doi: 10.1016/j.berh.2009.04.003. [DOI] [PubMed] [Google Scholar]
  • [67].Budhram A, Chu R, Rusta-Sallehy S, Ioannidis G, Denburg JA, Adachi JD, Haaland DA, Anti-cyclic citrullinated peptide antibody as a marker of erosive arthritis in patients with systemic lupus erythematosus: A systematic review and meta-analysis, Lupus. 23 (2015) 1156–1163. doi: 10.1177/0961203314540967. [DOI] [PubMed] [Google Scholar]
  • [68].Lins CF, Santiago MB, Ultrasound evaluation of joints in systemic lupus erythematosus: a systematic review, Eur. Radiol 25 (2015) 2688–2692. doi: 10.1007/s00330-015-3670-y. [DOI] [PubMed] [Google Scholar]
  • [69].Ostendorf B, Scherer A, Specker C, Mödder U, Schneider M, Jaccoud’s arthropathy in systemic lupus erythematosus: Differentiation of deforming and erosive patterns by magnetic resonance imaging, Arthritis Rheum. 48 (2003) 157–165. doi: 10.1002/art.10753. [DOI] [PubMed] [Google Scholar]
  • [70].Zollars ES, Hyer M, Wolf B, Chapin R, Measuring lupus arthritis activity using contrasted high-field MRI. Associations with clinical measures of disease activity and novel patterns of disease, Lupus Sci. Med 5 (2018) e000264. doi: 10.1136/lupus-2018-000264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Wolfe F, Petri M, Alarcón GS, Goldman J, Chakravarty EF, Katz RS, Karlson EW, Fibromyalgia, Systemic Lupus Erythematosus (SLE), and Evaluation of SLE Activity, J. Rheumatol 36 (2008) 82–88. doi: 10.3899/jrheum.080212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].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 37 (2010) 2032–2038. doi: 10.3899/jrheum.091366. [DOI] [PubMed] [Google Scholar]
  • [73].Bastian HM, Roseman JM, Mcgwin G, Alarcón GS, Friedman AW, Fessler BJ, Baethge BA, Reveille JD, Systemic lupus erythematosus in three ethnic groups. XII. Risk factors for lupus nephritis after diagnosis, Lupus. 11 (2002) 152–160. doi: 10.1191/0961203302lu158oa. [DOI] [PubMed] [Google Scholar]
  • [74].Bernatsky S, Boivin JF, Joseph L, Manzi S, Ginzler E, Gladman DD, Urowitz M, Fortin PR, Petri M, Barr S, Gordon C, Bae SC, Isenberg D, Zoma A, Aranow C, Dooley MA, Nived O, Sturfelt G, Steinsson K, Alarcón G, Senécal JL, Zummer M, Hanly J, Ensworth S, Pope J, Edworthy S, Rahman A, Sibley J, El-Gabalawy H, McCarthy T, St Y. Pierre, A. Clarke, R. Ramsey-Goldman, Mortality in systemic lupus erythematosus, Arthritis Rheum. 54 (2006) 2550–2557. doi: 10.1002/art.21955. [DOI] [PubMed] [Google Scholar]
  • [75].Faurschou M, Starklint H, Halberg P, Jacobsen S, Prognostic factors in lupus nephritis: Diagnostic and therapeutic delay increases the risk of terminal renal failure, J. Rheumatol 33 (2006) 1563–1569. [PubMed] [Google Scholar]
  • [76].WAKASUGI D, GONO T, KAWAGUCHI Y, HARA M, KOSEKI Y, KATSUMATA Y, HANAOKA M, YAMANAKA H, Frequency of Class III and IV Nephritis in Systemic Lupus Erythematosus Without Clinical Renal Involvement: An Analysis of Predictive Measures, J. Rheumatol 39 (2012) 79–85. doi: 10.3899/jrheum.110532. [DOI] [PubMed] [Google Scholar]
  • [77].Christopher-Stine L, Siedner M, Lin J, Haas M, Parekh H, Petri M, Fine DM, Renal biopsy in lupus patients with low levels of proteinuria, J. Rheumatol 34 (2007) 332–335. [PubMed] [Google Scholar]
  • [78].Christopher-Stine L, Petri M, Astor BC, Fine D, Urine protein-to-creatinine ratio is a reliable measure of proteinuria in lupus nephritis., J. Rheumatol 31 (2004) 1557–9. http://www.ncbi.nlm.nih.gov/pubmed/15290735. [PubMed] [Google Scholar]
  • [79].Olson SW, Lee JJ, Prince LK, Baker TP, Papadopoulos P, Edison J, Abbott KC, Elevated subclinical double-stranded DNA antibodies and future proliferative lupus nephritis, Clin. J. Am. Soc. Nephrol 8 (2013) 1702–1708. doi: 10.2215/CJN.01910213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Orbai A-M, Truedsson L, Sturfelt G, Nived O, Fang H, Alarcón G, Gordon C, Merrill J, Fortin P, Bruce I, Isenberg D, Wallace D, Ramsey-Goldman R, Bae S-C, Hanly J, Sanchez-Guerrero J, Clarke A, Aranow C, Manzi S, Urowitz M, Gladman D, Kalunian K, Costner M, Werth V, Zoma A, Bernatsky S, Ruiz-Irastorza G, Khamashta M, Jacobsen S, Buyon J, Maddison P, Dooley M, Van Vollenhoven R, Ginzler E, Stoll T, Peschken C, Jorizzo J, Callen J, Lim S, Fessler B, Inanc M, Kamen D, Rahman A, Steinsson K, Franks A, Sigler L, Hameed S, Pham N, Brey R, Weisman M, McGwin G, Magder L, Petri M, Anti-C1q antibodies in systemic lupus erythematosus., Lupus. 24 (2014) pii: 0961203314547791. doi: 10.1177/0961203314547791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Akhter E, Burlingame R, Seaman A, Magder L, Petri M, Anti-C1q antibodies have higher correlation with flares of lupus nephritis than other serum markers, Lupus. 20 (2011) 1267–1274. doi: 10.1177/0961203311411597. [DOI] [PubMed] [Google Scholar]
  • [82].Stojan G, Petri M, Anti-C1q in systemic lupus erythematosus, Lupus. 25 (2016) 873–877. doi: 10.1177/0961203316645205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Weening JJ, D’agati VD, Schwartz MM, Seshan SV, Alpers CE, Appel GB, Balow JE, Bruijn JANA, Cook T, Ferrario F, Fogo AB, Ginzler EM, Hebert LEE, Hill G, Hill P, Jennette JC, Kong NC, Lesavre P, Lockshin M, Looi L-M, Makino H, Moura LA, Nagata M, The classification of glomerulonephritis in systemic lupus erythematosus revisited, Kidney Int. 65 (2004) 521–530. doi: 10.1111/j.1523-1755.2004.00443.x. [DOI] [PubMed] [Google Scholar]
  • [84].Shea-Simonds P, Cairns TD, Roufosse C, Cook T, Vyse TJ, Lupus podocytopathy, Rheumatology. 48 (2009) 1616–1618. doi: 10.1093/rheumatology/kep256. [DOI] [PubMed] [Google Scholar]
  • [85].Descombes E, Droz D, Drouet L, Grünfeld JP, Lesavre P, Renal vascular lesions in lupus nephritis., Medicine (Baltimore). 76 (1997) 355–68. http://www.ncbi.nlm.nih.gov/pubmed/9352738. [DOI] [PubMed] [Google Scholar]
  • [86].Salvatore SP, Barisoni LMC, Herzenberg AM, Chander PN, Nickeleit V, Seshan SV, Collapsing glomerulopathy in 19 patients with systemic lupus erythematosus or Lupus-like disease, Clin. J. Am. Soc. Nephrol 7 (2012) 914–925. doi: 10.2215/CJN.11751111. [DOI] [PubMed] [Google Scholar]
  • [87].Ainiala H, Hietaharju A, Loukkola J, Peltola J, Korpela M, Metsänoja R, Auvinen A, Validity of the new American College of Rheumatology criteria for neuropsychiatric lupus syndromes: a population‐based evaluation, Arthritis Rheum. 45 (2001) 419–423. doi: 10.1016/j.measurement.2013.09.007. [DOI] [PubMed] [Google Scholar]
  • [88].Petri M, Orbai AM, Alarcõn GS, Gordon C, Merrill JT, Fortin PR, Bruce IN, Isenberg D, Wallace DJ, Nived O, Sturfelt G, Ramsey-Goldman R, Bae SC, Hanly JG, Sánchez-Guerrero J, Clarke A, Aranow C, Manzi S, Urowitz M, Gladman D, Kalunian K, Costner M, Werth VP, Zoma A, Bernatsky S, Ruiz-Irastorza G, Khamashta MA, Jacobsen S, Buyon JP, Maddison P, Dooley MA, Van Vollenhoven RF, Ginzler E, Stoll T, Peschken C, Jorizzo JL, Callen JP, Lim SS, Fessler BJ, Inanc M, Kamen DL, Rahman A, Steinsson K, Franks AG, Sigler L, Hameed S, Fang H, Pham N, Brey R, Weisman MH, McGwin G, Magder LS, Derivation and validation of the systemic lupus international collaborating clinics classification criteria for systemic lupus erythematosus, Arthritis Rheum. 64 (2012) 2677–2686. doi: 10.1002/art.34473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Sibbitt WL, Sibbitt RR, Brooks WM, Neuroimaging in neuropsychiatric systemic lupus erythematosus, Arthritis Rheum. 42 (1999) 2026–2038. doi:. [DOI] [PubMed] [Google Scholar]
  • [90].Birnbaum J, Petri M, Thompson R, Izbudak I, Kerr D, Distinct subtypes of myelitis in systemic lupus erythematosus, Arthritis Rheum. 60 (2009) 3378–3387. doi: 10.1002/art.24937. [DOI] [PubMed] [Google Scholar]
  • [91].Miller DH, Buchanan N, Barker G, Morrissey SP, Kendall BE, Rudge P, Khamashta M, Hughes GR, McDonald WI, Gadolinium-enhanced magnetic resonance imaging of the central nervous system in systemic lupus erythematosus., J. Neurol 239 (1992) 460–4. doi:1447575. [DOI] [PubMed] [Google Scholar]
  • [92].Sibbitt WL, Brooks WM, Haseler LJ, Griffey RH, Frank LM, Hart BL, Sibbitt RR, Spin-spin relaxation of brain tissues in systemic lupus erythematosus, Arthritis Rheum. 38 (1995) 810–818. doi: 10.1002/art.1780380615. [DOI] [PubMed] [Google Scholar]
  • [93].West SG, Emlen W, Wener MH, Kotzin BL, Neuropsychiatric lupus erythematosus: A 10-year prospective study on the value of diagnostic tests, Am. J. Med 99 (1995) 153–163. doi: 10.1016/S0002-9343(99)80135-1. [DOI] [PubMed] [Google Scholar]
  • [94].Carbotte RM, Denburg SD, Denburg JA, Prevalence of cognitive impairment in systemic lupus erythematosus., J. Nerv. Ment. Dis 174 (1986) 357–64. http://www.ncbi.nlm.nih.gov/pubmed/3711879. [DOI] [PubMed] [Google Scholar]
  • [95].Kozora E, Ellison MC, West S, Reliability and validity of the proposed American College of Rheumatology neuropsychological battery for systemic lupus erythematosus, Arthritis Care Res. 51 (2004) 810–818. doi: 10.1002/art.20692. [DOI] [PubMed] [Google Scholar]
  • [96].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 35 (2008) 1776–1781. doi:08/13/0716 [pii]. [PubMed] [Google Scholar]
  • [97].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 37 (2010) 2032–2038. doi: 10.3899/jrheum.091366. [DOI] [PubMed] [Google Scholar]
  • [98].Leadbetter EA, Rifkin IR, Hohlbaum AM, Beaudette BC, Shlomchik MJ, Marshak-Rothstein A, Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors, Nature. 416 (2002) 603–607. doi: 10.1038/416603a. [DOI] [PubMed] [Google Scholar]
  • [99].Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD, Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9., J. Clin. Invest 115 (2005) 407–17. doi: 10.1172/JCI23025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B, Jerala R, Mechanism of Endosomal TLR Inhibition by Antimalarial Drugs and Imidazoquinolines, J. Immunol 186 (2011) 4794–4804. doi: 10.4049/jimmunol.1000702. [DOI] [PubMed] [Google Scholar]
  • [101].Lafyatis R, York M, Marshak-Rothstein A, Antimalarial agents: Closing the gate on toll-like receptors?, Arthritis Rheum. 54 (2006) 3068–3070. doi: 10.1002/art.22157. [DOI] [PubMed] [Google Scholar]
  • [102].Petri M, Hydroxychloroquine use in the Baltimore Lupus Cohort: effects on lipids, glucose and thrombosis., Lupus. 5 Suppl 1 (1996) S16–22. http://www.ncbi.nlm.nih.gov/pubmed/8803905. [PubMed] [Google Scholar]
  • [103].Fox R, Anti-malarial drugs: possible mechanisms of action in autoimmune disease and prospects for drug development., Lupus. 5 Suppl 1 (1996) S4–10. http://www.ncbi.nlm.nih.gov/pubmed/8803903. [PubMed] [Google Scholar]
  • [104].Wozniacka A, Lesiak A, Narbutt J, McCauliffe DP, Sysa-Jedrzejowska A, Chloroquine treatment influences proinflammatory cytokine levels in systemic lupus erythematosus patients, Lupus. 15 (2006) 268–275. doi: 10.1191/0961203306lu2299oa. [DOI] [PubMed] [Google Scholar]
  • [105].An J, Woodward JJ, Sasaki T, Minie M, Elkon KB, Cutting Edge: Antimalarial Drugs Inhibit IFN-β Production through Blockade of Cyclic GMP-AMP Synthase–DNA Interaction, J. Immunol 194 (2015) 4089–4093. doi: 10.4049/jimmunol.1402793. [DOI] [PubMed] [Google Scholar]
  • [106].Alarcón GS, McGwin G, Bertoli AM, Fessler BJ, Calvo-Alén J, Bastian HM, Vilà LM, Reveille JD, Effect of hydroxychloroquine on the survival of patients with systemic lupus erythematosus: Data from LUMINA, a multiethnic US cohort (LUMINA L), Ann. Rheum. Dis 66 (2007) 1168–1172. doi: 10.1136/ard.2006.068676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Ruiz-Irastorza G, Egurbide M-V, Pijoan J-I, Garmendia M, Villar I, Martinez-Berriotxoa A, Erdozain J-G, Aguirre C, Effect of antimalarials on thrombosis and survival in patients with systemic lupus erythematosus, Lupus. 15 (2006) 577–583. doi: 10.1177/0961203306071872. [DOI] [PubMed] [Google Scholar]
  • [108].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 69 (2010) 20–28. doi: 10.1136/ard.2008.101766. [DOI] [PubMed] [Google Scholar]
  • [109].Mok CC, Tse SM, Chan KL, Ho LY, Effect of immunosuppressive therapies on survival of systemic lupus erythematosus: a propensity score analysis of a longitudinal cohort, Lupus. 27 (2018) 722–727. doi: 10.1177/0961203317739129. [DOI] [PubMed] [Google Scholar]
  • [110].Canadian Hydroxychloroquine Study Group, A randomized study of the effect of withdrawing hydroxychloroquine sulfate in systemic lupus erythematosus., N. Engl. J. Med 324 (1991) 150–4. doi: 10.1056/NEJM199101173240303. [DOI] [PubMed] [Google Scholar]
  • [111].Fessler BJ, Alarcón GS, McGwin G, Roseman J, Bastian HM, Friedman AW, Baethge BA, Vilá L, Reveille JD, Systemic lupus erythematosus in three ethnic groups: XVI. Association of hydroxychloroquine use with reduced risk of damage accrual, Arthritis Rheum. 52 (2005) 1473–1480. doi: 10.1002/art.21039. [DOI] [PubMed] [Google Scholar]
  • [112].Fasano S, Pierro L, Pantano I, Iudici M, Valentini G, Longterm Hydroxychloroquine Therapy and Low-dose Aspirin May Have an Additive Effectiveness in the Primary Prevention of Cardiovascular Events in Patients with Systemic Lupus Erythematosus, J. Rheumatol 44 (2017) 1032–1038. doi: 10.3899/jrheum.161351. [DOI] [PubMed] [Google Scholar]
  • [113].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. 15 (2006) 366–370. doi: 10.1191/0961203306lu2313oa. [DOI] [PubMed] [Google Scholar]
  • [114].Hanly JG, Urowitz MB, Su L, Gordon C, Bae S-C, Sanchez-Guerrero J, Romero-Diaz J, Wallace DJ, Clarke AE, Ginzler E, Merrill JT, Isenberg DA, Rahman A, Petri M, Fortin PR, Gladman D, Bruce IN, Steinsson K, Dooley M, Khamashta MA, Alarcón GS, Fessler BJ, Ramsey-Goldman R, Manzi S, Zoma AA, Sturfelt GK, Nived O, Aranow C, Mackay M, Ramos-Casals M, van Vollenhoven R, Kalunian KC, Ruiz-Irastorza G, Lim S, Kamen DL, Peschken CA, Inanc M, Theriault C, Thompson K, Farewell V, Seizure disorders in systemic lupus erythematosus results from an international, prospective, inception cohort study, Ann. Rheum. Dis 71 (2012) 1502–1509. doi: 10.1136/annrheumdis-2011-201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Andrade RM, Alarcón GS, González LA, Fernández M, Apte M, Vilá LM, McGwin G, Reveille JD, Seizures in patients with systemic lupus erythematosus: Data from LUMINA, a multiethnic cohort (LUMINA LIV), Ann. Rheum. Dis 67 (2008) 829–834. doi: 10.1136/ard.2007.077594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].González LA, Pons-Estel GJ, Zhang J, Vilá LM, Reveille JD, Alarcón GS, Time to neuropsychiatric damage occurrence in LUMINA (LXVI): A multi-ethnic lupus cohort, Lupus. 18 (2009) 822–830. doi: 10.1177/0961203309104392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Ruzicka T, Sommerburg C, Goerz G, Kind P, Mensing H, Treatment of cutaneous lupus erythematosus with acitretin and hydroxychloroquine., Br. J. Dermatol 127 (1992) 513–8. http://www.ncbi.nlm.nih.gov/pubmed/1467292. [DOI] [PubMed] [Google Scholar]
  • [118].Kuhn A, Ruland V, Bonsmann G, Cutaneous lupus erythematosus: Update of therapeutic options: Part i, J. Am. Acad. Dermatol 65 (2011) e179–e193. doi: 10.1016/j.jaad.2010.06.018. [DOI] [PubMed] [Google Scholar]
  • [119].Williams HJ, Egger MJ, Singer JZ, Willkens RF, Kalunian KC, Clegg DO, Skosey JL, Brooks RH, Alarcón GS, Steen VD, Comparison of hydroxychloroquine and placebo in the treatment of the arthropathy of mild systemic lupus erythematosus., J. Rheumatol 21 (1994) 1457–62. http://www.ncbi.nlm.nih.gov/pubmed/7983646. [PubMed] [Google Scholar]
  • [120].Babary H, Liu X, Ayatollahi Y, Chen XP, Doo L, Uppaluru LK, Kwak MK, Kulaga C, Modjinou D, Olech E, Yoo JW, Favorable effects of hydroxychloroquine on serum low density lipid in patients with systemic lupus erythematosus: A systematic review and meta-analysis, Int. J. Rheum. Dis 21 (2018) 84–92. doi: 10.1111/1756-185X.13159. [DOI] [PubMed] [Google Scholar]
  • [121].Petri M, Lakatta C, Magder L, Goldman D, Effect of prednisone and hydroxychloroquine on coronary artery disease risk factors in systemic lupus erythematosus: A longitudinal data analysis, Am. J. Med 96 (1994) 254–259. doi: 10.1016/0002-9343(94)90151-1. [DOI] [PubMed] [Google Scholar]
  • [122].Cairoli E, Rebella M, Danese N, Garra V, Borba EF, Hydroxychloroquine reduces low-density lipoprotein cholesterol levels in systemic lupus erythematosus: a longitudinal evaluation of the lipid-lowering effect., Lupus. 21 (2012) 1178–1182. doi: 10.1177/0961203312450084. [DOI] [PubMed] [Google Scholar]
  • [123].Wasko MCM, McClure CK, Kelsey SF, Huber K, Orchard T, Toledo FGS, Antidiabetogenic effects of hydroxychloroquine on insulin sensitivity and beta cell function: a randomised trial, Diabetologia. 58 (2015) 2336–2343. doi: 10.1007/s00125-015-3689-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Petri M, Thrombosis and systemic lupus erythematosus: the Hopkins Lupus Cohort perspective., Scand. J. Rheumatol 25 (1996) 191–3. http://www.ncbi.nlm.nih.gov/pubmed/8792794. [DOI] [PubMed] [Google Scholar]
  • [125].Pierangeli SS, Harris EN, In vivo models of thrombosis for the antiphospholipid syndrome, Lupus. 5 (1996) 451–455. doi: 10.1177/096120339600500524. [DOI] [PubMed] [Google Scholar]
  • [126].Mok CC, Mak A, Ma KM, Bone mineral density in postmenopausal Chinese patients with systemic lupus erythematosus, Lupus. 14 (2005) 106–112. doi: 10.1191/0961203305lu2039oa. [DOI] [PubMed] [Google Scholar]
  • [127].Herrinton LJ, Liu L, Goldfien R, Michaels MA, Tran TN, Risk of Serious Infection for Patients with Systemic Lupus Erythematosus Starting Glucocorticoids with or without Antimalarials, J. Rheumatol 43 (2016) 1503–1509. doi: 10.3899/jrheum.150671. [DOI] [PubMed] [Google Scholar]
  • [128].Fang Y, Chen Y, Chung T, See L, Yu K, Luo S, Kuo C, Lai J, Hydroxychloroquine and risk of cancer in patients with primary Sjögren syndrome: propensity score matched landmark analysis., Oncotarget. 8 (2017) 80461–80471. doi: 10.18632/oncotarget.19057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Wolfe F, Marmor MF, Rates and predictors of hydroxychloroquine retinal toxicity in patients with rheumatoid arthritis and systemic lupus erythematosus, Arthritis Care Res. (Hoboken) 62 (2010) 775–784. doi: 10.1002/acr.20133. [DOI] [PubMed] [Google Scholar]
  • [130].Marmor MF, Kellner U, Lai TYY, Lyons JS, Mieler WF, American Academy of Ophthalmology, Revised recommendations on screening for chloroquine and hydroxychloroquine retinopathy., Ophthalmology. 118 (2011) 415–22. doi: 10.1016/j.ophtha.2010.11.017. [DOI] [PubMed] [Google Scholar]
  • [131].Wang Y, Zhu J, DeLuca HF, Where is the vitamin D receptor?, Arch. Biochem. Biophys 523 (2012) 123–133. doi: 10.1016/j.abb.2012.04.001. [DOI] [PubMed] [Google Scholar]
  • [132].Daniel C, Sartory NA, Zahn N, Radeke HH, Stein JM, Immune modulatory treatment of trinitrobenzene sulfonic acid colitis with calcitriol is associated with a change of a T helper (Th) 1/Th17 to a Th2 and regulatory T cell profile, J. Pharmacol. Exp. Ther 324 (2008) 23–33. doi: 10.1124/jpet.107.127209. [DOI] [PubMed] [Google Scholar]
  • [133].Yu S, Cantorna MT, The vitamin D receptor is required for iNKT cell development., Proc. Natl. Acad. Sci. U. S. A 105 (2008) 5207–12. doi: 10.1073/pnas.0711558105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].V Bonventre J, Antifibrotic vitamin D analogs, 123 (2013) 22–25. doi: 10.1172/JCI72748.4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Der E, Suryawanshi H, Ranabothu S, Goilav B, Belmont HM, Izmirly P, Bornkamp N, Jordan N, Wang T, Wu M, James J, Guthridge J, Raychaudhuri S, Tuschl T, Buyon J, Putterman C, Kidney and Skin Single-Cell RNA Sequencing in Lupus Nephritis Provides Mechanistic Insights and Novel Potential Biomarkers, Arthritis Rheumatol. 69 (suppl (2017). https://acrabstracts.org/abstract/kidney-and-skin-single-cell-rna-sequencing-in-lupus-nephritis-provides-mechanistic-insights-and-novel-potential-biomarkers/. [Google Scholar]
  • [136].Shi Y, Liu T, Yao L, Xing Y, Zhao X, Fu J, Xue X, Chronic Vitamin D deficiency induces lung fibrosis through activation of the renin-angiotensin system, Sci. Rep 7 (2017) 1–10. doi: 10.1038/s41598-017-03474-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Ruiz-irastorza G, Egurbide MV, Olivares N, Martinez-Berriotxoa A, Aguirre C, Vitamin D deficiency in systemic lupus erythematosus: Prevalence, predictors and clinical consequences, Rheumatology. 47 (2008) 920–923. doi: 10.1093/rheumatology/ken121. [DOI] [PubMed] [Google Scholar]
  • [138].Yang CY, Leung PSC, Adamopoulos IE, Gershwin ME, The implication of vitamin D and autoimmunity: A comprehensive review, Clin. Rev. Allergy Immunol 45 (2013) 217–226. doi: 10.1007/s12016-013-8361-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Mok CC, Vitamin D and systemic lupus erythematosus: An update, Expert Rev. Clin. Immunol 9 (2013) 453–463. doi: 10.1586/eci.13.19. [DOI] [PubMed] [Google Scholar]
  • [140].Amital H, Szekanecz Z, Szücs G, Dankó K, Nagy E, Csépány T, Kiss E, Rovensky J, Tuchynova A, Kozakova D, Doria A, Corocher N, Agmon-Levin N, Barak V, Orbach H, Zandman-Goddard G, Shoenfeld Y, Serum concentrations of 25-OH vitamin D in patients with systemic lupus erythematosus (SLE) are inversely related to disease activity: Is it time to routinely supplement patients with SLE with vitamin D?, Ann. Rheum. Dis 69 (2010) 1155–1157. doi: 10.1136/ard.2009.120329. [DOI] [PubMed] [Google Scholar]
  • [141].Andreoli L, Piantoni S, Dall’Ara F, Allegri F, Meroni PL, Tincani A, Vitamin D and antiphospholipid syndrome, Lupus. 21 (2012) 736–740. doi: 10.1177/0961203312446386. [DOI] [PubMed] [Google Scholar]
  • [142].Piantoni S, Andreoli L, Allegri F, Meroni PL, Tincani A, Low levels of vitamin D are common in primary antiphospholipid syndrome with thrombotic disease, Reumatismo. 64 (2012) 307–13. doi: 10.4081/reumatismo.2012.307. [DOI] [PubMed] [Google Scholar]
  • [143].Petri M, Bello KJ, Fang H, Magder LS, Vitamin D in systemic lupus erythematosus: Modest association with disease activity and the urine protein-to-creatinine ratio, Arthritis Rheum. 65 (2013) 1865–1871. doi: 10.1002/art.37953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Lima GL, Paupitz J, Aikawa NE, Takayama L, Bonfa E, Pereira RMR, Vitamin D Supplementation in Adolescents and Young Adults With Juvenile Systemic Lupus Erythematosus for Improvement in Disease Activity and Fatigue Scores: A Randomized, Double-Blind, Placebo-Controlled Trial, Arthritis Care Res. (Hoboken). 68 (2016) 91–98. doi: 10.1002/acr.22621. [DOI] [PubMed] [Google Scholar]
  • [145].Durcan L, Petri M, Immunomodulators in SLE: Clinical evidence and immunologic actions, J. Autoimmun 74 (2016) 73–84. doi: 10.1016/j.jaut.2016.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Labrie F, Bélanger A, Simard J, Van Luu-The C Labrie, DHEA and peripheral androgen and estrogen formation: intracinology., Ann. N. Y. Acad. Sci 774 (1995) 16–28. doi: 10.1111/j.1749-6632.1995.tb17369.x. [DOI] [PubMed] [Google Scholar]
  • [147].McMurray RW, May W, Sex hormones and systemic lupus erythematosus: review and meta-analysis., Arthritis Rheum. 48 (2003) 2100–10. doi: 10.1002/art.11105. [DOI] [PubMed] [Google Scholar]
  • [148].Lahita RG, Bradlow HL, Ginzler E, Pang S, New M, Low plasma androgens in women with systemic lupus erythematosus., Arthritis Rheum. 30 (1987) 241–8. http://www.ncbi.nlm.nih.gov/pubmed/3032210. [DOI] [PubMed] [Google Scholar]
  • [149].Lucas JA, Ahmed SA, Casey ML, MacDonald PC, Prevention of autoantibody formation and prolonged survival in New Zealand Black/New Zealand White F1mice fed dehydroisoandrosterone, J. Clin. Invest 75 (1985) 2091–2093. doi: 10.1172/JCI111929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Chang DM, Chu SJ, Chen HC, Kuo SY, Lai JH, Dehydroepiandrosterone suppresses interleukin 10 synthesis in women with systemic lupus erythematosus, Ann. Rheum. Dis 63 (2004) 1623–1626. doi: 10.1136/ard.2003.016576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Chen CCG, Parker CR, Adrenal androgens and the immune system, Semin. Reprod. Med 22 (2004) 369–377. doi: 10.1055/s-2004-861553. [DOI] [PubMed] [Google Scholar]
  • [152].Dillon JS, Dehydroepiandrosterone, dehydroepiandrosterone sulfate and related steroids: their role in inflammatory, allergic and immunological disorders., Curr. Drug Targets. Inflamm. Allergy 4 (2005) 377–85. http://www.ncbi.nlm.nih.gov/pubmed/16101547. [DOI] [PubMed] [Google Scholar]
  • [153].Petri MA, Lahita RG, Van Vollenhoven RF, Merrill JT, Schiff M, Ginzler EM, Strand V, Kunz A, Gorelick KJ, Schwartz KE, Effects of prasterone on corticosteroid requirements of women with systemic lupus erythematosus: A double-blind, randomized, placebo-controlled trial, Arthritis Rheum. 46 (2002) 1820–1829. doi: 10.1002/art.10364. [DOI] [PubMed] [Google Scholar]
  • [154].Sanchez-Guerrero J, Fragoso-Loyo HE, Neuwelt CM, Wallace DJ, Ginzler EM, Sherrer YRS, McIlwain HH, Freeman PG, Aranow C, Petri MA, Deodhar AA, Blanton E, Manzi S, Kavanaugh A, Lisse JR, Ramsey-Goldman R, McKay JD, Kivitz AJ, Mease PJ, Winkler AE, Kahl LE, Lee AH, Furie RA, Strand CV, Lou L, Ahmed M, Quarles B, Schwartz KE, Effects of prasterone on bone mineral density in women with active systemic lupus erythematosus receiving chronic glucocorticoid therapy., J. Rheumatol 35 (2008) 1567–1575. [PubMed] [Google Scholar]
  • [155].Cain DW, Cidlowski JA, Immune regulation by glucocorticoids, Nat. Rev. Immunol 17 (2017) 233–247. doi: 10.1038/nri.2017.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Hahn BH, McMahon MA, Wilkinson A, Wallace WD, Daikh DI, Fitzgerald JD, Karpouzas GA, Merrill JT, Wallace DJ, Yazdany J, Ramsey-Goldman R, Singh K, Khalighi M, Choi SI, Gogia M, Kafaja S, Kamgar M, Lau C, Martin WJ, Parikh S, Peng J, Rastogi A, Chen W, Grossman JM, American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis, Arthritis Care Res. 64 (2012) 797–808. doi: 10.1002/acr.21664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Bertsias GK, Tektonidou M, Amoura Z, Aringer M, Bajema I, Berden JHM, Boletis J, Cervera R, Dörner T, Doria A, Ferrario F, Floege J, Houssiau FA, Ioannidis JPA, Isenberg DA, Kallenberg CGM, Lightstone L, Marks SD, Martini A, Moroni G, Neumann I, Praga M, Schneider M, Starra A, Tesar V, Vasconcelos C, Van Vollenhoven RF, Zakharova H, Haubitz M, Gordon C, Jayne D, Boumpas DT, Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis, Ann. Rheum. Dis 71 (2012) 1771–1782. doi: 10.1136/annrheumdis-2012-201940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Condon MB, Ashby D, Pepper RJ, Cook HT, Levy JB, Griffith M, Cairns TD, Lightstone L, Prospective observational single-centre cohort study to evaluate the effectiveness of treating lupus nephritis with rituximab and mycophenolate mofetil but no oral steroids, Ann. Rheum. Dis 72 (2013) 1280–1286. doi: 10.1136/annrheumdis-2012-202844. [DOI] [PubMed] [Google Scholar]
  • [159].Dooley M, Pendergraft WI, Ginzler E, Olsen N, Tumlin J, Rovin B, Houssiau F, Wofsy D, Isenberg D, Solomons N, Huizinga R, Speed of Remission with the Use of Voclosporin, MMF and Low Dose Steroids: Results of a Global Lupus Nephritis Study [abstract], Arthritis Rheumatol. 68 (suppl (2016). https://acrabstracts.org/abstract/speed-of-remission-with-the-use-of-voclosporin-mmf-and-low-dose-steroids-results-of-a-global-lupus-nephritis-study/. [Google Scholar]
  • [160].Singh JA, Hossain A, Kotb A, Oliveira A, Mudano AS, Grossman J, Winthrop K, Wells GA, Treatments for lupus nephritis: A systematic review and network metaanalysis, J. Rheumatol 43 (2016) 1801–1815. doi: 10.3899/jrheum.160041. [DOI] [PubMed] [Google Scholar]
  • [161].Gladman DD, Urowitz MB, Rahman P, Ibañez D, Tam L-S, Accrual of organ damage over time in patients with systemic lupus erythematosus., J. Rheumatol 30 (2003) 1955–9. http://www.ncbi.nlm.nih.gov/pubmed/12966597. [PubMed] [Google Scholar]
  • [162].Magder LS, Petri M, Incidence of and risk factors for adverse cardiovascular events among patients with systemic lupus erythematosus, Am. J. Epidemiol 176 (2012) 708–719. doi: 10.1093/aje/kws130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Thamer M, Hernán MA, Zhang Y, Cotter D, Petri M, Prednisone, lupus activity, and permanent organ damage, J. Rheumatol 36 (2009) 560–564. doi: 10.3899/jrheum.080828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Zonana-Nacach A, Barr SG, Magder LS, Petri M, Damage in systemic lupus erythematosus and its association with corticosteroids, Arthritis Rheum. 43 (2000) 1801–1808. doi:. [DOI] [PubMed] [Google Scholar]
  • [165].Danowski A, Magder L, Petri M, Flares in Lupus: Outcome Assessment Trial (FLOAT), a comparison between oral methylprednisolone and intramuscular triamcinolone, J. Rheumatol 33 (2006) 57–60. [PubMed] [Google Scholar]
  • [166].Frangou EA, Bertsias G, Boumpas DT, Cytotoxic-Immunosuppressive Drug Treatment, in: Tsokos GC (Ed.), Syst. Lupus Erythematosus Basic, Appl. Clin. Asp, Elsevier, 2016. [Google Scholar]
  • [167].Steinberg AD, Decker JL, A double-blind controlled trial comparing cyclophosphamide, azathioprine and placebo in the treatment of lupus glomerulonephritis, Arthritis Rheum. 17 (1974) 923–937. doi: 10.1002/art.1780170602. [DOI] [PubMed] [Google Scholar]
  • [168].Austin HA, Klippel JH, Balow JE, Le Riche NGH, Steinberg AD, Plotz PH, Decker JL, Therapy of Lupus Nephritis, N. Engl. J. Med 314 (1986) 614–619. doi: 10.1056/NEJM198603063141004. [DOI] [PubMed] [Google Scholar]
  • [169].McCune WJ, Golbus J, Zeldes W, Bohlke P, Dunne R, Fox DA, Clinical and Immunologic Effects of Monthly Administration of Intravenous Cyclophosphamide in Severe Systemic Lupus Erythematosus, N. Engl. J. Med 318 (1988) 1423–1431. doi: 10.1056/NEJM198806023182203. [DOI] [PubMed] [Google Scholar]
  • [170].Tokunaga M, Saito K, Kawabata D, Imura Y, Fujii T, Nakayamada S, Tsujimura S, Nawata M, Iwata S, Azuma T, Mimori T, Tanaka Y, Efficacy of rituximab (anti-CD20) for refractory systemic lupus erythematosus involving the central nervous system, Ann. Rheum. Dis 66 (2007) 470–475. doi: 10.1136/ard.2006.057885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Magro-Checa C, Zirkzee EJ, Huizinga TW, Steup-Beekman GM, Management of Neuropsychiatric Systemic Lupus Erythematosus: Current Approaches and Future Perspectives, Drugs. 76 (2016) 459–483. doi: 10.1007/s40265-015-0534-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Mok CC, Con: Cyclophosphamide for the treatment of lupus nephritis, Nephrol. Dial. Transplant 31 (2016) 1053–1057. doi: 10.1093/ndt/gfw068. [DOI] [PubMed] [Google Scholar]
  • [173].Tiede I, Fritz G, Strand S, Poppe D, Dvorsky R, Strand D, Lehr HA, Wirtz S, Becker C, Atreya R, Mudter J, Hildner K, Bartsch B, Holtmann M, Blumberg R, Walczak H, Iven H, Galle PR, Ahmadian MR, Neurath MF, CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes., J. Clin. Invest 111 (2003) 1133–45. doi: 10.1172/JCI200316432.Introduction. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Ginzler E, Sharon E, Diamond H, Kaplan D, Long-term maintenance therapy with azathioprine in systemic lupus erythematosus., Arthritis Rheum. 18 (1975) 27–34. http://www.ncbi.nlm.nih.gov/pubmed/1115745. [DOI] [PubMed] [Google Scholar]
  • [175].Sztejnbok M, Stewart A, Diamond H, Kaplan D, Azathioprine in the treatment of systemic lupus erythematosus. A controlled study, Arthritis Rheum. 14 (1971) 639–645. [DOI] [PubMed] [Google Scholar]
  • [176].Grootscholten C, Ligtenberg G, Hagen EC, Van Den Wall Bake AWL, De Glas-Vos JW, Bijl M, Assmann KJ, Bruijn JA, Weening JJ, Van Houwelingen HC, Derksen RHWM, Berden JHM, Azathioprine/methylprednisolone versus cyclophosphamide in proliferative lupus nephritis. A randomized controlled trial, Kidney Int. 70 (2006) 732–742. doi: 10.1038/sj.ki.5001630. [DOI] [PubMed] [Google Scholar]
  • [177].Grootscholten C, Bajema IM, Florquin S, Steenbergen EJ, Peutz-Kootstra CJ, Goldschmeding R, Bijl M, Hagen EC, Van Houwelingen HC, Derksen RHWM, Berden JHM, Treatment with cyclophosphamide delays the progression of chronic lesions more effectively than does treatment with azathioprine plus methylprednisolone in patients with proliferative lupus nephritis, Arthritis Rheum. 56 (2007) 924–937. doi: 10.1002/art.22449. [DOI] [PubMed] [Google Scholar]
  • [178].Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, Eitner F, Appel GB, Contreras G, Lisk L, Solomons N, Mycophenolate versus Azathioprine as Maintenance Therapy for Lupus Nephritis, N. Engl. J. Med 365 (2011) 1886–1895. doi: 10.1056/NEJMoa1014460. [DOI] [PubMed] [Google Scholar]
  • [179].Houssiau FA, D’Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, Fiehn C, Garrido EDR, Gilboe IM, Tektonidou M, Blockmans D, Ravelingien I, Le Guern V, Depresseux G, Guillevin L, Cervera R, Abramowicz D, Atzeni F, Danieli MG, De Clercq L, De Keyser F, Delahousse M, Espinosa G, Golstein M, Hirsch M, Karras A, Lang P, Marchal M, Marinho A, Max R, Peeters P, Petera P, Quémeneur T, Raeman F, Sarzi-Puttini P, De Santis LV, Verresen L, Weiss L, Westhovens R, Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: Results from the MAINTAIN Nephritis Trial, Ann. Rheum. Dis 69 (2010) 2083–2089. doi: 10.1136/ard.2010.131995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Griffiths B, Emery P, Ryan V, Isenberg D, Akil M, Thompson R, Maddison P, Griffiths ID, Lorenzi A, Miles S, Situnayake D, Teh LS, Plant M, Hallengren C, Nived O, Sturfelt G, Chakravarty K, Tait T, Gordon C, The BILAG multi-centre open randomized controlled trial comparing ciclosporin vs azathioprine in patients with severe SLE., Rheumatology (Oxford). 49 (2010) 723–732. doi: 10.1093/rheumatology/kep396. [DOI] [PubMed] [Google Scholar]
  • [181].Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, Vidal X, Mitjavila F, Castro Salomó A, Cuquet Pedragosa J, Ortiz-Santamaria V, Mauri Plana M, Cortés-Hernández J, Enteric-coated mycophenolate sodium versus azathioprine in patients with active systemic lupus erythematosus: A randomised clinical trial, Ann. Rheum. Dis 76 (2017) 1575–1582. doi: 10.1136/annrheumdis-2016-210882. [DOI] [PubMed] [Google Scholar]
  • [182].Lateef A, Petri M, Systemic Lupus Erythematosus and Pregnancy, Rheum. Dis. Clin. North Am 43 (2017) 215–226. doi: 10.1016/j.rdc.2016.12.009. [DOI] [PubMed] [Google Scholar]
  • [183].De Boer NKH, Jarbandhan SVA, De Graaf P, Mulder CJJ, Van Elburg RM, Van Bodegraven AA, Azathioprine use during pregnancy: Unexpected intrauterine exposure to metabolites, Am. J. Gastroenterol 101 (2006) 1390–1392. doi: 10.1111/j.1572-0241.2006.00538.x. [DOI] [PubMed] [Google Scholar]
  • [184].Brown PM, Pratt AG, Isaacs JD, Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers, Nat. Rev. Rheumatol 12 (2016) 731–742. doi: 10.1038/nrrheum.2016.175. [DOI] [PubMed] [Google Scholar]
  • [185].Miescher PA, Riethmueller D, Diagnosis and treatment of systemic lupus erythematosus, Semin. Hematol 2 (1965) 1–28. http://www.ncbi.nlm.nih.gov/pubmed/14268735. [PubMed] [Google Scholar]
  • [186].Sakthiswary RSE, Methotrexate in systemic lupus erythematosus: a systematic review of its efficacy., Lupus. 23(3) (2014) 225–35. http://www.ncbi.nlm.nih.gov/pubmed/14268735. [DOI] [PubMed] [Google Scholar]
  • [187].Fortin PR, Abrahamowicz M, Ferland D, Lacaille D, Smith CD, Zummer M, Steroid-sparing effects of methotrexate in systemic lupus erythematosus: A double-blind, randomized, placebo-controlled trial, Arthritis Rheum. 59 (2008) 1796–1804. doi: 10.1002/art.24068. [DOI] [PubMed] [Google Scholar]
  • [188].Allison A, Mechanisms of action of mycophenolate mofetil, Lupus. 14 (2005) 2–8. doi: 10.1191/0961203305LU2109OA. [DOI] [PubMed] [Google Scholar]
  • [189].Chan TM, Li FK, Tang CSO, Wong RWS, Fang GX, Ji YL, Lau CS, Wong AKM, Tong MKL, Chan KW, Lai KN, Efficacy of Mycophenolate Mofetil in Patients with Diffuse Proliferative Lupus Nephritis, N. Engl. J. Med 343 (2000) 1156–1162. doi: 10.1056/NEJM200010193431604. [DOI] [PubMed] [Google Scholar]
  • [190].Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT, Petri M, Gilkeson GS, Wallace DJ, Weisman MH, Appel GB, Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis., N. Engl. J. Med 353 (2005) 2219–28. doi: 10.1056/NEJMoa043731. [DOI] [PubMed] [Google Scholar]
  • [191].Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, Li L-S, Mysler E, Sanchez-Guerrero J, Solomons N, Wofsy D, Mycophenolate Mofetil versus Cyclophosphamide for Induction Treatment of Lupus Nephritis, J. Am. Soc. Nephrol 20 (2009) 1103–1112. doi: 10.1681/ASN.2008101028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Tunnicliffe DJ, Palmer SC, Henderson L, Masson P, Craig JC, Tong A, Singh-Grewal D, Flanc RS, Roberts MA, Webster AC, Strippoli GF, Immunosuppressive treatment for proliferative lupus nephritis, Cochrane Database Syst. Rev (2018). doi: 10.1002/14651858.CD002922.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Mok CC, Mycophenolate mofetil for non-renal manifestations of systemic lupus erythematosus: A systematic review, Scand. J. Rheumatol 36 (2007) 329–337. doi: 10.1080/03009740701607042. [DOI] [PubMed] [Google Scholar]
  • [194].Ginzler EM, Wofsy D, Isenberg D, Gordon C, Lisk L, Dooley MA, Nonrenal disease activity following mycophenolate mofetil or intravenous cyclophosphamide as induction treatment for lupus nephritis: Findings in a multicenter, prospective, randomized, open-label, parallel-group clinical trial, Arthritis Rheum. 62 (2010) 211–221. doi: 10.1002/art.25052. [DOI] [PubMed] [Google Scholar]
  • [195].Mok CC, Calcineurin inhibitors in systemic lupus erythematosus, Best Pract. Res. Clin. Rheumatol 31 (2017) 429–438. doi: 10.1016/j.berh.2017.09.010. [DOI] [PubMed] [Google Scholar]
  • [196].Ikezumi Y, Kanno K, Koike H, Tomita M, Uchiyama M, Shimizu F, Kawachi H, FK506 ameliorates proteinuria and glomerular lesions induced by anti-Thy 1.1 monoclonal antibody 1-22-3, Kidney Int. 61 (2002) 1339–1350. doi: 10.1046/j.1523-1755.2002.00259.x. [DOI] [PubMed] [Google Scholar]
  • [197].Liao R, Liu Q, Zheng Z, Fan J, Peng W, Kong Q, He H, Yang S, Chen W, Tang X, Yu X, Tacrolimus protects podocytes from injury in lupus nephritis partly by stabilizing the cytoskeleton and inhibiting podocyte apoptosis, PLoS One. 10 (2015) 1–16. doi: 10.1371/journal.pone.0132724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Faul C, Donnelly M, Merscher-Gomez S, Chang YH, Franz S, Delfgaauw J, Chang JM, Choi HY, Campbell KN, Kim K, Reiser J, Mundel P, The actin cytoskeleton of kidney podocytes is a direct target of the antiproteinuric effect of cyclosporine A, Nat. Med 14 (2008) 931–938. doi: 10.1038/nm.1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Krämer BK, Montagnino G, del Castillo D, Margreiter R, Sperschneider H, Olbricht CJ, Krüger B, Ortuño J, Köhler H, Kunzendorf U, Stummvoll HK, Tabernero JM, Mühlbacher F, Rivero M, Arias M, Dietl KH, del Castillo D, Pascual J, Olbrícht C, Kammerl M, Krüger B, Köhler K, Sester U, Mühlbacher F, Kunzendorf U, Hauser I, Ponticelli C, Bonomini V, Tabernero JM, Rivero M, Ancona E, Calconi G, Cambi V, la Greca G, Mosca F, Selvaggi F, Meurisse M, Detry O, González Molina M, Piccoli G, Salvadori M, Schilling M, Raedelli C, Arns W, Suarez F, Rodríguez Algarra G, Asensio C, Maiorca R, Ancona G, Sparacino V, Morales JM, Gutierrez JA, Lauzurica R, Verpooten GA, Sennesael J, Heidecke C, García J, Capdevila L, Abendroth D, Biesenbach G, Altieri P, Sorba G, Valente U, Castagneto M, Purroy A, Duhoux P, Kremar C, Stiegler G, Schleibner S, Efficacy and safety of tacrolimus compared with cyclosporin A microemulsion in renal transplantation: 2 year follow-up results, Nephrol. Dial. Transplant 20 (2005) 968–973. doi: 10.1093/ndt/gfh739. [DOI] [PubMed] [Google Scholar]
  • [200].Li X, Ren H, Zhang Q, Zhang W, Wu X, Xu Y, Shen P, Chen N, Mycophenolate mofetil or tacrolimus compared with intravenous cyclophosphamide in the induction treatment for active lupus nephritis, Nephrol. Dial. Transplant 27 (2012) 1467–1472. doi: 10.1093/ndt/gfr484. [DOI] [PubMed] [Google Scholar]
  • [201].Chen W, Tang X, Liu Q, Chen W, Fu P, Liu F, Liao Y, Yang Z, Zhang J, Chen J, Lou T, Fu J, Kong Y, Liu Z, Fan A, Rao S, Li Z, Yu X, Short-term outcomes of induction therapy with tacrolimus versus cyclophosphamide for active lupus nephritis: A multicenter randomized clinical trial, Am. J. Kidney Dis 57 (2011) 235–244. doi: 10.1053/j.ajkd.2010.08.036. [DOI] [PubMed] [Google Scholar]
  • [202].Závada J, Pešičkova SS, Ryšavá R, Olejárova M, Horák P, Hrnčíř Z, Rychlík I, Havrda M, Vítová J, Lukáč J, Rovenský J, Tegzova D, Böhmova J, Zadražil J, Hána J, Dostál C, Tesar V, Cyclosporine A or intravenous cyclophosphamide for lupus nephritis: The Cyclofa-Lune study, Lupus. 19 (2010) 1281–1289. doi: 10.1177/0961203310371155. [DOI] [PubMed] [Google Scholar]
  • [203].Závada J, Pešičková SS, Ryšavá R, Horák P, Hrnčíř Z, Lukáč J, Rovenský J, Vítová J, Havrda M, Rychlík I, Böhmova J, Vlasáková V, Slatinská J, Zadražil J, Olejárová M, Tegzova D, Tesar V, Extended follow-up of the CYCLOFA-LUNE trial comparing two sequential induction and maintenance treatment regimens for proliferative lupus nephritis based either on cyclophosphamide or on cyclosporine A, Lupus. 23 (2014) 69–74. doi: 10.1177/0961203313511555. [DOI] [PubMed] [Google Scholar]
  • [204].Hannah J, Casian A, D’Cruz D, Tacrolimus use in lupus nephritis: A systematic review and meta-analysis, Autoimmun. Rev 15 (2016) 93–101. doi: 10.1016/j.autrev.2015.09.006. [DOI] [PubMed] [Google Scholar]
  • [205].Mok CC, Ying KY, Yim CW, Siu YP, Tong KH, To CH, Ng WL, Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: A randomised controlled trial and long-term follow-up, Ann. Rheum. Dis 75 (2016) 30–36. doi: 10.1136/annrheumdis-2014-206456. [DOI] [PubMed] [Google Scholar]
  • [206].Lanata CM, Mahmood T, Fine DM, Petri M, Combination therapy of mycophenolate mofetil and tacrolimus in lupus nephritis, Lupus. 19 (2010) 935–940. doi: 10.1177/0961203310365714. [DOI] [PubMed] [Google Scholar]
  • [207].Cortés-Hernández J, Torres-Salido MT, Medrano AS, Tarrés MV, Ordi-Ros J, Long-term outcomes Mycophenolate mofetil treatment for lupus nephritis with addition of tacrolimus for resistant cases, Nephrol. Dial. Transplant 25 (2010) 3939–3948. doi: 10.1093/ndt/gfq322. [DOI] [PubMed] [Google Scholar]
  • [208].Mok CC, To CH, Yu KL, Ho LY, Combined low-dose mycophenolate mofetil and tacrolimus for lupus nephritis with suboptimal response to standard therapy: A 12-month prospective study, Lupus. 22 (2013) 1135–1141. doi: 10.1177/0961203313502864. [DOI] [PubMed] [Google Scholar]
  • [209].Liu Z, Zhang H, Liu Z, Xing C, Fu P, Ni Z, Chen J, Lin H, Liu F, He Y, He Y, Miao L, Chen N, Li Y, Gu Y, Shi W, Hu W, Liu Z, Bao H, Zeng C, Zhou M, Multitarget therapy for induction treatment of lupus nephritis: A randomized trial, Ann. Intern. Med 162 (2015) 18–26. doi: 10.7326/M14-1030. [DOI] [PubMed] [Google Scholar]
  • [210].Moroni G, Doria A, Mosca M, Alberighi ODC, Ferraccioli G, Todesco S, Manno C, Altieri P, Ferrara R, Greco S, Ponticelli C, A randomized pilot trial comparing cyclosporine and azathioprine for maintenance therapy in diffuse lupus nephritis over four years., Clin. J. Am. Soc. Nephrol 1 (2006) 925–932. doi: 10.2215/CJN.02271205. [DOI] [PubMed] [Google Scholar]
  • [211].Chen W, Liu Q, Chen W, 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. 21 (2012) 944–952. doi: 10.1177/0961203312442259. [DOI] [PubMed] [Google Scholar]
  • [212].Radhakrishnan J, Kunis CL, D’Agati V, Appel GB, Cyclosporine treatment of lupus membranous nephropathy., Clin. Nephrol 42 (1994) 147–54. http://www.ncbi.nlm.nih.gov/pubmed/7994932. [PubMed] [Google Scholar]
  • [213].Yap DY, Yu X, Chen XM, Lu F, Chen N, Li XW, Tang CS, Chan TM, Pilot 24 month study to compare mycophenolate mofetil and tacrolimus in the treatment of membranous lupus nephritis with nephrotic syndrome, Nephrology. 17 (2012) 352–357. doi: 10.1111/j.1440-1797.2012.01574.x. [DOI] [PubMed] [Google Scholar]
  • [214].Hallegua D, Wallace DJ, Metzger AL, Rinaldi RZ, Klinenberg JR, Cyclosporine for lupus membranous nephritis: Experience with ten patients and review of the literature, Lupus. 9 (2000) 241–251. doi: 10.1191/096120300680198935. [DOI] [PubMed] [Google Scholar]
  • [215].Austin HA, Illei GG, Braun MJ, Balow JE, Randomized, controlled trial of prednisone, cyclophosphamide, and cyclosporine in lupus membranous nephropathy., J. Am. Soc. Nephrol 20 (2009) 901–11. doi: 10.1681/ASN.2008060665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Busque S, Cantarovich M, Mulgaonkar S, Gaston R, Gaber AO, Mayo PR, Ling S, Huizinga RB, Meier-Kriesche HU, The PROMISE study: A phase 2b multicenter study of voclosporin (ISA247) versus tacrolimus in de Novo kidney transplantation, Am. J. Transplant 11 (2011) 2675–2684. doi: 10.1111/j.1600-6143.2011.03763.x. [DOI] [PubMed] [Google Scholar]
  • [217].Parikh S, Pendergraft W, Tumlin J, Saxena R, Solomons N, Huizinga R, Treatment of active Lupus nephritis with Voclosporiim: 48 week data from the aura-LV study, Am. J. Kidney Dis. Conf. Natl. Kidney Found. 2017 Spring Clin. Meet. United States 69 (2017) A2. doi: 10.1053/j.ajkd.2017.03.005. [DOI] [Google Scholar]
  • [218].Cobo-Ibáñez T, Loza-Santamaría E, Pego-Reigosa JM, Marqués AO, Rúa-Figueroa Í, Fernández-Nebro A, Cáliz RC, López Longo FJ, Muñoz-Fernández S, Efficacy and safety of rituximab in the treatment of non-renal systemic lupus erythematosus: A systematic review, Semin. Arthritis Rheum 44 (2015) 175–185. doi: 10.1016/j.semarthrit.2014.04.002. [DOI] [PubMed] [Google Scholar]
  • [219].Lan L, Han F, Chen J, Efficacy and safety of rituximab therapy for systemic lupus erythematosus: a systematic review and meta-analysis, J. Zhejiang Univ. Sci. B 13 (2012) 731–744. doi: 10.1631/jzus.B1200057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].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. 62 (2010) 222–233. doi: 10.1002/art.27233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, Maciuca R, Zhang D, Garg JP, Brunetta P, Appel G, LUNAR Investigator Group, Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study., Arthritis Rheum. 64 (2012) 1215–1226. doi: 10.1002/art.34359. [DOI] [PubMed] [Google Scholar]
  • [222].John Looney R, Anolik J, Sanz I, A perspective on B-cell-targeting therapy for SLE, Mod. Rheumatol 20 (2010) 1–10. doi: 10.1007/s10165-009-0213-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Cambridge G, Isenberg DA, Edwards JCW, Leandro MJ, Migone T-S, Teodorescu M, Stohl W, B cell depletion therapy in systemic lupus erythematosus: relationships among serum B lymphocyte stimulator levels, autoantibody profile and clinical response., Ann. Rheum. Dis 67 (2008) 1011–6. doi: 10.1136/ard.2007.079418. [DOI] [PubMed] [Google Scholar]
  • [224].Ehrenstein MR, Wing C, The BAFFling effects of rituximab in lupus: Danger ahead?, Nat. Rev. Rheumatol 12 (2016) 367–372. doi: 10.1038/nrrheum.2016.18. [DOI] [PubMed] [Google Scholar]
  • [225].Carter LM, Isenberg DA, Ehrenstein MR, Elevated serum BAFF levels are associated with rising anti-double-stranded DNA antibody levels and disease flare following B cell depletion therapy in systemic lupus erythematosus, Arthritis Rheum. 65 (2013) 2672–2679. doi: 10.1002/art.38074. [DOI] [PubMed] [Google Scholar]
  • [226].Kraaij T, Kamerling SWA, de Rooij ENM, van Daele PLA, Bredewold OW, Bakker JA, Bajema IM, Scherer HU, Toes REM, Huizinga TJW, Rabelink TJ, van Kooten C, Teng YKO, The NET-effect of combining rituximab with belimumab in severe systemic lupus erythematosus, J. Autoimmun 91 (2018) 45–54. doi: 10.1016/j.jaut.2018.03.003. [DOI] [PubMed] [Google Scholar]
  • [227].Aranow C, Dall’Era M, Byron M, Ding L, Smilek D, Diamond B, Wofsy D, FRI0305 Phase 2 trial of induction therapy with anti-cd20 (RITUXIMAB) followed by maintenance therapy with anti-baff (BELIMUMAB) in patients with active lupus nephritis, Ann. Rheum. Dis 77 (2018) 690 LP–690. http://ard.bmj.com/content/77/Suppl_2/690.1.abstract.29343507 [Google Scholar]
  • [228].MacKay F, Schneider P, Cracking the BAFF code, Nat. Rev. Immunol 9 (2009) 491–502. doi: 10.1038/nri2572. [DOI] [PubMed] [Google Scholar]
  • [229].Treml JF, Hao Y, Stadanlick JE, Cancro MP, The BLyS family: Toward a molecular understanding of B cell homeostasis, Cell Biochem. Biophys 53 (2009) 1–16. doi: 10.1007/s12013-008-9036-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Petri MA, van Vollenhoven RF, Buyon J, Levy RA, Navarra SV, Cervera R, Zhong ZJ, Freimuth WW, Baseline Predictors of Systemic Lupus Erythematosus Flares: Data From the Combined Placebo Groups in the Phase III Belimumab Trials, Arthritis Rheum. 65 (2013) 2143–2153. doi: 10.1002/art.37995. [DOI] [PubMed] [Google Scholar]
  • [231].Groom JR, Fletcher CA, Walters SN, Grey ST, Watt SV, Sweet MJ, Smyth MJ, Mackay CR, Mackay F, BAFF and MyD88 signals promote a lupuslike disease independent of T cells, J. Exp. Med 204 (2007) 1959–1971. doi: 10.1084/jem.20062567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzová D, Sanchez-Guerrero J, Schwarting A, Merrill JT, Chatham WW, Stohl W, Ginzler EM, Hough DR, Zhong ZJ, Freimuth W, Van Vollenhoven RF, 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. 63 (2011) 3918–3930. doi: 10.1002/art.30613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Navarra SV, Guzmán RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, Li EKM, Thomas M, Kim HY, León MG, Tanasescu C, Nasonov E, Lan JL, Pineda L, Zhong ZJ, Freimuth W, Petri MA, Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: A randomised, placebo-controlled, phase 3 trial, Lancet. 377 (2011) 721–731. doi: 10.1016/S0140-6736(10)61354-2. [DOI] [PubMed] [Google Scholar]
  • [234].Van Vollenhoven RF, Petri MA, Cervera R, Roth DA, Ji BN, Kleoudis CS, Zhong ZJ, Freimuth W, Belimumab in the treatment of systemic lupus erythematosus: High disease activity predictors of response, Ann. Rheum. Dis 71 (2012) 1343–1349. doi: 10.1136/annrheumdis-2011-200937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, Illei GG, Drappa J, Wang L, Yoo S, Anifrolumab, an Anti–Interferon-α Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus, Arthritis Rheumatol. 69 (2017) 376–386. doi: 10.1002/art.39962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].AstraZeneca, Update on TULIP 1 Phase III trial for anifrolumab in systemic lupus erythematosus, (n.d.). https://www.astrazeneca.com/media-centre/press-releases/2018/update-on-tulip-1-phase-iii-trial-for-anifrolumab-in-systemic-lupus-erythematosus-31082018.html.
  • [237].Yang XO, Panopoulos AD, Nurieva R, Seon HC, Wang D, Watowich SS, Dong C, STAT3 regulates cytokine-mediated generation of inflammatory helper T cells, J. Biol. Chem 282 (2007) 9358–9363. doi: 10.1074/jbc.C600321200. [DOI] [PubMed] [Google Scholar]
  • [238].Ma C, Avery D, Chan A, Batten M, Bustamante J, Boisson-Dupuis S, Arkwright P, Kreins A, Averbuch D, Engelhard D, Magdorf K, Kilic S, Minegishi Y, Nonoyama S, French M, Choo S, Smart J, Peake J, Wong M, Gray P, Cook M, Fulcher D, Casanova J-L, Deenick E, Tangye S, Functional STAT3 deficiency compromises the generation of human T follicular helper cells., Blood. 119 (2012) 3997–4008. doi: 10.1182/blood-2011-11-392985.The. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Ueno H, Banchereau J, Vinuesa CG, Pathophysiology of T follicular helper cells in humans and mice, Nat. Immunol 16 (2015) 142–152. doi: 10.1038/ni.3054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Shaltout AS, Sayed D, Badary MS, Nafee AM, El Zohri MH, Bakry R, Ahmed SH, Effect of IL6 and IL23 on double negative T cells and anti ds-DNA in systemic lupus erythematosus patients, Hum. Immunol 77 (2016) 937–943. doi: 10.1016/j.humimm.2016.06.007. [DOI] [PubMed] [Google Scholar]
  • [241].Dai H, He F, Tsokos GC, Kyttaris VC, IL-23 Limits the Production of IL-2 and Promotes Autoimmunity in Lupus, J. Immunol 199 (2017) 903–910. doi: 10.4049/jimmunol.1700418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].van Vollenhoven R, Hahn B, Tsokos G, Wagner C, Lipsky P, Hsu B, Chevrier M, Gordon R, Triebel M, Rose S, S7A:8 Efficacy and safety of ustekinumab, an interleukin 12/23 inhibitor, in patients with active systemic lupus erythematosus: results of a phase 2, randomised placebo-controlled study, Lupus Sci. Med 5 (2018) A28 LP–A29. http://lupus.bmj.com/content/5/Suppl_1/A28.abstract. [DOI] [PubMed] [Google Scholar]
  • [243].Gadina M, Johnson C, Schwartz D, Bonelli M, Hasni S, Kanno Y, Changelian P, Laurence A, O’Shea JJ, Translational and clinical advances in JAK-STAT biology: The present and future of jakinibs., J. Leukoc. Biol (2018) 1–16. doi: 10.1002/JLB.5RI0218-084R. [DOI] [PubMed] [Google Scholar]
  • [244].Wallace DJ, Furie RA, Tanaka Y, Kalunian KC, Mosca M, Petri MA, Dörner T, Cardiel MH, Bruce IN, Gomez E, Carmack T, DeLozier AM, Janes JM, Linnik MD, de Bono S, Silk ME, Hoffman RW, Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial, Lancet. 392 (2018) 222–231. doi: 10.1016/S0140-6736(18)31363-1. [DOI] [PubMed] [Google Scholar]
  • [245].Dall’Era M, Chakravarty E, Wallace D, Genovese M, Weisman M, Kavanaugh A, Kalunian K, Dhar P, Vincent E, Pena-Rossi C, Wofsy D, Alum N, Dubois A, Kinnman N, Picard M, Bortolotti A, O’Grady L, Hill J, Nestorov I, Salmon E, Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: Results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial, Arthritis Rheum. 56 (2007) 4142–4150. doi: 10.1002/art.23047. [DOI] [PubMed] [Google Scholar]
  • [246].Merrill JT, Wallace DJ, Wax S, Kao A, Fraser PA, Chang P, Isenberg D, Efficacy and Safety of Atacicept in Patients With Systemic Lupus Erythematosus: Results of a Twenty-Four–Week, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Arm, Phase IIb Study, Arthritis Rheumatol. 70 (2018) 266–276. doi: 10.1002/art.40360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Wallace DJ, Improving the prognosis of SLE without prescribing lupus drugs and the primary care paradox, Lupus. 17 (2008) 91–92. doi: 10.1177/0961203307086267. [DOI] [PubMed] [Google Scholar]
  • [248].Kuhn A, Gensch K, Haust M, Meuth AM, Boyer F, Dupuy P, Lehmann P, Metze D, Ruzicka T, Photoprotective effects of a broad-spectrum sunscreen in ultraviolet-induced cutaneous lupus erythematosus: A randomized, vehicle-controlled, double-blind study, J. Am. Acad. Dermatol 64 (2011) 37–48. doi: 10.1016/j.jaad.2009.12.053. [DOI] [PubMed] [Google Scholar]
  • [249].Wolfe F, Petri M, Alarcón GS, Goldman J, Chakravarty EF, Katz RS, Karlson EW, Fibromyalgia, systemic lupus erythematosus (SLE), and evaluation of SLE activity, J. Rheumatol 36 (2009) 82–88. doi: 10.3899/jrheum.080212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Torrente-Segarra V, Salman-Monte TC, Rúa-Figueroa I, Pérez-Vicente S, López-Longo FJ, Galindo-Izquierdo M, Calvo-Alén J, Olivé-Marqués A, Ibáñez-Ruán J, Horcada L, Sánchez-Atrio A, Montilla C, Rodríguez-Gómez M, Díez-álvarez E, Martinez-Taboada V, Andreu JL, Fernández-Berrizbeitia O, Hernández-Beriain JA, Gantes M, Hernández-Cruz B, Pecondón-Español ángela, Marras C, Bonilla G, Pego-Reigosa JM, Blanco R, Vela-Casasempere P, Melero-González R, Otón-Sánchez T, Tornero-Muriel E, Uriarte-Isacelaya E, Freire-González M, Fito-Manteca MC, Fernández-Nebro A, Narváez J, Zea-Mendoza A, Rosas-Gómez de Salazar JC, Ibáñez-Barceló M, Pérez-Venegas JJ, de la Torre Ortega I, Pérez LC, Delgado PC, Rodríguez-Almaraz E, Heredia ES, Soler GS, Ramírez CS, Senabre Gallego JM, Collado MA, Bernal JA, Vilamajó IR, Mas AJ, Murillo C, Barranco IC, Melchor EG, Fontova JC, Manrique MG, Lencastre CG, Martínez-Losa MM, Almagro RM, González-Gay Mantecón MA, Martíny IP, del Carmen Bejerano M, Blanco IV, Moreira B, Aurrecoechea E, Jimeno TR, Zamorano ángeles A., Magro C, álvarez ER, Arruabarrena CE, ángeles M Mérida A, Egües Dubuc CA, Fanlo JC, álvarez ED, Vitovi C, Robles AL, Rodríguez TV, Oyarzábal MVI, ángeles M López B, Romero Barco CM, López JAM, Pernaute OS, de Vicuña Pinedo TG, Expósito MV, de la Peña PG, Rubio SR, González Martín JJ, Gómez AP, Bohórquez C, Hajkhan AM, Turrión Nieves AI, Lois Iglesias AJ, Boteanu AL, Gamir MLG, Alberti PR, Fernández SM, Oliva MR, Stoye, Rodríguez íñigo H., Rodríguez CM, de Aspe de la Iglesia B, González RL, Sarabia FN, Toyos Sáenz de Miera FJ, de la Fuente JLM, Montañés JU, Sánchez RH, Pérez RM, Lozano BR, úcar Angulo E, Lucea MER, Domínguez LL, Alegre Sancho JJ, de La Morena Barrio I, Valls E, Ruiz JM, Quevedo Vila VE, Machín, Nóvoa J, Fernández LS, Fibromyalgia prevalence and related factors in a large registry of patients with systemic lupus erythematosus, Clin. Exp. Rheumatol 34 (2016) 40–47. [PubMed] [Google Scholar]
  • [251].Wang C, Schmid CH, Rones R, Kalish R, Yinh J, Goldenberg DL, Lee Y, McAlindon T, A randomized trial of tai chi for fibromyalgia., N. Engl. J. Med 363 (2010) 743–54. doi: 10.1056/NEJMoa0912611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Gomez-Pinilla F, Hillman C, The influence of exercise on cognitive abilities., Compr. Physiol 3 (2013) 403–28. doi: 10.1002/cphy.c110063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Tench CM, McCarthy J, McCurdie I, White PD, D’Cruz DP, Fatigue in systemic lupus erythematosus: a randomized controlled trial of exercise., Rheumatology (Oxford). 42 (2003) 1050–4. doi: 10.1093/rheumatology/keg289. [DOI] [PubMed] [Google Scholar]
  • [254].Giannelou M, Mavragani CP, Cardiovascular disease in systemic lupus erythematosus: A comprehensive update, J. Autoimmun 82 (2017) 1–12. doi: 10.1016/j.jaut.2017.05.008. [DOI] [PubMed] [Google Scholar]
  • [255].Petri M, Roubenoff R, Dallal GE, Nadeau MR, Selhub J, Rosenberg IH, Plasma homocysteine as a risk factor for atherothrombotic events in systemic lupus erythematosus, Lancet. 348 (1996) 1120–1124. doi: 10.1016/S0140-6736(96)03032-2. [DOI] [PubMed] [Google Scholar]
  • [256].Homocysteine Studies Collaboration Homocysteine and Risk of Ischemic Heart Disease and Stroke, JAMA. 288 (2002) 2015. doi: 10.1001/jama.288.16.2015. [DOI] [PubMed] [Google Scholar]
  • [257].Humphrey LL, Fu R, Rogers K, Freeman M, Helfand M, Homocysteine Level and Coronary Heart Disease Incidence: A Systematic Review and Meta-analysis, Mayo Clin. Proc 83 (2016) 1203–1212. doi: 10.4065/83.11.1203. [DOI] [PubMed] [Google Scholar]
  • [258].Veeranna V, Zalawadiya SK, Niraj A, Pradhan J, Ference B, Burack RC, Jacob S, Afonso L, Homocysteine and reclassification of cardiovascular disease risk, J. Am. Coll. Cardiol 58 (2011) 1025–1033. doi: 10.1016/j.jacc.2011.05.028. [DOI] [PubMed] [Google Scholar]
  • [259].Yi F, Dos Santos EA, Xia M, Chen QZ, Li PL, Li N, Podocyte injury and glomerulosclerosis in hyperhomocysteinemic rats, Am. J. Nephrol 27 (2007) 262–268. doi: 10.1159/000101471. [DOI] [PubMed] [Google Scholar]
  • [260].Long Y, Nie J, Homocysteine in Renal Injury, Kidney Dis. (Basel, Switzerland). 2 (2016) 80–87. doi: 10.1159/000444900 PM - 27536696 M4 - Citavi. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Stojan G, Fu W, Petri M, Homocysteine, antiphospholipid antibodies and risk of vascular events in patients with systemic lupus erythematosus [Abstract], Ann. Rheum. Dis 76 (2017). [Google Scholar]
  • [262].Ross SC, Densen P, Complement deficiency states and infection: epidemiology, pathogenesis and consequences of neisserial and other infections in an immune deficiency., Medicine (Baltimore). 63 (1984) 243–73. http://www.ncbi.nlm.nih.gov/pubmed/6433145. [PubMed] [Google Scholar]
  • [263].Danza A, Ruiz-Irastorza G, Infection risk in systemic lupus erythematosus patients: susceptibility factors and preventive strategies., Lupus. 22 (2013) 1286–1294. doi: 10.1177/0961203313493032. [DOI] [PubMed] [Google Scholar]
  • [264].Carli L, Tani C, Spera V, Vagelli R, Vagnani S, Mazzantini M, Di Munno O, Mosca M, Risk factors for osteoporosis and fragility fractures in patients with systemic lupus erythematosus, Lupus Sci. Med 3 (2016) e000098. doi: 10.1136/lupus-2015-000098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Edens C, Robinson AB, Systemic lupus erythematosus, bone health, and osteoporosis, Curr. Opin. Endocrinol. Diabetes Obes 22 (2015) 422–431. doi: 10.1097/MED.0000000000000197. [DOI] [PubMed] [Google Scholar]
  • [266].Borba VZC, Matos PG, Da Silva Viana PR, Fernandas A, Sato EI, Lazaretti-Castro M, High prevalence of vertebral deformity in premenopausal systemic lupus erythematosus patients, Lupus. 14 (2005) 529–533. doi: 10.1191/0961203305lu2154oa. [DOI] [PubMed] [Google Scholar]
  • [267].Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, Humphrey MB, Lane NE, Magrey M, Miller M, Morrison L, Rao M, Robinson AB, Saha S, Wolver S, Bannuru RR, Vaysbrot E, Osani M, Turgunbaev M, Miller AS, McAlindon T, 2017 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis, Arthritis Rheumatol. 69 (2017) 1521–1537. doi: 10.1002/art.40137. [DOI] [PubMed] [Google Scholar]
  • [268].Anderson JJB, Kruszka B, Delaney JAC, He K, Burke GL, Alonso A, Bild DE, Budoff M, Michos ED, Calcium intake from diet and supplements and the risk of coronary artery calcification and its progression among older adults: 10-year follow-up of the multi-ethnic study of atherosclerosis (MESA), J. Am. Heart Assoc 5 (2016) 1–13. doi: 10.1161/JAHA.116.003815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Der E, Ranabothu S, Suryawanshi H, Akat KM, Clancy R, Morozov P, Kustagi M, Czuppa M, Izmirly P, Belmont HM, Wang T, Jordan N, Bornkamp N, Nwaukoni J, Martinez J, Goilav B, Buyon JP, Tuschl T, Putterman C, Single cell RNA sequencing to dissect the molecular heterogeneity in lupus nephritis, JCI Insight. 2 (2017). doi: 10.1172/jci.insight.93009. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES