Abstract
This review aimed to evaluate the mechanism of premature cardiovascular disease (CVD) in systemic lupus erythematosus (SLE) patients, particularly in the female population, and emphasize the need for early management interventions; explore the association between SLE and two autoimmune diseases, myasthenia gravis (MG) and antiphospholipid antibody syndrome (APS), and their management strategies; and evaluate the effectiveness of pharmacological and non-pharmacological interventions in managing SLE, focusing on premenopausal females, females of childbearing age, and pregnant patients. We conducted a comprehensive literature review to achieve these objectives using various databases, including PubMed, Google Scholar, and Cochrane. The collected data were analyzed and synthesized to provide an evidence-based overview of SLE, its management strategies as an independent disease, and some disease associations. The treatment should be focused on remission, preventing organ damage, and improving the overall quality of life (QOL). Extensive emphasis should also be focused on diagnosing SLE and concurrent underlying secondary diseases timely and managing them appropriately.
Keywords: antiphospholipid antibody syndrome, myasthenia gravis, therapeutic interventions, cardiac manifestations, systemic lupus erythematosus, adverse pregnancy outcomes, serum biomarkers
Introduction and background
Systemic lupus erythematosus (SLE) can be defined as a chronic multisystem autoimmune disease with a wide range of systemic manifestations involving almost all organs and tissues with its effects varying from minor cutaneous involvement to major organ damage. The burden of SLE and its survival rates vary across Asia-Pacific countries, with crude incidence rates (100,000 per year) ranging from 0.9 to 3.1, while prevalence rates range from 4.3 to 45.3 (per 100,000). A higher rate of renal involvement, which is one of the main systems involved in death, was reported for Asians as compared to whites [1]. Most of the pathology in SLE is due to immune complex (IC) deposition in various organs, which leads to the activation of complement and other mediators of inflammation. The symptoms of SLE vary widely. Fatigue in SLE is multifactorial [2]. SLE attacks the immune system, which, in addition to the immunosuppressive drugs used to treat the disease, reduces the body’s ability to fight infections [3]. General symptoms include weight loss, appetite loss, headache, myalgias, arthralgias, fever, and malaise. Weight gain may also happen due to corticosteroid treatment or nephrotic syndrome anasarca [4]. Involvement of the musculoskeletal system is common in patients with SLE [5]. Arthritis, arthralgia, osteonecrosis, and myopathy are the main manifestations [6]. Dermatological manifestations include a malar rash, which comprises an erythematous rash over the cheeks and nasal bridge.
Other features include photosensitivity, discoid rash, and alopecia. Telangiectasias, urticaria, vasculitic purpura, bullous lesions, panniculitis, livedo reticularis (LR), and Raynaud’s phenomenon are features that are related to but not specific to SLE. Deposits of immunoglobulins are found in all cases of SLE, but only 50% lead to nephritis [2]. Sepsis and renal failure are two main causes of death in patients with SLE, and the kidney is the most common organ involved. Glomerular disease usually manifests in the first few years and is usually asymptomatic. Symptoms of uremia and fluid overload may be caused by acute or chronic renal failure. Nephritic syndrome may present as hematuria and hypertension, while nephrotic syndrome may present as edema, weight gain, or hyperlipidemia. Lupus nephritis is one of the most common manifestations of SLE and one of the most damaging too. It is caused by the deposition of immune complexes and occurs in more than 50% of SLE patients [7]. Of patients with SLE, 25%-75% report neurological involvement [8]. SLE may cause status epilepticus, aseptic meningitis, optic neuropathy, myelopathy, and transverse myelitis with spastic paraparesis [7]. Stroke and transient ischemic attack may be related to antiphospholipid antibody syndrome (APS) or vasculitis. Migraines may be related to APS [9]. Headache and mood disorders might be the most common neurological manifestations of SLE [10,11]. Serositis can affect both the cardiac and pulmonary systems, and pleurisy with pleuritic chest pain with or without pleural effusion is the most common pulmonary manifestation of SLE [2]. Alveolar hemorrhage, pulmonary hypertension (PH), and thromboembolic disease are some of the other effects of SLE on the pulmonary system [2]. Oral ulcers are common in SLE, and abdominal pain may signify peritonitis, pancreatitis, mesenteric ischemia, and vasculitis. Autoimmune hepatitis leading to jaundice can also occur [7].
The most common cardiac presentation of SLE is pericarditis. Myocarditis leading to heart failure can also occur. Libman-Sacks endocarditis is also a manifestation of SLE, while coronary vasculitis presenting as angina can occur rarely. Accelerated ischemic heart disease is also commonly associated with SLE. Vasculitis, peripheral vascular disease, digital ulcers, livedo reticularis, and Raynaud’s phenomenon are common vascular manifestations of SLE [3]. Conjunctivitis, interstitial keratitis, episcleritis, and diffuse or nodular scleritis can occur with SLE, with keratoconjunctivitis sicca being the most common ocular manifestation [12,13]. SLE is associated with worse pregnancy outcomes with an increased risk of fetal death in utero, spontaneous abortions, and fetal retardations [8]. Neonatal lupus affects 3% of babies born to mothers with SLE [4]. Patients with SLE suffer from thyroid dysfunction more commonly than the general population [14]. The rate of fractures in lupus is five times higher than in the general population [15]. Glucocorticoid use can suppress pituitary function, while vitamin D deficiency is common due to avoidance of sun exposure [16]. Cytopenias such as leukopenia, lymphopenia, anemia, or thrombocytopenia can be seen in SLE, while leukopenia and lymphopenia are more common. ESR is frequently elevated in active disease, and plasma homocysteine levels are considered a risk factor for stroke in SLE [2].
Diagnosis and classification criteria
SLE diagnosis can be challenging given a wide variety of clinical features with the potential for multiple organ system involvement, as well as the overlap of clinical features with other autoimmune conditions. Initial evaluation requires a comprehensive history and physical examination along with a careful interpretation of relevant laboratory testing. It is also important to know that there are no diagnostic criteria established for the diagnosis of SLE so far. Current practices, therefore, rely on classification criteria to identify SLE cases. The 2019 European League Against Rheumatism/American College of Rheumatology (EULAR/ACR) classification criteria [17,18] for systemic lupus erythematosus has improved sensitivity (96.1%) and specificity (93.4%) compared to previous American College of Rheumatology (ACR) 1997 [19] and Systemic Lupus International Collaborating Clinics (SLICC) 2012 criteria [20]. Table 1 demonstrates the improvement in specificity from the previous ACR 1997 while maintaining the sensitivity of SLICC 2012 criteria.
Table 1. Operating characteristics of new EULAR/ACR 2019 criteria compared to ACR 1997 and SLICC 2012 classification criteria.
ACR 1997 criteria | SLICC 2012 criteria | EULAR/ACR 2019 criteria | |
Derivation | |||
Sensitivity (95% CI) | 0.85 (0.81-0.88) | 0.97 (0.95-0.98) | 0.98 (0.97-0.99) |
Specificity (95% CI) | 0.95 (0.93-0.97) | 0.90 (0.87-0.92) | 0.96 (0.95-0.98) |
Combined (95% CI) | 1.80 (1.76-1.83) | 1.87 (1.84-1.90) | 1.94 (1.92-1.96) |
Validation | |||
Sensitivity (95% CI) | 0.83 (0.80-0.85) | 0.97 (0.95-0.98) | 0.96 (0.95-0.98) |
Specificity (95% CI) | 0.93 (0.91-0.95) | 0.84 (0.80-0.87) | 0.93 (0.91-0.95) |
Combined (95% CI) | 1.76 (1.73-1.80) | 1.80 (1.77-1.84) | 1.90 (1.87-1.92) |
The recent classification designates antinuclear antibody (ANA) positivity as the qualifying factor to enter the SLE criteria given its high sensitivity. It also emphasizes the concept that all criteria must only be taken into account if there is no alternative cause suspected and the manifestation is most likely explained by SLE. Moreover, the presence of at least one clinical criterion is a must along with 10 or more points to classify as having SLE. Figure 1 discusses these points further along with a mention of clinical and immunologic criteria that are central to this classification.
While the sensitivity of the most recent EULAR/ACR classification criteria is 96%-99%, it should be kept in mind that about 1%-4% of cases of SLE can be missed if the classification is strictly followed for the purpose of diagnosis. We must also keep in mind that while the classification encompasses several clinical and immunologic manifestations of the disease, it does not incorporate various other and less common features [21]. This is particularly pertinent to diagnosing patients with ANA-negative disease [22,23]. Table 2 summarizes other features relevant to the diagnosis of a disease that is not included in the EULAR/ACR 2019 criteria.
Table 2. Organ domains and relevant features included/not included in the EULAR/ACR 2019 criteria.
Domain | EULAR/ACR 2019 classification criteria | Other features relevant for SLE diagnosis |
Autoantibodies | ||
1 | ANA (obligatory entry criterion) Anti-Sm: 6, anti-dsDNA (highly specific test): 6 | Anti-Ro/anti-La, anti-U1RNP, anti-dsDNA (tests of lesser specificity), anti-nucleosome/anti-chromatin, anti-histone, anti-C1q, anti-ribosomal P, positive Coombs test without hemolysis, false-positive serology for syphilis |
2 | Anti-cardiolipin (medium to high titer): 2, anti-β-2-glycoprotein 1: 2, lupus anticoagulant: 2 | |
Complement | ||
3 | C3 and C4 low: 4, C3 or C4 low: 3 | CH50 low, complement split products on erythrocytes |
Mucocutaneous manifestations | ||
4 | ACLE: 6, SCLE: 4, DLE: 4, oral ulcers: 2, non-scarring alopecia: 2 | Lupus tumidus, lupus panniculitis/lupus profundus, chilblains lupus, leukocytoclastic vasculitis, urticarial vasculitis, nasal ulcers |
Lupus nephritis | ||
5 | ISN/RPS class III or IV nephritis: 10, ISN/RPS class II or V nephritis: 8, proteinuria >0.5 g/day: 4 | IgA nephritis, cellular casts |
Musculoskeletal manifestations | ||
6 | Joint involvement: 6 | Myositis |
Serositis | ||
7 | Acute pericarditis: 6, pleural or pericardial effusion: 5 | Sterile peritonitis |
Neuropsychiatric manifestations | ||
8 | Seizure: 5, psychosis: 3, delirium: 2 | (Transverse) myelitis (often APS-related), chorea, mononeuritis multiplex, cranial neuropathy, peripheral neuropathy, lupus headache |
Hematological manifestations | ||
9 | Thrombocytopenia: 4, autoimmune hemolytic anemia: 4, leukopenia: 3 | Thrombotic thrombocytopenic purpura, other forms of hemolytic anemia, anemia of chronic disease, lymphopenia |
Constitutional symptoms | ||
10 | Fever:2 | Arthralgias, myalgias, fatigue, lymphadenopathy |
Other uncommon SLE organ manifestations | ||
Pneumonitis, interstitial lung disease, pulmonary artery hypertension, Libman-Sacks endocarditis (APS-related), myocarditis, hepatitis, pancreatitis. gastrointestinal vasculitis, interstitial cystitis |
Hence, making the diagnosis through an individualized approach is of utmost importance to avoid excluding potential SLE patients from receiving appropriate therapies [24].
Treatment of SLE in general
Immunomodulators
Hydroxychloroquine (HCQ): Hydroxychloroquine (HCQ) modulates the immune response pleiotropically by inhibiting B-cell receptor and Toll-like receptor (TLR) signaling, as well as intracellular TLR-3 and TLR-7 activation, which is important in nucleic acid sensing [25,26]. It raises lysosomal pH, interfering with major histocompatibility complex (MHC) antigen binding and thus processing autoantigens and cytokine secretions [27]. By interfering with the stimulator of interferon gene (STING) pathway, HCQ has an anti-type 1 interferon effect [28]. Unless there is a clear contraindication, HCQ is recommended for all patients with SLE. It is the only medication that has been shown to increase survival in lupus patients [29,30]. It has been shown to reduce lupus flares [31], prevent organ damage [32] including cardiovascular events [33] and triple mycophenolate response in lupus nephritis [34], prevent seizures [35,36], and lower the risk of developing neuropsychiatric lupus [37]. HCQ alleviates skin manifestations [38,39] and arthritis [40]. HCQ improves lipids [27], lowers insulin resistance [41] and the risk of thrombosis [42,43], and improves bone density [44].
HCQ has no immunosuppressive properties and does not increase the risk of infection or cancer [45,46]. Retinal toxicity is a rare complication that becomes more common after 20 years of treatment [47]. Retinal screening is done at the beginning, at five years, and then every year [48]. The screening test of choice is optical coherence tomography [49]. Hyperpigmentation is possible, while cardiomegaly and myopathy are two extremely rare complications. The daily dose should not exceed the threshold of 5 mg/kg real body weight based on existing evidence, which suggests that the risk of toxicity is very low for doses less than 5 mg/kg real body weight. It is worth noting that the efficacy of HCQ in lupus has been established in studies with a prescribed dose of 6.5 mg/kg/day, so it remains to be seen whether a lower dose will have comparable clinical effects. Patients in long-term remission may have their dose reduced, although no formal studies have addressed this strategy. In patients with cutaneous manifestations and HCQ-induced retinal toxicity, quinacrine, an alternative antimalarial, may be considered [50]. Figure 2 illustrates the mechanism of action of HCQ.
Corticosteroids: By non-selectively decreasing the expression of adhesion molecules and cytokines (such as interleukin (IL)-2, IL-6, tumor necrosis factor-α (TNF-α), and prostaglandins), corticosteroids have strong anti-inflammatory and immunosuppressive effects. The initial to long-term goal should be to reduce the daily dose to less than 7.5 mg/day of prednisone equivalent or to stop them because long-term glucocorticoid therapy can have several adverse effects, including irreversible organ damage [52-55]. Continuous glucocorticoid doses > 7.5 mg/day carry a markedly increased risk, and some studies even indicated a lower dose as potentially harmful [56-59]. As a result, the management strategy is to use corticosteroids as bridging therapy (oral or intramuscular (IM)) as part of an induction regimen or to treat an acute flare rather than as a maintenance treatment [60]. After ruling out infections, high-dose intravenous (IV) methylprednisolone (typically 250-1,000 mg/day for three days) is frequently used in acute, organ-threatening diseases (such as renal and neuropsychiatric) [61]. Figure 3 illustrates the genomic mechanisms of glucocorticoid-induced anti-inflammation.
Immunosuppressants
Methotrexate: Methotrexate is an antimetabolite that prevents the synthesis of purines by irreversibly binding to the enzyme dihydrofolate reductase and thus inhibits DNA synthesis, repair, and replication. Although the mechanism of its anti-inflammatory effects is not fully understood, it goes beyond arresting the cell cycle through folate depletion. For instance, co-administration of folate reduces side effects while not impairing its efficacy. Reduced circulating pro-inflammatory T cells, increased anti-inflammatory adenosine signaling, apoptosis of activated lymphocytes, reduced reactive oxygen species, and decreased adhesion molecules on endothelial and synovial cells are some pleiotropic effects of low-dose methotrexate [63]. Methotrexate (15-20 mg per week) was effective in treating cutaneous and articular disease, according to Carneiro and Sato [64], and it made it easier to lower the dosage of prednisone. A steroid-sparing effect was demonstrated by Fortin et al. [65], along with a decline in disease activity. A 60% improvement in the joint count was discovered by Rahman et al. [66]. Methotrexate is an effective and widely used treatment for cutaneous disease resistant to antimalarial and topical medications [67]. However, because it is a teratogen, methotrexate cannot be used during pregnancy.
Cyclophosphamide (CYC): A highly toxic alkylating agent, cyclophosphamide suppresses the production of antibodies by depleting T and B cells [68]. Cyclophosphamide (CYC) should only be used as a last resort in non-major organ manifestations that are refractory to other treatments and should only be considered in organ-threatening diseases (renal, cardiopulmonary, or neuropsychiatric). Due to its gonadotoxic effects, it should be used with caution in females and males of fertile age [69-71]. In premenopausal patients with SLE, concurrent use of gonadotropin hormone-releasing hormone (GnRH) analogs reduces the ovarian reserve reduction brought on by CYC therapy [72-74]. Prior to starting treatment, it is important to provide information about the potential for ovarian cryopreservation. Infections and other risks associated with CYC therapy, such as cancer, should also be taken into account [75,76].
Azathioprine: Azathioprine, a purine analog, is inactive until it is metabolized by the liver and erythrocytes into mercaptopurine, at which point it inhibits DNA synthesis and prevents immune system cell proliferation. There is a high prevalence of gastrointestinal tract toxicity, oral ulcers, nausea, vomiting, diarrhea, and epigastric pain. Leukopenia and, less frequently, thrombocytopenia and anemia are the results of dose-related toxicity to the bone marrow [77]. Compared to corticosteroids alone, azathioprine has been shown in two small randomized studies to decrease mortality, the frequency of flare-ups, and corticosteroid use, including in patients with severe renal or central nervous system (CNS) disease [78,79]. Azathioprine is frequently used as a corticosteroid-sparing agent in extra-renal lupus [80]. As the metabolite 6-methyl mercaptopurine (6-MMP) is not produced in the developing fetus, azathioprine is still a great option for controlling renal and extra-renal disease during pregnancy [81]. Figure 4 illustrates the immunomodulatory effects of azathioprine.
Mycophenolate mofetil (MMF): The rate-limiting enzyme in the synthesis of guanosine nucleotides, inosine monophosphate dehydrogenase, is inhibited by mycophenolate mofetil, a prodrug of mycophenolic acid, an effective immunosuppressant for both renal and non-renal lupus (although not in neuropsychiatric disease) [83-85]. Enteric-coated mycophenolate sodium (EC-MPS) outperformed azathioprine in a recent randomized, open-label trial for extra-renal SLE in terms of achieving remission and lowering flares [86]. However, because of its teratogenic potential, it needs to be discontinued at least six weeks prior to conception, and a higher price compared to azathioprine or methotrexate limits its ability to be recommended universally in females of reproductive age with non-renal manifestations. Figure 5 illustrates the inhibition of nucleotide synthesis by mycophenolate mofetil.
Calcineurin inhibitors: Calcineurin inhibitors block T cells through the inhibition of calcineurin [76]. This inhibits T cells and lowers levels of IL-1b, interferon-gamma (IFN-γ), IL-6, and IL-10 by preventing transcription factors such as nuclear factor of activated T cells (NFAT) from translocating. B-cell activation is also compromised, in addition to class switching and immunoglobulin production. Furthermore, calcineurin inhibitors stabilize podocytes and decrease mesangial proliferation in the kidneys, improving proteinuria [87-89]. Tacrolimus is preferred for SLE. Tacrolimus has been shown to be additive to mycophenolate mofetil for the treatment of lupus nephritis in particular [90] and has been studied as a monotherapy and as a component of a multi-targeted approach [90-92]. Other studies have shown that tacrolimus is a suitable substitute for mycophenolate mofetil [93]. A meta-analysis found that tacrolimus was superior to CYC for lupus nephritis induction therapy [94]. Tacrolimus is a treatment option for refractory cutaneous disease during pregnancy [95] and can be applied topically. Figure 6 illustrates the inhibition of T cells by tacrolimus.
Biological Agents
Belimumab: In SLE, irregular B-cell pathways are usually present. B cells play a role in autoantibody production, T-cell antigen presentation, and cytokine release (including interferon-α, IL-6, IL-10, B-cell activating factor (BAFF), TNF-α, and a proliferation-inducing ligand (APRIL)) [96]. Belimumab, a fully humanized monoclonal antibody, binds to soluble BAFF, resulting in a decrease in the number of peripheral naive and transitional activated B cells [97,98]. Belimumab should be considered in extra-renal disease with insufficient control (ongoing disease activity or frequent flares) to first-line treatments (typically a combination of HCQ and prednisone with or without immunosuppressive agents) and an inability to taper glucocorticoid daily dose to acceptable levels (i.e., maximum of 7.5 mg/day). Belimumab may benefit patients with persistent disease; patients with high disease activity (e.g., Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) > 10), prednisone dose > 7.5 mg/day, and serological activity (low C3/C4 and high anti-dsDNA titers), with cutaneous, musculoskeletal, and serological manifestations, are more likely to respond [99-101].
Rituximab (RTX): Rituximab (RTX) is a chimeric monoclonal antibody that kills mature B cells and B-cell precursors by attacking CD20 on B cells. Due to the negative findings of randomized controlled trials (RCTs), RTX is currently only used off-label in patients with severe renal or extra-renal (primarily hematological and neuropsychiatric) disease that is resistant to other immunosuppressive agents and/or belimumab or in patients who have contraindications to these medications. More than one immunosuppressive drug must have failed before RTX is administered [102-105], with the possible exception of severe autoimmune thrombocytopenia (ITP) and hemolytic anemia, for which RTX has shown efficacy in both lupus patients and those with isolated immune thrombocytopenia (ITP) [106-108]. In lupus nephritis, RTX is typically considered when first-line therapies (CYC and MMF) have failed or when the condition is relapsing [102,109]. Other therapies such as anifrolumab, baricitinib, and atacicept are under study. Figure 7 illustrates targeted biological agents available in present or previous clinical trials of systemic lupus erythematosus.
Figure 8 illustrates the overview of the management of SLE based on the severity of the disease.
Review
Managing SLE in the premenopausal period, females of childbearing age, and pregnancy
Recent SLE epidemiological studies report the global prevalence of SLE to be 15.87-108.92 per 100,000 persons, with 1.4-15.13 new cases per 100,000 persons per year [112]. Studies show that the disease prevalence and incidence are several folds in females compared to males [112,113] with a prevalence ratio ranging between 7:1 and 15:1 between females of childbearing age and males [114,115]. Given that SLE affects females of reproductive age, understanding issues regarding reproductive health including fertility, family planning, contraception, pregnancy implications, drug safety during pregnancy, and breastfeeding is of utmost importance.
Reproductive Ability
Several risk factors play a role in decreasing fertility in females with SLE [116]. Chronic disease activity and recurrent flares diminish ovarian reserves by creating a state of inflammation within the ovaries and disturbing the hypothalamic pituitary ovarian axis [117-119]. A cross-sectional study also demonstrated an association between premature ovarian failure and specific SLE-related autoantibodies as well as different immunosuppressants used to treat the disease [120].
Management of SLE in Pregnant Mothers
For a long time, doctors advised females with SLE to put off having children out of fear of a disease flare or a bad outcome for the fetus. Having patients under good control on drugs suitable for pregnancy before conception is a crucial part of managing SLE during pregnancy. Patients should preferably be cared for by a multidisciplinary team with expertise in rheumatic illnesses and maternal-fetal medicine. There is a lack of information on the safety of medications for females who are pregnant or nursing. Providers have long adhered to the A, B, C, D, and X pregnancy grades established by the US Food and Drug Administration (FDA) [121]. Table 3 lists the medications that can be safely used or discontinued during pregnancy.
Table 3. Medications to continue or discontinue during pregnancy.
Medications to continue during pregnancy | Medications to discontinue prior to pregnancy |
Prenatal multivitamin | Cyclophosphamide |
Low-dose aspirin | Mycophenolate mofetil |
Hydroxychloroquine | Methotrexate |
Prednisone (moderate dose) | Leflunomide |
Azathioprine | |
Aspirin (81 mg) |
Contraception and Family Planning
Studies report that many females with SLE do not use effective contraception adequately [123,124]. Extensive contraception counseling should be provided to avoid the risks of unwanted pregnancies during periods of moderate to severe disease activity and teratogen intake. Counseling should include individualized discussions about effective available contraceptive options and should take disease-related factors into account, especially disease activity and the risk of thrombosis (presence of antiphospholipid antibodies (aPL) in particular). Table 4 illustrates the association of these factors with various contraceptive modalities.
Table 4. Summary of recommendations for contraceptives in SLE patients.
Clinical presentation | Copper IUD | LNG IUD | Progesterone-only pill | DMPA | COC | Vaginal ring | Patch |
SLE, low disease activity, (-) aPL | Effective, long-acting | No increase flare | Risk of osteoporosis with prolonged use | No increase flare | Similar estrogen level to COC, no data | Higher estrogen levels than COC, avoid | |
SLE, active disease, (-) aPL | Effective, long-acting | No increase flare | Risk of osteoporosis with prolonged use | No studies, avoid | |||
SLE stable on immunosuppressive medication, (-) aPL | Effective, long-acting, no infection data but likely low-risk | No increase flare | Risk of osteoporosis with prolonged use | Check for medication interactions | |||
SLE with renal impairment, (-) aPL | Effective, long-acting | No increase flare | Risk of osteoporosis with prolonged use | Avoid drospirenone-containing COC due to risk of hyperkalemia | |||
SLE with (+) aPL | Effective, long- acting | Low/no increase thrombosis | Low/no increase thrombosis | Low/uncertain risk thrombosis | Increased risk of thrombosis, avoid | ||
SLE with thrombosis/on anticoagulation | Increase menstrual bleeding | Low/no increase thrombosis; decreases menstrual bleeding /amenorrhea | Low/no increase thrombosis | Low/uncertain risk thrombosis | Increased risk of thrombosis, avoid |
Copper and levonorgestrel intrauterine devices (IUDs) are safe options compatible with all levels of disease activity [126] and have the advantage of providing long-term contraception. Both IUDs can also be safely used in patients with concomitant aPL. Levonorgestrel IUD has the additional benefit of controlling menorrhagia in patients who are on anticoagulation medications [127]. Previous concerns regarding the increased risk of pelvic infections with a copper IUD have not been entirely supported in many studies [128]. Moreover, immunosuppressive therapy is not a contraindication for using IUDs [129]. Other options, for the purpose of long-term contraception, include subdermal progestin-based implants. Progestin-only contraception does not increase the risk of thromboembolism as seen in a large meta-analysis study [130].
Among hormonal contraceptives, combined oral contraceptive (COC) pills, and progesterone-only pills, safety has been established for use in inactive or stable active disease with negative antiphospholipid antibodies (aPL) [126-131]. However, the use of combined hormonal contraceptive pills should be discouraged in patients with positive antiphospholipid antibodies (aPL) [126]. A case-control study found that in patients with positive aPL, COCs increased the risk of arterial thrombosis [132]. Lastly, other combined hormonal contraceptives such as vaginal rings and transdermal patches have estrogen levels either similar or more to COCs, respectively, and hence should be avoided for use in SLE [133]. The use of depot medroxyprogesterone acetate (DMPA) injections should be avoided for long-term contraception in patients being treated with corticosteroids due to the potential risk of osteoporosis [134].
Pregnancy
The management of SLE in pregnant females is guided by the risk of disease-related complications for pregnant females and the fetus, as well as the benefits versus risks associated with different therapies available for this population.
Adverse Pregnancy Outcomes and Predictors
SLE is associated with an increased risk of obstetric complications and unfavorable pregnancy outcomes. Preeclampsia is the most frequently occurring complication among several others, including increased length of hospital stay, hypertension, intrauterine growth retardation (IUGR), preterm birth, and stillbirth [135-140]. Increased disease activity, in the preconception period, is strongly associated with adverse pregnancy outcomes including premature birth and fetal loss [141], and pregnancy should be delayed for at least six months after remission is achieved [135,141]. In addition to a high SLEDAI score, having lupus nephritis within six months before pregnancy is associated with higher rates of maternal complications including disease flares during pregnancy [142,143]. Other factors associated with adverse outcomes are major organ involvement, the presence of anti-Ro/La antibodies [144,145], and the presence of hypercoagulability [146]. Discontinuation of hydroxychloroquine is associated with the risk of disease flares during pregnancy and the postpartum period [147,148] as well as preeclampsia [149].
Management Therapies During Pregnancy and Lactation
Pregnancies were previously discouraged given the high risk of lupus flares and adverse obstetric outcomes, but advancements in medical knowledge and the development of compatible medication have made successful pregnancies possible. The teratogenicity of medication must be considered while treating pregnant patients. Table 5 summarizes the recommendations regarding the use of various medications during pregnancy and lactation period.
Table 5. Safety of common SLE treatments during pregnancy and lactation.
Medication | Preconception | During pregnancy | Lactation |
Compatible | |||
Hydroxychloroquine | + | + | + |
Sulfasalazine | + | + | + |
Azathioprine | + | + | + |
Cyclosporine | + | + | + |
Tacrolimus | + | + | + |
Prednisone | Keep dose <20 mg/day | Keep dose <20 mg/day | Keep dose <20 mg/day |
NSAIDs | Discontinue with difficulty conceiving | Stop at week 20 | + |
Stop at conception | |||
Belimumab | Stop with positive pregnancy test | - | + |
Rituximab | Stop with positive pregnancy test | - | + |
Abatacept | Stop with positive pregnancy test | - | + |
Stopping hydroxychloroquine (HCQ) precipitates flares of lupus, while continuation during the pregnancy decreases the need for average glucocorticoid dosing [147,148,151]. HCQ is known to be safe for the fetus. A case series study documented no association between HCQ and congenital abnormalities [152]. Another double-blinded placebo-controlled study documented higher delivery age and Apgar score with zero incidences of ophthalmological and auditory abnormalities in early childhood [151]. Moreover, it can be safely continued during breastfeeding [153]. According to the US Preventive Services Task Force (USPSTF) recommendation statement, low-dose aspirin should be initiated from approximately 12 weeks of gestation in females at high risk of developing preeclampsia, which includes patients with SLE [154]. The lowest possible dose of prednisone should be used to control the disease as glucocorticoids are well known to cause gestational complications such as hypertension and diabetes. While earlier studies have mentioned an increased risk of cleft palate, a large study of 51,973 infants exposed to glucocorticoids in utero did not demonstrate this association [155]. Prednisone transfer to breast milk is minimal and is considered safe for the nursing infant when taken in low doses (<20 mg/day) [156].
Nonsteroidal anti-inflammatory drugs (NSAIDs) are widely used to help control pain in patients with musculoskeletal involvement. The US Food and Drug Administration (FDA) recommends against using NSAIDs around 20 weeks of gestation due to the risk of fetal renal dysfunction leading to oligohydramnios [157,158]. NSAIDs are widely used as analgesics after parturition to treat pain in postpartum females and are generally considered safe [159]. Azathioprine use in central nervous system (CNS) disease and lupus nephritis is associated with improved survival and fewer hospitalizations [78]. A study of 178 pregnancies showed that the rate of poor pregnancy outcomes did not differ between the azathioprine-exposed and non-exposed groups, and no congenital abnormalities were noted in the infants [160]. Calcineurin inhibitors such as tacrolimus and cyclosporine are well known for achieving and maintaining remission in moderate to severe lupus, including lupus nephritis, and can be used as options where other modalities are either not tolerated or contraindicated, such as during gestation. A study of 54 pregnancies showed no difference in adverse maternal and fetal outcomes between tacrolimus-exposed and non-exposed pregnancies [161]. Subsequently, an observational study of 60 pregnancies did not demonstrate a difference in rates of fetal mortality, preterm delivery, hypertensive disorders of gestation, and small for gestational age infants in either of the groups [162]. Limited observation of cyclosporine also suggests an acceptable benefit/risk ratio for use in pregnancy [163].
Biological medications such as rituximab and belimumab are immunoglobulin G (IgG)-based medications that do not significantly cross the placenta until after 12 weeks of gestation. Hence, they can be cautiously continued through conception and should be stopped after the first missed period where possible. Alternatively, several studies [164-166] have provided enough evidence to support the continued use of biologics throughout pregnancies where alternate therapies fail to achieve disease control. Cyclosporine, tacrolimus, azathioprine, and anti-TNF-α inhibitors are also considered safe to be used during the lactation period [156].
Antineoplastic therapies and leflunomide are used for moderate to severe disease/major organ involvement but are strictly contraindicated in pregnancy [72,167-170]. The teratogenicity of CYC has been extensively studied in the past and is well known to cause fetal malformations and CNS and musculoskeletal defects [171] if used in the first trimester of pregnancy. Mycophenolate mofetil is one of the most commonly used drugs for lupus nephritis. It is not only associated with pregnancy loss [172] but is also associated with multiple fetal malformations including facial, esophageal, and ear defects [173-176]. Methotrexate is also a well-known abortifacient, hence resulting in pregnancy loss simultaneously predisposing the exposed fetus to intrauterine growth restriction and CNS, skull, and other musculoskeletal defects [177]. Leflunomide, while not yet established to be a human teratogen, is known to be embryotoxic and teratogenic in non-human models. Hence, it is classified as pregnancy category X by the FDA [178]. Leflunomide has a half-life of two weeks, and it can take a long time for levels to become undetectable in the blood after the therapy is discontinued [179]. Patients who wish to become pregnant should undergo a washout procedure with cholestyramine to aid in the elimination of the drug before conception [179,180]. Literature on CYC, mycophenolate, methotrexate, and leflunomide pertaining to breastfeeding is not sufficient to support or contradict their use during lactation. While most of the other therapies appear to be safe, these medications should not be continued during lactation [167].
Cardiovascular disease (CVD) risk in premenopausal patients with SLE
Cardiovascular disease (CVD) is one of the main causes of mortality globally [181]. Obesity, smoking, hypertension, physical inactivity, dyslipidemia, and diabetes were considered to be risk factors for CVD in females. However, in recent years, other risk factors have risen dramatically, which include preterm delivery, hypertensive disorders in pregnancy, breast cancer treatment, depression, and autoimmune diseases such as rheumatoid arthritis (RA) and SLE [182]. SLE affects every aspect of the immune system, and humoral autoimmunity with the development of specific autoantibodies and serum cytokine dysregulation are the disease hallmarks. Female bias in autoimmunity might be attributed to a variety of factors, which include the obvious sex-related variations in the immune systems of males and females that lead to females’ increased immunologic reactivity and may predispose them to acquire an autoimmune illness, possibly because estrogen inhibits Th1- dependent disorders but potentiates Th2-dependent disorders [183]. The prevalence of premature CVD in premenopausal females with SLE is estimated to be up to 50 times higher than in the general population with a male/female ratio of 9:1 [184].
The mechanism of CVD in premenopausal females with SLE is multifactorial. They are induced because of accelerated atherosclerosis, and autoimmune disorders are related to an increase in cardiovascular illnesses. These are presently regarded as nontraditional risk factors, with an increase in coronary and cerebral-vascular mortality [185]. Chronic inflammation associated with SLE leads to endothelial dysfunction, which is a key factor in the pathogenesis of CVD. This inflammation also increases the risk of atherosclerosis and promotes the development of thrombosis. It leads to the aging of vessels and raises the cumulative cardiovascular risk, just like metabolic syndrome, obesity, diabetes, chronic renal insufficiency, and sleep apnea syndrome (SAS) [185]. The recruitment of monocytes to the artery wall is an important step in the development of atherosclerosis and endothelial production of vascular cell adhesion molecule-1 (VCAM-1), which contributes to this process [186].
SLE is an inflammatory systemic illness with a typical sequela of early atherosclerosis [187]. Circulating immune complexes (IC) are also widely seen in the serum of SLE patients and are responsible for many of the disease’s acute inflammatory symptoms. Our findings that immune complex IC-C1q reduces cholesterol 27-hydroxylase mRNA and protein levels in endothelial cells and macrophages add to the theory that IC-C1q contributes to the development of atherosclerosis and may alter crucial metabolic processes in the vessel wall. Circulating IC, therefore, promotes atherosclerosis through two mechanisms, either by encouraging macrophage recruitment to the arterial wall and/or by decreasing endothelial and macrophage capacity to undertake reverse cholesterol transport by lowering intracellular cholesterol 27-hydroxylase levels. The immune response to self-antigens causes tissue damage or dysfunction in autoimmune diseases, which can occur systemically or affect specific organs or body systems. Numerous studies have linked IFN-α to SLE. During self-material uptake, IFN-α activates antigen-presenting cells, breaking immunologic self-tolerance [188]. Many SLE patients have increased serum IFN-α, which correlates with disease activity [189,190].
Recombinant human IFN-α used to treat chronic viral hepatitis and cancer may produce de novo SLE [191]. IFN-α-induced SLE usually disappears after discontinuation [192,193]. Of healthy first-degree relatives of SLE patients, 20% have excessively high serum IFN-α compared to 5% of healthy unrelated persons [194]. These findings imply that elevated serum IFN-α is heritable for SLE [194]. Patients and healthy first-degree relatives have the highest serum IFN-α activity during peak SLE incidence [195]. Polygenic inheritance may explain SLE families’ elevated IFN-α characteristics. These studies strengthen the idea that IFN-α pathway dysregulation causes human SLE. Also, there was a trend toward an inverse connection between age and serum IFN-α activity in both male and female patient groups, as well as male and female healthy relatives [195].
Managing CVD manifestations of SLE in premenopausal females
There are several therapeutic interventions that can be used to mitigate CVD manifestations in premenopausal females with SLE.
Lifestyle Modifications
Lifestyle modifications such as regular exercise, healthy diet, and smoking cessation can reduce the risk of CVD in this population [196], considering that obesity is more common among patients with SLE than in the general population [197], with prevalence ranging from 28% to 50% [198]. Established risk factors such as hypertension, dyslipidemia, and diabetes must be managed appropriately [199].
NSAIDs
Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used to manage SLE-related symptoms and should be used cautiously in patients with a high risk of CVD. NSAIDs are used to treat fever, arthritis, serositis, and headaches in over 80% of individuals with SLE. NSAID-induced hepatotoxicity, acute renal failure, cutaneous and allergy responses, and aseptic meningitis is enhanced in SLE patients. However, NSAIDs should be safely recommended to most lupus patients if their administration is reevaluated on a frequent basis [200]. Long-term NSAID use has no elevated CVD risks, according to studies, while some studies also suggest an increased CVD risk. More research into other autoimmune/auto-inflammatory disorders is needed to see if they have similar effects [201].
Reduction of Lipids
As cellular processes require both energy and associated signaling, lipogenesis is a prime therapeutic focus. Furthermore, immunocytes rely on lipids to exert their specialized roles in response to stimuli; hence, it is important to strike a balance between the effects of lipid metabolism on immunocytes and the internal environment in order to investigate additional targets. Furthermore, several factors have been implicated in the complicated pathways of lipid metabolism that operate directly or indirectly on SLE. The importance of studying the consequences of interfering with lipid metabolism in appropriate in vivo models, in particular in combination with standard immunosuppressive medications, is emphasized by the role that lipids play in the immune system, particularly among various cell populations [202]. Thus, statins, which are lipid-lowering agents, may be used to reduce the risk of atherosclerosis in these patients, but the proven benefit still requires more data to establish the same [203].
Corticosteroids
Long-term use of corticosteroids for their anti-inflammatory and immunosuppressive effects has been a mainstay of SLE treatment for decades. Just around 10% of prednisolone and prednisone are able to penetrate the placenta because they are inactivated by 11-hydroxysteroid dehydrogenase [204]. Inflammatory rashes on the skin can be treated with topical steroids, and intralesional preparations are an option for discoid lupus that has progressed to a severe stage. Intra-articular steroid injections, with or without a local anesthetic agent, may be a more direct and preferred therapy of choice for localized soft tissue and joint involvement. Due to their pro-atherogenic qualities, corticosteroids have been linked to deleterious effects on metabolic parameters such as body fat distribution, blood pressure, and glucose metabolism [205-207]. According to research [208,209], they increase low-density lipoprotein (LDL) cholesterol and triglyceride levels while decreasing high-density lipoprotein (HDL) cholesterol. There is growing evidence that inflammation itself predisposes to atherosclerosis, and steroids can reduce this risk [206], so it is important to evaluate the benefit/risk ratio of reducing chronic inflammation versus side effects and cardiovascular risk on an individual basis. Long-term usage of corticosteroids for the treatment of SLE [210] has been linked to weight increase (from under 10 to over 30 pounds). Therefore, the increased risk of cardiovascular disease associated with the excess body weight seen in SLE patients on prolonged use of corticosteroids creates a vicious loop in which weight gain might maintain disease activity and necessitate the maintenance of these medications.
Aspirin
Aspirin therapy may also be considered in selected patients with a high risk of thrombosis. To more precisely define its function in these patients, controlled, prospective investigations are required [211].
Hydroxychloroquine
Hydroxychloroquine (HCQ), a regularly used SLE treatment, may have cardioprotective benefits and is linked to a lower incidence of CVD in these patients. HCQ may lower the incidence of flares, allowing for a decrease in steroid dosage, lessen organ damage, and prevent the thrombotic effects of antiphospholipid antibodies. The medication is generally safe and can be given to pregnant women [212].
Myasthenia gravis (MG) and SLE
SLE generally has a relapsing and remitting course, and in 90% of cases, it is seen in females of childbearing age. Its severity varies from mild to rapidly progressing, along with symptoms such as facial rash, photosensitivity, arthritis, fever, serositis, cytopenia, and oral ulcers [213]. On the other hand, myasthenia gravis (MG) is a rare organ-specific autoimmune disease of the neuromuscular junction characterized by autoantibodies against the nicotinic acetylcholine receptor (nACHR), the lipoprotein receptor-related protein 4 (LRP4), or the muscle-specific tyrosine kinase (MuSK) receptors [214]. This leads to the blockage of the neuromuscular junction resulting in muscular weakness, which improves with rest and worsens with muscle use. It generally occurs in females of age <40 years with ocular symptoms (diplopia and ptosis); slurred speech; difficulty in swallowing, chewing, walking, and lifting objects; facial weakness; and dyspnea [215]. Rarely, SLE and MG have similar features that precede one another or can coexist in a patient [214].
However, it is quite interesting to note that in patients undergoing thymectomy for MG, the association between SLE and MG has been reported. For example, a case report of a 48-year-old female stated the occurrence of SLE and secondary antiphospholipid syndrome (APS) 28 years post-thymectomy for MG, with thymectomy being the sparkling factor [216]. Thymectomy is the first-line treatment for generalized or serious myasthenia because thymic abnormalities are frequently seen in MG patients and the thymus is known to produce autoantibodies [215,217]. Thymectomy, however, has no impact in cases of SLE that have already been diagnosed [217]. A study was conducted among 13 patients with MG and preexisting SLE, out of which 11 had sufficient data available. Table 6 summarizes the diagnoses of these patients based on the standards established by the ACR [218].
Table 6. Common clinical characterizations in SLE and MG overlap as noted in 11 out of 13 patients.
Total number | 11 |
ACR criteria for SLE diagnosis | Number (%) |
Malar rash | 2 (18.2%) |
Oral ulcers | 2 (18.2%) |
Photosensitivity | 2 (18.2%) |
Renal disorder | 2 (18.2%) |
Discoid rash | 3 (27.3%) |
Neurological disorder | 4 (36.4%) |
Serositis | 6 (54.5%) |
Hematological disorder | 8 (72.7%) |
Arthritis | 10 (90.9%) |
Immunologic disorder* | 10 (90.9%) |
Antinuclear antibody | 11 (100%) |
Immunologic Link in the Pathogenesis
The pathogenesis of both conditions has been linked to an alpha-chemokine subfamily (CXC) in particular [219,220]. According to studies, these chemokines are in charge of several immunoreactive cell mobility via chemoattraction. Additionally, they are involved in angiogenesis and may facilitate the activation of dendritic cells, monocytes, T cells, B cells, NK cells, basophils, and eosinophils [221]. Studies on animal models have demonstrated that CXCL13 interacts with B and T lymphocytes, causing the precipitation of SLE in patients with established MG [219]. Granulocyte-macrophage colony-stimulating factor (GM-CSF), which can be found exogenously and endogenously, is also a contributing component in the development of both diseases. It is worth noting that, in addition to vascular endothelial cells, fibroblasts, mast cells, monocytes, and macrophages, T and B cells are all involved in the endogenous synthesis of GM-CSF, indicating its significant immunologic association [222].
Thymectomy and the Increasing Risk for Autoimmune Diseases
The thymus is where T cells mature. Pathological processes in the thymus induce cell dysfunction and activate autoreactive CD4+ T lymphocytes, which interact with B lymphocytes to produce autoantibodies [223]. By suppressing the activity of CD4+ T lymphocytes, regulatory T lymphocytes are in charge of stopping the autoimmune process. It has been suggested that the absence or dysfunction of regulatory CD4+ CD25+ T lymphocytes is a cause of connective tissue disorders and consequently of MG and SLE [223]. The occurrence of systemic autoimmune disorders in patients who underwent thymectomy for MG has been documented in some situations [217]. Loss of central tolerance to its antigen and increased autoantibody production are side effects of thymectomy [224]. Polyarthritis and polyarthralgia were the most prevalent symptoms in post-thymectomy SLE cases, and laboratory results revealed mild T-cell lymphopenia, hypergammaglobulinemia, and B-cell hyperreactivity [225]. Many autoimmune illnesses, including SLE, Hashimoto’s disease, APS, idiopathic portal hypertension, and cutaneous vessel vasculitis, can be made worse by thymectomy [225]. Table 7 displays the prevalence and concomitance of SLE and MG as reported in various articles [224,226-228].
Table 7. Prevalence and concomitance of SLE and MG.
SN | MG (n) | SLE (concomitant) | Gender (M:F) | Age at diagnosis (Av) | Thymectomy associated with SLE | Prevalence | Reference |
1 | 127 | 1 | 0:01 | 48.8 | - | 0.80% | [14] |
2 | 132 | 5 | 1:04 | 24-58 | 2/5 | 3.78% | [15] |
3 | 78 | 6 | 0:06 | 44.5 | 2/6 | 7.70% | [16] |
SN | SLE (n) | MG (concomitant) | Gender (M:F) | Age at diagnosis (Av) | - | Prevalence | - |
1 | 1300 | 17 | 0:17 | 34.5 | - | 1.30% | [12] |
Although it was believed that HCQ can lead to the development of MG, patients who received this medication for SLE displayed weaker MG symptoms than other patients [224,226]. Antimalarial medications directly impact the neuromuscular junction, frequently leading to neuromyopathy and atrophic muscle fibers in muscle biopsies [224]. When symptoms persist even after discontinuing HCQ, MG should be ruled out because the drug can cause ocular symptoms and symmetrical muscle weakness [214].
Simultaneous Management of Both Diseases
Four patients with SLE-MG overlap syndrome were reported in a case series; two underwent thymectomy after the diagnosis of MG was established and received pyridostigmine, while the third case initially responded poorly to pyridostigmine and was later identified as having SLE-myositis overlap syndrome [217]. This series shows various treatment plans for different cases. For the case series reported by Minchenberg et al. [217], the events and the treatment are shown in Table 8.
Table 8. A case series of four patients who underwent thymectomy and other treatment strategies for SLE and MG overlap management.
Variables/findings | Case 1 | Case 2 | Case 3 | Case 4 |
Age/gender | 56/F | 57/M | 58/F | 62/F |
Age at MG onset | 10 | 54 | 58 (initially diagnosed as SLE) | 33 |
SLE | Present | Present | Present | Present |
Thymectomy | Yes | No | No | Yes |
Treatment (initial) | Thymectomy, pyridostigmine | Cholinesterase inhibitor | HCQ (discontinued by personal decision) | Thymectomy, pyridostigmine |
Treatment (later) | HCQ | Mycophenolate mofetil, HCQ | None | HCQ |
Telitacicept (Tai'ai®) is a fusion protein made up of a recombinant transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor fused to the fragment crystallizable (Fc) region of human immunoglobulin G (IgG). For the therapy of B-cell-mediated autoimmune conditions such as SLE, rheumatoid arthritis (RA), and multiple sclerosis (MS), Yantai Rongchang Pharmaceutical is developing telitacicept through its subsidiary RemeGen. Telitacicept inhibits the growth and survival of mature B cells and plasma cells by binding to and inhibiting the action of two cell-signaling molecules, B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL). The use of telitacicept for the treatment of patients with active SLE was given its initial permission in China in March 2021. China is currently conducting clinical trials of telitacicept for a number of additional indications, such as IgA nephropathy, MS, MG, neuromyelitis optica, spectrum diseases, RA, and Sjögren’s syndrome. The development of telitacicept, which resulted in this first approval for SLE, has been summarized in this paper [229]. Figure 9 shows the mechanism of action of telitacicept.
Managing antiphospholipid antibody syndrome (APS) with SLE
Antiphospholipid syndrome (APS) can be defined as the association of thrombosis and/or pregnancy morbidity with antiphospholipid antibodies (aPL), lupus anticoagulant (LA), anticardiolipin antibodies (aCL), and/or anti-β2-glycoprotein-I antibodies (aβ2GPI) [231]. APS can either be primary, which occurs in otherwise healthy individuals without an underlying disease, or it can occur simultaneously with autoimmune diseases, particularly SLE. Patients with SLE and APS can develop valvular heart disease, pulmonary hypertension (PH), livedo reticularis (LR)/racemosa, thrombocytopenia, hemolytic anemia, renal thrombotic microangiopathy (TMA), and cognitive dysfunction [232]. SLE and aPL-positive patients can both progress to end-organ damage with one-third of SLE patients progressing to end-organ damage within five years of diagnosis and one-third of aPL patients progressing to end-organ damage within 10 years of disease [233]. Around 30%-40% of patients with SLE test positive for aPL [234]. The risk of vascular events and death in SLE increases due to aPL [235,236]. Nearly 40% of aPL-positive patients among all SLE patients develop arterial and/or deep venous thrombosis (DVT) as compared to 10%-20% of aPL-negative patients [237]. Wahl et al. [238] conducted a meta-analysis that showed that patients with SLE and LA are at six times greater risk of developing venous thromboembolism and at 11 times greater risk of developing recurrent venous thromboembolism as compared to those without LA. Patients having SLE with aPL are twice at risk of developing DVT/pulmonary embolism (PE) and four times at risk of developing recurrent DVT after the first event as compared to those without aCL.
Pregnancy morbidity in patients with SLE and aPL is 25%-47%, while that in SLE without aPL is 0%-38% [239,240]. In patients with lupus nephritis, aPL increases the risk of maternal hypertension and premature births. aPL is also associated with an increased rate of induced abortion [241]. The presence of valvular lesions is 40%-50% in aPL-positive lupus, while it is around 20% in aPL-negative SLE [242]. The frequency of pulmonary hypertension in patients with SLE and aPL is 15%-100%, and in SLE without aPL, it is 11%-55% [243]. A meta-analysis conducted by DeFilippis et al. [244] showed that the percentage of livedo reticularis in aPL-positive SLE was 27%, and in aPL-negative SLE, it was 11%. SLE patients with LA or aCL are three times more likely to have thrombocytopenia as compared to those without LA or aCL, as shown by a meta-analysis conducted by Love et al. [237]. Autoimmune hemolytic anemia is found in 20%-28% of aPL-positive SLE, while its percentage is 1%-9% in aPL-negative SLE patients [245]. aPL nephropathy is present in 25%-39% of aPL-positive SLE patients, and its prevalence in aPL-negative SLE is 4%-16% [245]. A study conducted by Coín et al. [246] showed the prevalence of cognitive impairment in aPL-positive SLE patients to be 21% as compared to 11.9% in aPL-negative SLE patients.
In the majority of the cases, there is no difference in the management of aPL-positive patients with or without lupus. There are however certain exceptions. Multiple studies on aPL-positive patients with or without systemic autoimmune diseases suggested that low-dose aspirin may play a role in protecting against the first episode of thrombosis in aPL-positive SLE patients [247-249]. The use of low-dose aspirin in the first pregnancy in a patient with aPL-positive SLE is justified as both pregnancy and SLE are risk factors for thrombosis and low-dose aspirin may decrease the risk of preeclampsia in high-risk patients [245]. A few case reports have pointed out that adding warfarin, heparin, or aspirin to the standard treatment in patients with aPL nephropathy is beneficial [250-253]. The European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) suggested the use of hydroxychloroquine and/or antiplatelet/anticoagulant for lupus patients with aPL nephropathy [254]. A recent study suggested that lupus patients with APS had higher activation of the mTOR pathway as compared to those lupus patients without APS. For patients with aPL nephropathy who required kidney transplantation, those who were treated with rapamycin (10 patients) had decreased vascular proliferation, and no recurrence of vascular lesions proliferation was observed. At 144 months post-transplantation, seven of 10 (70%) aPL nephropathy patients treated with rapamycin had a functioning allograft in comparison with only three of 27 (11%) patients who were not treated with rapamycin [255]. The use of low-dose aspirin remains controversial for primary thrombosis and pregnancy morbidity prevention and so is the use of anticoagulation in lupus nephritis patients with aPL nephropathy [245].
Non-pharmacological interventions for the improvement of fatigue, depression, pain, and quality of life (QOL) in SLE patients
Fatigue, pain, and depression significantly influence the overall quality of life of SLE patients, in conjunction with disease activity. Therefore, it is imperative to implement effective interventions aimed at improving the quality of life (QOL) for these individuals. In a literature review published in 2016, a total of 12 randomized studies with 846 participants, comprising seven randomized trials, one non-randomized trial, and four prospective observational studies, were analyzed. The review concluded that physical exercise, behavioral and psychological interventions, acupuncture, dietary modifications, and phototherapy demonstrated efficacy in mitigating fatigue among SLE patients. Notably, aerobic exercise emerged as the most effective intervention, although the study findings were not always uniform [256]. A strategy was suggested to assess and handle fatigue among SLE patients. The proposed approach entails examining potential non-lupus factors that could contribute to fatigue, monitoring disease activity, evaluating for anxiety and depression, and administering suitable interventions such as managing non-lupus etiologies, treating active disease, and utilizing medication and psychological interventions to manage anxiety and depression [257]. Figure 10 shows the management of fatigue in SLE.
Another approach to manage fatigue, exhaustion, and pain is balneotherapy. It is a therapeutic approach that utilizes the healing properties of mineral waters, mud, and natural gases from springs that are widely acknowledged and accepted for their medicinal and legal benefits. The main objective of balneotherapy is to provide a rehabilitative and restorative effect on the body. According to the research findings, incorporating balneotherapy as a supplementary component to non-pharmacological treatments may potentially result in advantageous outcomes for individuals with SLE who are in remission or have minimal disease activity. Specifically, it may help to alleviate non-inflammatory pain and fatigue, thereby enhancing the QOL for these patients [258]. Physical exercise is an essential component of managing SLE, as it can effectively reduce fatigue and depression and enhance the overall QOL. In a study conducted on 20 patients diagnosed with SLE and 25 control patients, both groups underwent a three-month strengthening exercise program. The participants’ depression level was assessed using the self-rating depression scale, QOL was measured using a questionnaire, and the severity of fatigue was assessed using the fatigue severity scale. The study also utilized the six-minute walk test, two-minute step test, and body mass index (BMI) to evaluate the participants’ physical health before and after the three-month exercise program [259].
The inclusion of a psychoeducational program in the management of SLE patients resulted in an improvement in depression, anxiety, perceived stress, QOL, satisfaction with treatment, and medication adherence but did not have an impact on disease activity, according to a study that enrolled 80 SLE patients divided equally into intervention and control groups. The intervention group received 12 group sessions of psychotherapy and patient education, while disease activity was evaluated using the SLEDAI. Various questionnaires were administered, including the Symptom Checklist-90-Revised (SCL-90-R), perceived stress scale (PSS), Short Form 36 (SF36), Treatment Satisfaction Questionnaire for Medications (TSQM), and Medication Adherence Rating Scale-5 (MARS-5) [260].
Further research studies should be conducted on non-pharmacological interventions to enhance the QOL of SLE patients. These studies should involve larger sample sizes to increase statistical power and longer follow-up periods to obtain more comprehensive results.
Conclusions
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that requires individualized management. ANA positivity is the current qualifying factor for SLE criteria, but the diagnosis should be made through an individualized approach to avoid excluding potential patients from appropriate therapies. Females of reproductive age with SLE require careful management of fertility, pregnancy, and drug safety. Chronic inflammation associated with SLE leads to endothelial dysfunction, hastening vessel aging and raising cumulative cardiovascular risk. Thymectomy in myasthenia gravis (MG) patients has been associated with an increased risk of developing autoimmune diseases such as SLE, but it has no impact on cases of SLE that have already been diagnosed. The association between SLE and MG suggests a potential role for hydroxychloroquine (HCQ) in alleviating MG symptoms in SLE patients. Antiphospholipid syndrome (APS) is associated with an increased risk of thrombosis, pregnancy morbidity, and end-organ damage in SLE patients. Low-dose aspirin can protect against thrombosis in aPL-positive SLE patients. Non-pharmacological interventions such as exercise, psychotherapy, and balneotherapy can improve the quality of life of SLE patients. Optimal outcomes for SLE patients require a multidisciplinary approach involving rheumatologists, obstetricians, and other specialists.
Acknowledgments
All authors contributed equally to the study conceptualization, extensive literature review, manuscript writing, and critical proofreading prior to manuscript submission. Saleha Dar and Sabina Koirala share first authorship. Uzzam Ahmed Khawaja mentored and supervised the manuscript.
The authors have declared that no competing interests exist.
References
- 1.Systematic review of the epidemiology of systemic lupus erythematosus in the Asia-Pacific region: prevalence, incidence, clinical features, and mortality. Jakes RW, Bae SC, Louthrenoo W, Mok CC, Navarra SV, Kwon N. Arthritis Care Res (Hoboken) 2012;64:159–168. doi: 10.1002/acr.20683. [DOI] [PubMed] [Google Scholar]
- 2.Manifestations of systemic lupus erythematosus. Cojocaru M, Cojocaru IM, Silosi I, Vrabie CD. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3391953/ Maedica (Bucur) 2011;6:330–336. [PMC free article] [PubMed] [Google Scholar]
- 3.Review article: systemic lupus erythematosus: a review for anesthesiologists. Ben-Menachem E. Anesth Analg. 2010;111:665–676. doi: 10.1213/ANE.0b013e3181e8138e. [DOI] [PubMed] [Google Scholar]
- 4.Systemic lupus erythematosus and accelerated atherosclerosis. Hahn BH. N Engl J Med. 2003;349:2379–2380. doi: 10.1056/NEJMp038168. [DOI] [PubMed] [Google Scholar]
- 5.Monitoring systemic lupus erythematosus in standard clinical care. Petri M. Best Pract Res Clin Rheumatol. 2007;21:687–697. doi: 10.1016/j.berh.2007.01.003. [DOI] [PubMed] [Google Scholar]
- 6.Musculoskeletal involvement in systemic lupus erythematosus. Zoma A. Lupus. 2004;13:851–853. doi: 10.1191/0961203303lu2021oa. [DOI] [PubMed] [Google Scholar]
- 7.Identification of distinct gene expression profiles in the synovium of patients with systemic lupus erythematosus. Nzeusseu Toukap A, Galant C, Theate I, Maudoux AL, Lories RJ, Houssiau FA, Lauwerys BR. Arthritis Rheum. 2007;56:1579–1588. doi: 10.1002/art.22578. [DOI] [PubMed] [Google Scholar]
- 8.Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients. Cervera R, Khamashta MA, Font J, et al. Medicine (Baltimore) 2003;82:299–308. doi: 10.1097/01.md.0000091181.93122.55. [DOI] [PubMed] [Google Scholar]
- 9.Systemic lupus erythematosus. D’Cruz DP, Khamashta MA, Hughes GR. Lancet. 2007;369:587–596. doi: 10.1016/S0140-6736(07)60279-7. [DOI] [PubMed] [Google Scholar]
- 10.Transverse myelitis as the first manifestation of systemic lupus erythematosus or lupus-like disease: good functional outcome and relevance of antiphospholipid antibodies. D’Cruz DP, Mellor-Pita S, Joven B, et al. https://www.jrheum.org/content/31/2/280.long. J Rheumatol. 2004;31:280–285. [PubMed] [Google Scholar]
- 11.CNS lupus: a study of 41 patients. Joseph FG, Lammie GA, Scolding NJ. Neurology. 2007;69:644–654. doi: 10.1212/01.wnl.0000267320.48939.d0. [DOI] [PubMed] [Google Scholar]
- 12.Ocular manifestations of systemic lupus erythematosus. Arevalo JF, Lowder CY, Muci-Mendoza R. Curr Opin Ophthalmol. 2002;13:404–410. doi: 10.1097/00055735-200212000-00011. [DOI] [PubMed] [Google Scholar]
- 13.Ocular manifestations of systemic lupus erythematosus: a clinical review. Peponis V, Kyttaris VC, Tyradellis C, Vergados I, Sitaras NM. Lupus. 2006;15:3–12. doi: 10.1191/0961203306lu2250rr. [DOI] [PubMed] [Google Scholar]
- 14.Lupus update: perspective and clinical pearls. Manzi S. Cleve Clin J Med. 2009;76:137–142. doi: 10.3949/ccjm.76a.gr005. [DOI] [PubMed] [Google Scholar]
- 15.Antiphospholipid antibodies, systemic lupus erythematosus, and non-traumatic metatarsal fractures. Sangle S, D'Cruz DP, Khamashta MA, Hughes GR. Ann Rheum Dis. 2004;63:1241–1243. doi: 10.1136/ard.2003.016105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Prevalence and risk factors of osteoporosis in female SLE patients-extended report. Almehed K, Forsblad d'Elia H, Kvist G, Ohlsson C, Carlsten H. Rheumatology (Oxford) 2007;46:1185–1190. doi: 10.1093/rheumatology/kem105. [DOI] [PubMed] [Google Scholar]
- 17.2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Aringer M, Costenbader K, Daikh D, et al. Ann Rheum Dis. 2019;78:1151–1159. doi: 10.1136/annrheumdis-2018-214819. [DOI] [PubMed] [Google Scholar]
- 18.2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Aringer M, Costenbader K, Daikh D, et al. Arthritis Rheumatol. 2019;71:1400–1412. doi: 10.1002/art.40930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Hochberg MC. Arthritis Rheum. 1997;40:1725. doi: 10.1002/art.1780400928. [DOI] [PubMed] [Google Scholar]
- 20.Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus. Petri M, Orbai AM, Alarcón GS, et al. Arthritis Rheum. 2012;64:2677–2686. doi: 10.1002/art.34473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Classifying and diagnosing systemic lupus erythematosus in the 21st century. Aringer M, Johnson SR. Rheumatology (Oxford) 2020;59:0. doi: 10.1093/rheumatology/keaa379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Antinuclear antibody-negative systemic lupus erythematosus: how many patients and how to identify? Li H, Zheng Y, Chen L, Lin S. Arch Rheumatol. 2022;37:626–634. doi: 10.46497/ArchRheumatol.2022.9366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Leukoencephalopathy and cerebral edema as the presenting manifestations of SLE in an ANA-negative adolescent female: a case report and review of literature. Theisen A, Bose P, Knight C, Oliver M. Pediatr Rheumatol Online J. 2020;18:58. doi: 10.1186/s12969-020-00449-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.SLE classification criteria: science-based icons or algorithmic distractions - an intellectually demanding dilemma. [ May; 2023 ];Rekvig OP. Front Immunol. 2022 13:1011591. doi: 10.3389/fimmu.2022.1011591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors. Leadbetter EA, Rifkin IR, Hohlbaum AM, Beaudette BC, Shlomchik MJ, Marshak-Rothstein A. Nature. 2002;416:603–607. doi: 10.1038/416603a. [DOI] [PubMed] [Google Scholar]
- 26.Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9. Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD. J Clin Invest. 2005;115:407–417. doi: 10.1172/JCI23025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hydroxychloroquine use in the Baltimore Lupus Cohort: effects on lipids, glucose and thrombosis. Petri M. https://pubmed.ncbi.nlm.nih.gov/8803905/ Lupus. 1996;5 Suppl 1:0–22. [PubMed] [Google Scholar]
- 28.Cutting edge: antimalarial drugs inhibit IFN-β production through blockade of cyclic GMP-AMP synthase-DNA interaction. An J, Woodward JJ, Sasaki T, Minie M, Elkon KB. J Immunol. 2015;194:4089–4093. doi: 10.4049/jimmunol.1402793. [DOI] [PubMed] [Google Scholar]
- 29.Effect of hydroxychloroquine on the survival of patients with systemic lupus erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA L) Alarcón GS, McGwin G, Bertoli AM, et al. Ann Rheum Dis. 2007;66:1168–1172. doi: 10.1136/ard.2006.068676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Effect of antimalarials on thrombosis and survival in patients with systemic lupus erythematosus. Ruiz-Irastorza G, Egurbide MV, Pijoan JI, et al. Lupus. 2006;15:577–583. doi: 10.1177/0961203306071872. [DOI] [PubMed] [Google Scholar]
- 31.A randomized study of the effect of withdrawing hydroxychloroquine sulfate in systemic lupus erythematosus. Canadian Hydroxychloroquine Study Group. N Engl J Med. 1991;324:150–154. doi: 10.1056/NEJM199101173240303. [DOI] [PubMed] [Google Scholar]
- 32.Systemic lupus erythematosus in three ethnic groups: XVI. Association of hydroxychloroquine use with reduced risk of damage accrual. Fessler BJ, Alarcón GS, McGwin G Jr, et al. Arthritis Rheum. 2005;52:1473–1480. doi: 10.1002/art.21039. [DOI] [PubMed] [Google Scholar]
- 33.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. Fasano S, Pierro L, Pantano I, Iudici M, Valentini G. J Rheumatol. 2017;44:1032–1038. doi: 10.3899/jrheum.161351. [DOI] [PubMed] [Google Scholar]
- 34.Hydroxychloroquine use predicts complete renal remission within 12 months among patients treated with mycophenolate mofetil therapy for membranous lupus nephritis. Kasitanon N, Fine DM, Haas M, Magder LS, Petri M. Lupus. 2006;15:366–370. doi: 10.1191/0961203306lu2313oa. [DOI] [PubMed] [Google Scholar]
- 35.Seizure disorders in systemic lupus erythematosus results from an international, prospective, inception cohort study. Hanly JG, Urowitz MB, Su L, et al. Ann Rheum Dis. 2012;71:1502–1509. doi: 10.1136/annrheumdis-2011-201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Seizures in patients with systemic lupus erythematosus: data from LUMINA, a multiethnic cohort (LUMINA LIV) Andrade RM, Alarcón GS, González LA, et al. Ann Rheum Dis. 2008;67:829–834. doi: 10.1136/ard.2007.077594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Time to neuropsychiatric damage occurrence in LUMINA (LXVI): a multi-ethnic lupus cohort. González LA, Pons-Estel GJ, Zhang J, Vilá LM, Reveille JD, Alarcón GS. Lupus. 2009;18:822–830. doi: 10.1177/0961203309104392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Treatment of cutaneous lupus erythematosus with acitretin and hydroxychloroquine. Ruzicka T, Sommerburg C, Goerz G, Kind P, Mensing H. Br J Dermatol. 1992;127:513–518. doi: 10.1111/j.1365-2133.1992.tb14851.x. [DOI] [PubMed] [Google Scholar]
- 39.Cutaneous lupus erythematosus: update of therapeutic options part I. Kuhn A, Ruland V, Bonsmann G. J Am Acad Dermatol. 2011;65:0–93. doi: 10.1016/j.jaad.2010.06.018. [DOI] [PubMed] [Google Scholar]
- 40.Comparison of hydroxychloroquine and placebo in the treatment of the arthropathy of mild systemic lupus erythematosus. Williams HJ, Egger MJ, Singer JZ, et al. https://pubmed.ncbi.nlm.nih.gov/7983646/ J Rheumatol. 1994;21:1457–1462. [PubMed] [Google Scholar]
- 41.Antidiabetogenic effects of hydroxychloroquine on insulin sensitivity and beta cell function: a randomised trial. Wasko MC, McClure CK, Kelsey SF, Huber K, Orchard T, Toledo FG. Diabetologia. 2015;58:2336–2343. doi: 10.1007/s00125-015-3689-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Thrombosis and systemic lupus erythematosus: the Hopkins Lupus Cohort perspective. Petri M. Scand J Rheumatol. 1996;25:191–193. doi: 10.3109/03009749609069986. [DOI] [PubMed] [Google Scholar]
- 43.In vivo models of thrombosis for the antiphospholipid syndrome. Pierangeli SS, Harris EN. Lupus. 1996;5:451–455. doi: 10.1177/096120339600500524. [DOI] [PubMed] [Google Scholar]
- 44.Bone mineral density in postmenopausal Chinese patients with systemic lupus erythematosus. Mok CC, Mak A, Ma KM. Lupus. 2005;14:106–112. doi: 10.1191/0961203305lu2039oa. [DOI] [PubMed] [Google Scholar]
- 45.Risk of serious infection for patients with systemic lupus erythematosus starting glucocorticoids with or without antimalarials. Herrinton LJ, Liu L, Goldfien R, Michaels MA, Tran TN. J Rheumatol. 2016;43:1503–1509. doi: 10.3899/jrheum.150671. [DOI] [PubMed] [Google Scholar]
- 46.Hydroxychloroquine and risk of cancer in patients with primary Sjögren syndrome: propensity score matched landmark analysis. Fang YF, Chen YF, Chung TT, et al. Oncotarget. 2017;8:80461–80471. doi: 10.18632/oncotarget.19057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rates and predictors of hydroxychloroquine retinal toxicity in patients with rheumatoid arthritis and systemic lupus erythematosus. Wolfe F, Marmor MF. Arthritis Care Res (Hoboken) 2010;62:775–784. doi: 10.1002/acr.20133. [DOI] [PubMed] [Google Scholar]
- 48.Revised recommendations on screening for chloroquine and hydroxychloroquine retinopathy. Marmor MF, Kellner U, Lai TY, Lyons JS, Mieler WF. Ophthalmology. 2011;118:415–422. doi: 10.1016/j.ophtha.2010.11.017. [DOI] [PubMed] [Google Scholar]
- 49.A subset of patients with systemic lupus erythematosus fails to degrade DNA from multiple clinically relevant sources. Leffler J, Ciacma K, Gullstrand B, Bengtsson AA, Martin M, Blom AM. Arthritis Res Ther. 2015;17:205. doi: 10.1186/s13075-015-0726-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Fanouriakis A, Kostopoulou M, Alunno A, et al. Ann Rheum Dis. 2019;78:736–745. doi: 10.1136/annrheumdis-2019-215089. [DOI] [PubMed] [Google Scholar]
- 51.Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Schrezenmeier E, Dörner T. Nat Rev Rheumatol. 2020;16:155–166. doi: 10.1038/s41584-020-0372-x. [DOI] [PubMed] [Google Scholar]
- 52.Factors associated with damage accrual in patients with systemic lupus erythematosus: results from the Systemic Lupus International Collaborating Clinics (SLICC) Inception Cohort. Bruce IN, O'Keeffe AG, Farewell V, et al. Ann Rheum Dis. 2015;74:1706–1713. doi: 10.1136/annrheumdis-2013-205171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cumulative burden of glucocorticoid-related adverse events in patients with systemic lupus erythematosus: findings from a 12-year longitudinal study. Chen HL, Shen LJ, Hsu PN, Shen CY, Hall SA, Hsiao FY. J Rheumatol. 2018;45:83–89. doi: 10.3899/jrheum.160214. [DOI] [PubMed] [Google Scholar]
- 54.From childhood to adulthood: the trajectory of damage in patients with juvenile-onset systemic lupus erythematosus. Lim LS, Pullenayegum E, Lim L, Gladman D, Feldman B, Silverman E. Arthritis Care Res (Hoboken) 2017;69:1627–1635. doi: 10.1002/acr.23199. [DOI] [PubMed] [Google Scholar]
- 55.Birmingham SLE cohort: outcomes of a large inception cohort followed for up to 21 years. Yee CS, Su L, Toescu V, et al. Rheumatology (Oxford) 2015;54:836–843. doi: 10.1093/rheumatology/keu412. [DOI] [PubMed] [Google Scholar]
- 56.Both prolonged remission and Lupus Low Disease Activity State are associated with reduced damage accrual in systemic lupus erythematosus. Tsang-A-Sjoe MW, Bultink IE, Heslinga M, Voskuyl AE. Rheumatology (Oxford) 2017;56:121–128. doi: 10.1093/rheumatology/kew377. [DOI] [PubMed] [Google Scholar]
- 57.Prednisone, lupus activity, and permanent organ damage. Thamer M, Hernán MA, Zhang Y, Cotter D, Petri M. J Rheumatol. 2009;36:560–564. doi: 10.3899/jrheum.080828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Comparison of high versus low-medium prednisone doses for the treatment of systemic lupus erythematosus patients with high activity at diagnosis. Ruiz-Arruza I, Barbosa C, Ugarte A, Ruiz-Irastorza G. Autoimmun Rev. 2015;14:875–879. doi: 10.1016/j.autrev.2015.05.011. [DOI] [PubMed] [Google Scholar]
- 59.Effect of corticosteroid use by dose on the risk of developing organ damage over time in systemic lupus erythematosus-the Hopkins Lupus Cohort. Al Sawah S, Zhang X, Zhu B, Magder LS, Foster SA, Iikuni N, Petri M. Lupus Sci Med. 2015;2:0. doi: 10.1136/lupus-2014-000066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Flares in lupus: Outcome Assessment Trial (FLOAT), a comparison between oral methylprednisolone and intramuscular triamcinolone. Danowski A, Magder L, Petri M. https://www.jrheum.org/content/33/1/57.long. J Rheumatol. 2006;33:57–60. [PubMed] [Google Scholar]
- 61.Risk of serious infections with immunosuppressive drugs and glucocorticoids for lupus nephritis: a systematic review and network meta-analysis. Singh JA, Hossain A, Kotb A, Wells G. BMC Med. 2016;14:137. doi: 10.1186/s12916-016-0673-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Synthetic pharmacotherapy for systemic lupus erythematosus: potential mechanisms of action, efficacy, and safety. Téllez Arévalo AM, Quaye A, Rojas-Rodríguez LC, Poole BD, Baracaldo-Santamaría D, Tellez Freitas CM. Medicina (Kaunas) 2022;59 doi: 10.3390/medicina59010056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Brown PM, Pratt AG, Isaacs JD. Nat Rev Rheumatol. 2016;12:731–742. doi: 10.1038/nrrheum.2016.175. [DOI] [PubMed] [Google Scholar]
- 64.Double blind, randomized, placebo controlled clinical trial of methotrexate in systemic lupus erythematosus. Carneiro JR, Sato EI. https://pubmed.ncbi.nlm.nih.gov/10381042/ J Rheumatol. 1999;26:1275–1279. [PubMed] [Google Scholar]
- 65.Steroid-sparing effects of methotrexate in systemic lupus erythematosus: a double-blind, randomized, placebo-controlled trial. Fortin PR, Abrahamowicz M, Ferland D, Lacaille D, Smith CD, Zummer M. Arthritis Rheum. 2008;59:1796–1804. doi: 10.1002/art.24068. [DOI] [PubMed] [Google Scholar]
- 66.Efficacy and tolerability of methotrexate in antimalarial resistant lupus arthritis. Rahman P, Humphrey-Murto S, Gladman DD, Urowitz MB. https://pubmed.ncbi.nlm.nih.gov/9489814/ J Rheumatol. 1998;25:243–246. [PubMed] [Google Scholar]
- 67.Cutaneous lupus erythematosus: update of therapeutic options part II. Kuhn A, Ruland V, Bonsmann G. J Am Acad Dermatol. 2011;65:0–213. doi: 10.1016/j.jaad.2010.06.018. [DOI] [PubMed] [Google Scholar]
- 68.Frangou EA, Bertsias G, Boumpas DT. Systemic lupus erythematosus: basic, applied and clinical aspects. Academic Press; 2016. Chapter 62 - cytotoxic-immunosuppressive drug treatment; pp. 533–541. [Google Scholar]
- 69.Characteristics related to early secondary amenorrhoea and pregnancy among women diagnosed with systemic lupus erythematosus: an analysis using the GOAL study. Knight JH, Howards PP, Spencer JB, Tsagaris KC, Lim SS. Lupus Sci Med. 2016;3:0. doi: 10.1136/lupus-2015-000139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Brief report: the Euro-lupus low-dose intravenous cyclophosphamide regimen does not impact the ovarian reserve, as measured by serum levels of anti-Müllerian hormone. Tamirou F, Husson SN, Gruson D, Debiève F, Lauwerys BR, Houssiau FA. Arthritis Rheumatol. 2017;69:1267–1271. doi: 10.1002/art.40079. [DOI] [PubMed] [Google Scholar]
- 71.Anti-müllerian hormone and ovarian reserve in systemic lupus erythematosus. Mok CC, Chan PT, To CH. Arthritis Rheum. 2013;65:206–210. doi: 10.1002/art.37719. [DOI] [PubMed] [Google Scholar]
- 72.EULAR recommendations for women's health and the management of family planning, assisted reproduction, pregnancy and menopause in patients with systemic lupus erythematosus and/or antiphospholipid syndrome. Andreoli L, Bertsias GK, Agmon-Levin N, et al. Ann Rheum Dis. 2017;76:476–485. doi: 10.1136/annrheumdis-2016-209770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gonadotropin releasing hormone agonists may minimize cyclophosphamide associated gonadotoxicity in SLE and autoimmune diseases. Blumenfeld Z, Mischari O, Schultz N, Boulman N, Balbir-Gurman A. Semin Arthritis Rheum. 2011;41:346–352. doi: 10.1016/j.semarthrit.2011.05.008. [DOI] [PubMed] [Google Scholar]
- 74.Adjunctive GnRH-a treatment attenuates depletion of ovarian reserve associated with cyclophosphamide therapy in premenopausal SLE patients. Marder W, McCune WJ, Wang L, et al. Gynecol Endocrinol. 2012;28:624–627. doi: 10.3109/09513590.2011.650752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lymphoma risk in systemic lupus: effects of disease activity versus treatment. Bernatsky S, Ramsey-Goldman R, Joseph L, et al. Ann Rheum Dis. 2014;73:138–142. doi: 10.1136/annrheumdis-2012-202099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cumulative immunosuppressant exposure is associated with diversified cancer risk among 14 832 patients with systemic lupus erythematosus: a nested case-control study. Hsu CY, Lin MS, Su YJ, et al. Rheumatology (Oxford) 2017;56:620–628. doi: 10.1093/rheumatology/kew457. [DOI] [PubMed] [Google Scholar]
- 77.Current and novel therapeutics in the treatment of systemic lupus erythematosus. Yildirim-Toruner C, Diamond B. J Allergy Clin Immunol. 2011;127:303–312. doi: 10.1016/j.jaci.2010.12.1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Long-term maintenance therapy with azathioprine in systemic lupus erythematosus. Ginzler E, Sharon E, Diamond H, Kaplan D. Arthritis Rheum. 1975;18:27–34. doi: 10.1002/art.1780180106. [DOI] [PubMed] [Google Scholar]
- 79.Azathioprine in the treatment of systemic lupus erythematosus. A controlled study. Sztejnbok M, Stewart A, Diamond H, Kaplan D. Arthritis Rheum. 1971;14:639–645. doi: 10.1002/art.1780140511. [DOI] [PubMed] [Google Scholar]
- 80.The BILAG multi-centre open randomized controlled trial comparing ciclosporin vs azathioprine in patients with severe SLE. Griffiths B, Emery P, Ryan V, et al. Rheumatology (Oxford) 2010;49:723–732. doi: 10.1093/rheumatology/kep396. [DOI] [PubMed] [Google Scholar]
- 81.Azathioprine use during pregnancy: unexpected intrauterine exposure to metabolites. de Boer NK, Jarbandhan SV, de Graaf P, Mulder CJ, van Elburg RM, van Bodegraven AA. Am J Gastroenterol. 2006;101:1390–1392. doi: 10.1111/j.1572-0241.2006.00538.x. [DOI] [PubMed] [Google Scholar]
- 82.Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology. Broen JC, van Laar JM. Nat Rev Rheumatol. 2020;16:167–178. doi: 10.1038/s41584-020-0374-8. [DOI] [PubMed] [Google Scholar]
- 83.Mycophenolate mofetil for non-renal manifestations of systemic lupus erythematosus: a systematic review. Mok CC. Scand J Rheumatol. 2007;36:329–337. doi: 10.1080/03009740701607042. [DOI] [PubMed] [Google Scholar]
- 84.Mycophenolate mofetil in nonrenal manifestations of systemic lupus erythematosus: an observational cohort study. Tselios K, Gladman DD, Su J, Urowitz MB. J Rheumatol. 2016;43:552–558. doi: 10.3899/jrheum.150779. [DOI] [PubMed] [Google Scholar]
- 85.Mycophenolate mofetil for induction treatment of lupus nephritis: a systematic review and metaanalysis. Touma Z, Gladman DD, Urowitz MB, Beyene J, Uleryk EM, Shah PS. J Rheumatol. 2011;38:69–78. doi: 10.3899/jrheum.100130. [DOI] [PubMed] [Google Scholar]
- 86.Enteric-coated mycophenolate sodium versus azathioprine in patients with active systemic lupus erythematosus: a randomised clinical trial. Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, et al. Ann Rheum Dis. 2017;76:1575–1582. doi: 10.1136/annrheumdis-2016-210882. [DOI] [PubMed] [Google Scholar]
- 87.FK506 ameliorates proteinuria and glomerular lesions induced by anti-Thy 1.1 monoclonal antibody 1-22-3. Ikezumi Y, Kanno K, Koike H, Tomita M, Uchiyama M, Shimizu F, Kawachi H. Kidney Int. 2002;61:1339–1350. doi: 10.1046/j.1523-1755.2002.00259.x. [DOI] [PubMed] [Google Scholar]
- 88.Tacrolimus protects podocytes from injury in lupus nephritis partly by stabilizing the cytoskeleton and inhibiting podocyte apoptosis. Liao R, Liu Q, Zheng Z, et al. PLoS One. 2015;10:0. doi: 10.1371/journal.pone.0132724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.The actin cytoskeleton of kidney podocytes is a direct target of the antiproteinuric effect of cyclosporine A. Faul C, Donnelly M, Merscher-Gomez S, et al. Nat Med. 2008;14:931–938. doi: 10.1038/nm.1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Multitarget therapy for induction treatment of lupus nephritis: a randomized trial. Liu Z, Zhang H, Liu Z, et al. Ann Intern Med. 2015;162:18–26. doi: 10.7326/M14-1030. [DOI] [PubMed] [Google Scholar]
- 91.Tacrolimus versus cyclophosphamide as treatment for diffuse proliferative or membranous lupus nephritis: a non-randomized prospective cohort study. Wang S, Li X, Qu L, et al. Lupus. 2012;21:1025–1035. doi: 10.1177/0961203312448105. [DOI] [PubMed] [Google Scholar]
- 92.Low-dose tacrolimus in treating lupus nephritis refractory to cyclophosphamide: a prospective cohort study. Fei Y, Wu Q, Zhang W, et al. https://www.clinexprheumatol.org/abstract.asp?a=5805. Clin Exp Rheumatol. 2013;31:62–68. [PubMed] [Google Scholar]
- 93.Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: a randomised controlled trial and long-term follow-up. Mok CC, Ying KY, Yim CW, Siu YP, Tong KH, To CH, Ng WL. Ann Rheum Dis. 2016;75:30–36. doi: 10.1136/annrheumdis-2014-206456. [DOI] [PubMed] [Google Scholar]
- 94.Tacrolimus use in lupus nephritis: a systematic review and meta-analysis. Hannah J, Casian A, D'Cruz D. Autoimmun Rev. 2016;15:93–101. doi: 10.1016/j.autrev.2015.09.006. [DOI] [PubMed] [Google Scholar]
- 95.Lupus nephritis and pregnancy: concerns and management. Lightstone L, Hladunewich MA. Semin Nephrol. 2017;37:347–353. doi: 10.1016/j.semnephrol.2017.05.006. [DOI] [PubMed] [Google Scholar]
- 96.Management strategies and future directions for systemic lupus erythematosus in adults. Durcan L, O’Dwyer T, Petri M. Lancet. 2019;393:2332–2343. doi: 10.1016/S0140-6736(19)30237-5. [DOI] [PubMed] [Google Scholar]
- 97.A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Furie R, Petri M, Zamani O, et al. Arthritis Rheum. 2011;63:3918–3930. doi: 10.1002/art.30613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Navarra SV, Guzmán RM, Gallacher AE, et al. Lancet. 2011;377:721–731. doi: 10.1016/S0140-6736(10)61354-2. [DOI] [PubMed] [Google Scholar]
- 99.Clinical predictors of response and discontinuation of belimumab in patients with systemic lupus erythematosus in real life setting. Results of a large, multicentric, nationwide study. Iaccarino L, Andreoli L, Bocci EB, et al. J Autoimmun. 2018;86:1–8. doi: 10.1016/j.jaut.2017.09.004. [DOI] [PubMed] [Google Scholar]
- 100.Effects of belimumab, a B lymphocyte stimulator-specific inhibitor, on disease activity across multiple organ domains in patients with systemic lupus erythematosus: combined results from two phase III trials. Manzi S, Sánchez-Guerrero J, Merrill JT, et al. Ann Rheum Dis. 2012;71:1833–1838. doi: 10.1136/annrheumdis-2011-200831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Belimumab in the treatment of systemic lupus erythematosus: high disease activity predictors of response. van Vollenhoven RF, Petri MA, Cervera R, et al. Ann Rheum Dis. 2012;71:1343–1349. doi: 10.1136/annrheumdis-2011-200937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Efficacy of rituximab in 164 patients with biopsy-proven lupus nephritis: pooled data from European cohorts. Díaz-Lagares C, Croca S, Sangle S, et al. Autoimmun Rev. 2012;11:357–364. doi: 10.1016/j.autrev.2011.10.009. [DOI] [PubMed] [Google Scholar]
- 103.Rituximab in systemic lupus erythematosus: an updated systematic review and meta-analysis. Duxbury B, Combescure C, Chizzolini C. Lupus. 2013;22:1489–1503. doi: 10.1177/0961203313509295. [DOI] [PubMed] [Google Scholar]
- 104.Rituximab in systemic lupus erythematosus: a systematic review of off-label use in 188 cases. Ramos-Casals M, Soto MJ, Cuadrado MJ, Khamashta MA. Lupus. 2009;18:767–776. doi: 10.1177/0961203309106174. [DOI] [PubMed] [Google Scholar]
- 105.Efficacy and safety of off-label use of rituximab in refractory lupus: data from the Italian Multicentre Registry. Iaccarino L, Bartoloni E, Carli L, et al. https://www.clinexprheumatol.org/abstract.asp?a=8681. Clin Exp Rheumatol. 2015;33:449–456. [PubMed] [Google Scholar]
- 106.Rituximab therapy has a rapid and durable response for refractory cytopenia in childhood-onset systemic lupus erythematosus. Olfat M, Silverman ED, Levy DM. Lupus. 2015;24:966–972. doi: 10.1177/0961203315578764. [DOI] [PubMed] [Google Scholar]
- 107.Safety and efficacy of rituximab in systemic lupus erythematosus: results from 136 patients from the French AutoImmunity and Rituximab registry. Terrier B, Amoura Z, Ravaud P, et al. Arthritis Rheum. 2010;62:2458–2466. doi: 10.1002/art.27541. [DOI] [PubMed] [Google Scholar]
- 108.Rituximab plus standard of care for treatment of primary immune thrombocytopenia: a systematic review and meta-analysis. Chugh S, Darvish-Kazem S, Lim W, et al. Lancet Haematol. 2015;2:0–81. doi: 10.1016/S2352-3026(15)00003-4. [DOI] [PubMed] [Google Scholar]
- 109.Rituximab and mycophenolate mofetil for relapsing proliferative lupus nephritis: a long-term prospective study. Boletis JN, Marinaki S, Skalioti C, Lionaki SS, Iniotaki A, Sfikakis PP. Nephrol Dial Transplant. 2009;24:2157–2160. doi: 10.1093/ndt/gfp002. [DOI] [PubMed] [Google Scholar]
- 110.Systemic lupus erythematosus and other autoimmune rheumatic diseases: challenges to treatment. Murphy G, Lisnevskaia L, Isenberg D. Lancet. 2013;382:809–818. doi: 10.1016/S0140-6736(13)60889-2. [DOI] [PubMed] [Google Scholar]
- 111.Update οn the diagnosis and management of systemic lupus erythematosus. Fanouriakis A, Tziolos N, Bertsias G, Boumpas DT. Ann Rheum Dis. 2021;80:14–25. doi: 10.1136/annrheumdis-2020-218272. [DOI] [PubMed] [Google Scholar]
- 112.Global epidemiology of systemic lupus erythematosus: a comprehensive systematic analysis and modelling study. Tian J, Zhang D, Yao X, Huang Y, Lu Q. Ann Rheum Dis. 2023;82:351–356. doi: 10.1136/ard-2022-223035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Epidemiology of systemic lupus erythematosus: an update. Stojan G, Petri M. Curr Opin Rheumatol. 2018;30:144–150. doi: 10.1097/BOR.0000000000000480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Prevalence of adult systemic lupus erythematosus in California and Pennsylvania in 2000: estimates obtained using hospitalization data. Chakravarty EF, Bush TM, Manzi S, Clarke AE, Ward MM. Arthritis Rheum. 2007;56:2092–2094. doi: 10.1002/art.22641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.The role of sex hormones in systemic lupus erythematosus. Lahita RG. Curr Opin Rheumatol. 1999;11:352–356. doi: 10.1097/00002281-199909000-00005. [DOI] [PubMed] [Google Scholar]
- 116.Evaluation of ovarian reserve tests in women with systemic lupus erythematosus. Ulug P, Oner G, Kasap B, Akbas EM, Ozcicek F. Am J Reprod Immunol. 2014;72:85–88. doi: 10.1111/aji.12249. [DOI] [PubMed] [Google Scholar]
- 117.Menstrual disturbances in patients with systemic lupus erythematosus without alkylating therapy: clinical, hormonal and therapeutic associations. Pasoto SG, Mendonça BB, Bonfá E. Lupus. 2002;11:175–180. doi: 10.1191/0961203302lu163oa. [DOI] [PubMed] [Google Scholar]
- 118.Protein-losing enteropathy and premature ovarian failure in a young woman with systemic lupus erythematosus. Chang YS, Lai CC, Chen WS, Wang SH, Chou CT, Tsai CY. Lupus. 2012;21:1237–1239. doi: 10.1177/0961203312449492. [DOI] [PubMed] [Google Scholar]
- 119.Reproductive aspects of systemic lupus erythematosus. Oktem O, Yagmur H, Bengisu H, Urman B. J Reprod Immunol. 2016;117:57–65. doi: 10.1016/j.jri.2016.07.001. [DOI] [PubMed] [Google Scholar]
- 120.Premature ovarian failure in patients affected by systemic lupus erythematosus: a cross-sectional study. Ceccarelli F, Orefice V, Perrone G, et al. https://www.clinexprheumatol.org/abstract.asp?a=14151. Clin Exp Rheumatol. 2020;38:450–454. [PubMed] [Google Scholar]
- 121.Pregnancy and management in women with rheumatoid arthritis, systemic lupus erythematosus, and obstetric antiphospholipid syndrome. Castro-Gutierrez A, Young K, Bermas BL. Med Clin North Am. 2021;105:341–353. doi: 10.1016/j.mcna.2020.10.002. [DOI] [PubMed] [Google Scholar]
- 122.A better family plan. [ May; 2023 ];https://www.the-rheumatologist.org/article/a-better-family-plan/5/ 2007 5:8–3. [Google Scholar]
- 123.Risk of unintended pregnancy among women with systemic lupus erythematosus. Schwarz EB, Manzi S. Arthritis Rheum. 2008;59:863–866. doi: 10.1002/art.23712. [DOI] [PubMed] [Google Scholar]
- 124.Contraceptive counseling and use among women with systemic lupus erythematosus: a gap in health care quality? Yazdany J, Trupin L, Kaiser R, Schmajuk G, Gillis JZ, Chakravarty E, Schwarz EB. Arthritis Care Res (Hoboken) 2011;63:358–365. doi: 10.1002/acr.20402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Summary of recommendations for contraceptives in SLE/APS patients. [ Apr; 2023 ]. 2023. https://www.uptodate.com/contents/image?csi=d04efd99-2dff-473e-8fcb-adde5945abc3&source=contentShare&imageKey=RHEUM%2F98708 https://www.uptodate.com/contents/image?csi=d04efd99-2dff-473e-8fcb-adde5945abc3&source=contentShare&imageKey=RHEUM%2F98708
- 126.A trial of contraceptive methods in women with systemic lupus erythematosus. Sánchez-Guerrero J, Uribe AG, Jiménez-Santana L, Mestanza-Peralta M, Lara-Reyes P, Seuc AH, Cravioto MD. N Engl J Med. 2005;353:2539–2549. doi: 10.1056/NEJMoa050817. [DOI] [PubMed] [Google Scholar]
- 127.Treatment of menorrhagia associated with oral anticoagulation: efficacy and safety of the levonorgestrel releasing intrauterine device (Mirena coil) Pisoni CN, Cuadrado MJ, Khamashta MA, Hunt BJ. Lupus. 2006;15:877–880. doi: 10.1177/0961203306071706. [DOI] [PubMed] [Google Scholar]
- 128.A randomized trial of the intrauterine contraceptive device vs hormonal contraception in women who are infected with the human immunodeficiency virus. Stringer EM, Kaseba C, Levy J, et al. Am J Obstet Gynecol. 2007;197:144–148. doi: 10.1016/j.ajog.2007.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.U.S. medical eligibility criteria for contraceptive use, 2016. Curtis KM, Tepper NK, Jatlaoui TC, et al. MMWR Recomm Rep. 2016;65:1–103. doi: 10.15585/mmwr.rr6503a1. [DOI] [PubMed] [Google Scholar]
- 130.Assessing the risk of venous thromboembolic events in women taking progestin-only contraception: a meta-analysis. Mantha S, Karp R, Raghavan V, Terrin N, Bauer KA, Zwicker JI. BMJ. 2012;345:0. doi: 10.1136/bmj.e4944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Combined oral contraceptives in women with systemic lupus erythematosus. Petri M, Kim MY, Kalunian KC, et al. N Engl J Med. 2005;353:2550–2558. doi: 10.1056/NEJMoa051135. [DOI] [PubMed] [Google Scholar]
- 132.Antiphospholipid antibodies and risk of myocardial infarction and ischaemic stroke in young women in the ratio study: a case-control study. Urbanus RT, Siegerink B, Roest M, Rosendaal FR, de Groot PG, Algra A. Lancet Neurol. 2009;8:998–1005. doi: 10.1016/S1474-4422(09)70239-X. [DOI] [PubMed] [Google Scholar]
- 133.Comparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptive. van den Heuvel MW, van Bragt AJ, Alnabawy AK, Kaptein MC. Contraception. 2005;72:168–174. doi: 10.1016/j.contraception.2005.03.005. [DOI] [PubMed] [Google Scholar]
- 134.Prevalence of low bone mass and osteoporosis in long-term users of the injectable contraceptive depot medroxyprogesterone acetate. Modesto W, Bahamondes MV, Bahamondes L. J Womens Health (Larchmt) 2015;24:636–640. doi: 10.1089/jwh.2014.5077. [DOI] [PubMed] [Google Scholar]
- 135.Predictors of maternal and fetal outcomes in pregnancies of patients with systemic lupus erythematosus. Kwok LW, Tam LS, Zhu T, Leung YY, Li E. Lupus. 2011;20:829–836. doi: 10.1177/0961203310397967. [DOI] [PubMed] [Google Scholar]
- 136.Factors that predict prematurity and preeclampsia in pregnancies that are complicated by systemic lupus erythematosus. Chakravarty EF, Colón I, Langen ES, Nix DA, El-Sayed YY, Genovese MC, Druzin ML. Am J Obstet Gynecol. 2005;192:1897–1904. doi: 10.1016/j.ajog.2005.02.063. [DOI] [PubMed] [Google Scholar]
- 137.Obstetric hospitalizations in the United States for women with systemic lupus erythematosus and rheumatoid arthritis. Chakravarty EF, Nelson L, Krishnan E. Arthritis Rheum. 2006;54:899–907. doi: 10.1002/art.21663. [DOI] [PubMed] [Google Scholar]
- 138.A national study of the complications of lupus in pregnancy. Clowse ME, Jamison M, Myers E, James AH. Am J Obstet Gynecol. 2008;199:127–126. doi: 10.1016/j.ajog.2008.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Maternal and perinatal outcomes of pregnancies in systemic lupus erythematosus: a nationwide population-based study. Chen YJ, Chang JC, Lai EL, et al. Semin Arthritis Rheum. 2020;50:451–457. doi: 10.1016/j.semarthrit.2020.01.014. [DOI] [PubMed] [Google Scholar]
- 140.Influence of disease activity and medications on offspring birth weight, pre-eclampsia and preterm birth in systemic lupus erythematosus: a population-based study. Skorpen CG, Lydersen S, Gilboe IM, et al. Ann Rheum Dis. 2018;77:264–269. doi: 10.1136/annrheumdis-2017-211641. [DOI] [PubMed] [Google Scholar]
- 141.The impact of increased lupus activity on obstetric outcomes. Clowse ME, Magder LS, Witter F, Petri M. Arthritis Rheum. 2005;52:514–521. doi: 10.1002/art.20864. [DOI] [PubMed] [Google Scholar]
- 142.Impact of previous lupus nephritis on maternal and fetal outcomes during pregnancy. Saavedra MA, Cruz-Reyes C, Vera-Lastra O, Romero GT, Cruz-Cruz P, Arias-Flores R, Jara LJ. Clin Rheumatol. 2012;31:813–819. doi: 10.1007/s10067-012-1941-4. [DOI] [PubMed] [Google Scholar]
- 143.Maternal and foetal outcomes in pregnant patients with active lupus nephritis. Wagner SJ, Craici I, Reed D, et al. Lupus. 2009;18:342–347. doi: 10.1177/0961203308097575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Anti-SSA/Ro positivity and congenital heart block: obstetric and foetal outcome in a cohort of anti-SSA/Ro positive pregnant patients with and without autoimmune diseases. Fredi M, Argolini LM, Angeli F, et al. Clin Exp Rheumatol. 2023;41:685–693. doi: 10.55563/clinexprheumatol/2ju0yv. [DOI] [PubMed] [Google Scholar]
- 145.Neonatal lupus erythematosus - prevention is better than cure. Liszewska A, Woźniacka A. Postepy Dermatol Alergol. 2022;39:1021–1026. doi: 10.5114/ada.2022.122601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Brief report: changes in antiphospholipid antibody titers during pregnancy: effects on pregnancy outcomes. Yelnik CM, Porter TF, Branch DW, et al. Arthritis Rheumatol. 2016;68:1964–1969. doi: 10.1002/art.39668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Hydroxychloroquine and lupus pregnancy: review of a series of 36 cases. Buchanan NM, Toubi E, Khamashta MA, Lima F, Kerslake S, Hughes GR. Ann Rheum Dis. 1996;55:486–488. doi: 10.1136/ard.55.7.486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Effect of pregnancy on disease flares in patients with systemic lupus erythematosus. Eudy AM, Siega-Riz AM, Engel SM, Franceschini N, Howard AG, Clowse ME, Petri M. Ann Rheum Dis. 2018;77:855–860. doi: 10.1136/annrheumdis-2017-212535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Use of antimalarial drugs is associated with a lower risk of preeclampsia in lupus pregnancy: a prospective cohort study. Saavedra MÁ, Miranda-Hernández D, Lara-Mejía A, et al. Int J Rheum Dis. 2020;23:633–640. doi: 10.1111/1756-185X.13830. [DOI] [PubMed] [Google Scholar]
- 150.Systemic lupus erythematosus management in pregnancy. Dao KH, Bermas BL. Int J Womens Health. 2022;14:199–211. doi: 10.2147/IJWH.S282604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Hydroxychloroquine in pregnant patients with systemic lupus erythematosus. Parke A, West B. https://pubmed.ncbi.nlm.nih.gov/8895146/ J Rheumatol. 1996;23:1715–1718. [PubMed] [Google Scholar]
- 152.Hydroxychloroquine (HCQ) in lupus pregnancy: double-blind and placebo-controlled study. Levy RA, Vilela VS, Cataldo MJ, et al. Lupus. 2001;10:401–404. doi: 10.1191/096120301678646137. [DOI] [PubMed] [Google Scholar]
- 153.Maternal systemic lupus erythematosus (SLE) high risk for preterm delivery and not for long-term neurological morbidity of the offspring. Davidov D, Sheiner E, Wainstock T, Miodownik S, Pariente G. J Clin Med. 2021;10 doi: 10.3390/jcm10132952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Aspirin use to prevent preeclampsia and related morbidity and mortality: US Preventive Services Task Force recommendation statement. Davidson KW, Barry MJ, Mangione CM, et al. JAMA. 2021;326:1186–1191. doi: 10.1001/jama.2021.14781. [DOI] [PubMed] [Google Scholar]
- 155.Corticosteroid use during pregnancy and risk of orofacial clefts. Hviid A, Mølgaard-Nielsen D. CMAJ. 2011;183:796–804. doi: 10.1503/cmaj.101063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Rheumatoid arthritis medications and lactation. Sammaritano LR, Bermas BL. Curr Opin Rheumatol. 2014;26:354–360. doi: 10.1097/BOR.0000000000000055. [DOI] [PubMed] [Google Scholar]
- 157.Commissioner O of the. Nonsteroidal anti-inflammatory drugs (NSAIDs): drug safety communication - avoid use of NSAIDs in pregnancy at 20 weeks or later. [ May; 2023 ]. 2020. https://www.fda.gov/safety/medical-product-safety-information/nonsteroidal-anti-inflammatory-drugs-nsaids-drug-safety-communication-avoid-use-nsaids-pregnancy-20 https://www.fda.gov/safety/medical-product-safety-information/nonsteroidal-anti-inflammatory-drugs-nsaids-drug-safety-communication-avoid-use-nsaids-pregnancy-20
- 158.FDA recommends avoiding use of NSAIDs in pregnancy at 20 weeks or later because they can result in low amniotic fluid. [ May; 2023 ]. 2022. https://www.fda.gov/drugs/fda-drug-safety-podcasts/fda-recommends-avoiding-use-nsaids-pregnancy-20-weeks-or-later-because-they-can-result-low-amniotic https://www.fda.gov/drugs/fda-drug-safety-podcasts/fda-recommends-avoiding-use-nsaids-pregnancy-20-weeks-or-later-because-they-can-result-low-amniotic
- 159.Nonsteroidal anti-inflammatory drugs during pregnancy and the initiation of lactation. Bloor M, Paech M. Anesth Analg. 2013;116:1063–1075. doi: 10.1213/ANE.0b013e31828a4b54. [DOI] [PubMed] [Google Scholar]
- 160.Azathioprine during pregnancy in systemic lupus erythematosus patients is not associated with poor fetal outcome. Saavedra MÁ, Sánchez A, Morales S, Ángeles U, Jara LJ. Clin Rheumatol. 2015;34:1211–1216. doi: 10.1007/s10067-015-2987-x. [DOI] [PubMed] [Google Scholar]
- 161.The efficacy of adjunct tacrolimus treatment in pregnancy outcomes in patients with systemic lupus erythematosus. Ichinose K, Sato S, Kitajima Y, et al. Lupus. 2018;27:1312–1320. doi: 10.1177/0961203318770536. [DOI] [PubMed] [Google Scholar]
- 162.Safety of tacrolimus use during pregnancy and related pregnancy outcomes in patients with systemic lupus erythematosus: a retrospective single-center analysis in Japan. Kitada A, Nakai T, Fukui S, et al. Lupus. 2023;32:352–362. doi: 10.1177/09612033221148334. [DOI] [PubMed] [Google Scholar]
- 163.Cyclosporine in the treatment of lupus nephritis including two patients treated during pregnancy. Hussein MM, Mooij JM, Roujouleh H. https://pubmed.ncbi.nlm.nih.gov/8403571/ Clin Nephrol. 1993;40:160–163. [PubMed] [Google Scholar]
- 164.Pregnancy outcomes in patients treated with belimumab: report from real-world experience. Kao JH, Lan TY, Lu CH, Cheng CF, Huang YM, Shen CY, Hsieh SC. Semin Arthritis Rheum. 2021;51:963–968. doi: 10.1016/j.semarthrit.2021.06.005. [DOI] [PubMed] [Google Scholar]
- 165.Different pregnancy outcomes in patients with systemic lupus erythematosus treated with belimumab. Lai Y, Li B, Huang J, et al. Lupus. 2023;32:149–154. doi: 10.1177/09612033221141805. [DOI] [PubMed] [Google Scholar]
- 166.Dilemma of belimumab therapy (dis)continuation during pregnancy: results of a retrospective study in eudravigilance. Ghalandari N, Crijns HJ, Dolhain RJ, Hazes JM, van Puijenbroek EP. Lupus. 2023;32:189–197. doi: 10.1177/09612033221143286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. Ann Rheum Dis. 2016;75:795–810. doi: 10.1136/annrheumdis-2015-208840. [DOI] [PubMed] [Google Scholar]
- 168.BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding-part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Flint J, Panchal S, Hurrell A, et al. Rheumatology (Oxford) 2016;55:1693–1697. doi: 10.1093/rheumatology/kev404. [DOI] [PubMed] [Google Scholar]
- 169.BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding-part II: analgesics and other drugs used in rheumatology practice. Flint J, Panchal S, Hurrell A, et al. Rheumatology (Oxford) 2016;55:1698–1702. doi: 10.1093/rheumatology/kev405. [DOI] [PubMed] [Google Scholar]
- 170.How safe are anti-rheumatic drugs during pregnancy? Østensen M, Förger F. Curr Opin Pharmacol. 2013;13:470–475. doi: 10.1016/j.coph.2013.03.004. [DOI] [PubMed] [Google Scholar]
- 171.Cyclophosphamide teratogenesis: a review. Mirkes PE. Teratog Carcinog Mutagen. 1985;5:75–88. doi: 10.1002/tcm.1770050202. [DOI] [PubMed] [Google Scholar]
- 172.Risk of pregnancy loss in patients exposed to mycophenolate compared to azathioprine: a retrospective cohort study. Thai TN, Sarayani A, Wang X, Albogami Y, Rasmussen SA, Winterstein AG. Pharmacoepidemiol Drug Saf. 2020;29:716–724. doi: 10.1002/pds.5017. [DOI] [PubMed] [Google Scholar]
- 173.The safety of mycophenolate mofetil in pregnancy. Pisoni CN, D'Cruz DP. Expert Opin Drug Saf. 2008;7:219–222. doi: 10.1517/14740338.7.3.219. [DOI] [PubMed] [Google Scholar]
- 174.A case of coloboma in a newborn to a woman taking mycophenolate mofetil in pregnancy after kidney transplantation. Dei Malatesta MF, Rocca B, Gentile T, et al. Transplant Proc. 2009;41:1407–1409. doi: 10.1016/j.transproceed.2009.03.088. [DOI] [PubMed] [Google Scholar]
- 175.Congenital esophageal atresia and microtia in a newborn secondary to mycophenolate mofetil exposure during pregnancy: a case report and review of the literature. Alsebayel MM, Abaalkhail FA, Alsebayel FM, Alissa DA, Al-Jedai AH, Elsiesy H. Am J Case Rep. 2018;19:523–526. doi: 10.12659/AJCR.908433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Reviewing the evidence for mycophenolate mofetil as a new teratogen: case report and review of the literature. Anderka MT, Lin AE, Abuelo DN, Mitchell AA, Rasmussen SA. Am J Med Genet A. 2009;149A:1241–1248. doi: 10.1002/ajmg.a.32685. [DOI] [PubMed] [Google Scholar]
- 177.Teratogen update: methotrexate. Hyoun SC, Običan SG, Scialli AR. Birth Defects Res A Clin Mol Teratol. 2012;94:187–207. doi: 10.1002/bdra.23003. [DOI] [PubMed] [Google Scholar]
- 178.FDA labeling system for drugs in pregnancy. Boothby LA, Doering PL. Ann Pharmacother. 2001;35:1485–1489. doi: 10.1345/aph.1A034. [DOI] [PubMed] [Google Scholar]
- 179.Clinical pharmacokinetics of leflunomide. Rozman B. Clin Pharmacokinet. 2002;41:421–430. doi: 10.2165/00003088-200241060-00003. [DOI] [PubMed] [Google Scholar]
- 180.Birth outcomes in women who have taken leflunomide during pregnancy. Chambers CD, Johnson DL, Robinson LK, et al. Arthritis Rheum. 2010;62:1494–1503. doi: 10.1002/art.27358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Cardiovascular diseases. [ May; 2023 ]. 2023. https://www.who.int/health-topics/cardiovascular-diseases https://www.who.int/health-topics/cardiovascular-diseases
- 182.Cardiovascular disease in women: clinical perspectives. Garcia M, Mulvagh SL, Merz CN, Buring JE, Manson JE. Circ Res. 2016;118:1273–1293. doi: 10.1161/CIRCRESAHA.116.307547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Cardiovascular and autoimmune diseases in females: the role of microvasculature and dysfunctional endothelium. Gianturco L, Bodini BD, Atzeni F, et al. Atherosclerosis. 2015;241:259–263. doi: 10.1016/j.atherosclerosis.2015.03.044. [DOI] [PubMed] [Google Scholar]
- 184.The unexplained female predominance of systemic lupus erythematosus: clues from genetic and cytokine studies. Weckerle CE, Niewold TB. Clin Rev Allergy Immunol. 2011;40:42–49. doi: 10.1007/s12016-009-8192-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Utility of Framingham Coronary Heart Disease Risk Score for predicting cardiac risk after stroke. Towfighi A, Markovic D, Ovbiagele B. Stroke. 2012;43:2942–2947. doi: 10.1161/STROKEAHA.112.668319. [DOI] [PubMed] [Google Scholar]
- 186.Role of vascular cell adhesion molecule-1 and fibronectin connecting segment-1 in monocyte rolling and adhesion on early atherosclerotic lesions. Huo Y, Hafezi-Moghadam A, Ley K. Circ Res. 2000;87:153–159. doi: 10.1161/01.res.87.2.153. [DOI] [PubMed] [Google Scholar]
- 187.Chronic inflammatory diseases and endothelial dysfunction. Castellon X, Bogdanova V. Aging Dis. 2016;7:81–89. doi: 10.14336/AD.2015.0803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J. Science. 2001;294:1540–1543. doi: 10.1126/science.1064890. [DOI] [PubMed] [Google Scholar]
- 189.Immune interferon in the circulation of patients with autoimmune disease. Hooks JJ, Moutsopoulos HM, Geis SA, Stahl NI, Decker JL, Notkins AL. N Engl J Med. 1979;301:5–8. doi: 10.1056/NEJM197907053010102. [DOI] [PubMed] [Google Scholar]
- 190.Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK. Arthritis Rheum. 2005;52:1491–1503. doi: 10.1002/art.21031. [DOI] [PubMed] [Google Scholar]
- 191.Possible induction of systemic lupus erythematosus by interferon-alpha treatment in a patient with a malignant carcinoid tumour. Rönnblom LE, Alm GV, Oberg KE. J Intern Med. 1990;227:207–210. doi: 10.1111/j.1365-2796.1990.tb00144.x. [DOI] [PubMed] [Google Scholar]
- 192.Systemic lupus erythematosus arising during interferon-alpha therapy for cryoglobulinemic vasculitis associated with hepatitis C. Niewold TB, Swedler WI. Clin Rheumatol. 2005;24:178–181. doi: 10.1007/s10067-004-1024-2. [DOI] [PubMed] [Google Scholar]
- 193.Interferon alpha-induced lupus: proof of principle. Niewold TB. J Clin Rheumatol. 2008;14:131–132. doi: 10.1097/RHU.0b013e318177627d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.High serum IFN-alpha activity is a heritable risk factor for systemic lupus erythematosus. Niewold TB, Hua J, Lehman TJ, Harley JB, Crow MK. Genes Immun. 2007;8:492–502. doi: 10.1038/sj.gene.6364408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Age- and sex-related patterns of serum interferon-alpha activity in lupus families. Niewold TB, Adler JE, Glenn SB, Lehman TJ, Harley JB, Crow MK. Arthritis Rheum. 2008;58:2113–2119. doi: 10.1002/art.23619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Lifestyle interventions and weight management in systemic lupus erythematosus patients: a systematic literature review and metanalysis. Goessler KF, Gualano B, Nonino CB, Bonfá E, Nicoletti CF. J Lifestyle Med. 2022;12:37–46. doi: 10.15280/jlm.2022.12.1.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.The impact of obesity and a high-fat diet on clinical and immunological features in systemic lupus erythematosus. Kono M, Nagafuchi Y, Shoda H, Fujio K. Nutrients. 2021;13 doi: 10.3390/nu13020504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.The impact of obesity on SLE disease activity: findings from the Southern California Lupus Registry (SCOLR) Teh P, Zakhary B, Sandhu VK. Clin Rheumatol. 2019;38:597–600. doi: 10.1007/s10067-018-4336-3. [DOI] [PubMed] [Google Scholar]
- 199.Diabetes, hypertension, and cardiovascular disease: clinical insights and vascular mechanisms. Petrie JR, Guzik TJ, Touyz RM. Can J Cardiol. 2018;34:575–584. doi: 10.1016/j.cjca.2017.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Risk:benefit ratio of nonsteroidal anti-inflammatory drugs in systemic lupus erythematosus. Horizon AA, Wallace DJ. Expert Opin Drug Saf. 2004;3:273–278. doi: 10.1517/14740338.3.4.273. [DOI] [PubMed] [Google Scholar]
- 201.Long-term frequent use of non-steroidal anti-inflammatory drugs might protect patients with ankylosing spondylitis from cardiovascular diseases: a nationwide case-control study. Tsai WC, Ou TT, Yen JH, Wu CC, Tung YC. PLoS One. 2015;10:0. doi: 10.1371/journal.pone.0126347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lipid metabolism: immune regulation and therapeutic prospectives in systemic lupus erythematosus. [ May; 2023 ];Sun W, Li P, Cai J, Ma J, Zhang X, Song Y, Liu Y. Front Immunol. 2022 13:860586. doi: 10.3389/fimmu.2022.860586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Response to: 'statins in systemic lupus erythematosus' by Abud-Mendoza. de Jong HJ, van Staa TP, Cohen Tervaert JW. Ann Rheum Dis. 2019;78:0. doi: 10.1136/annrheumdis-2017-212902. [DOI] [PubMed] [Google Scholar]
- 204.Anti-inflammatory and immunosuppressive drugs and reproduction. Østensen M, Khamashta M, Lockshin M, et al. Arthritis Res Ther. 2006;8:209. doi: 10.1186/ar1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.High sensitivity C-reactive protein in systemic lupus erythematosus: relation to disease activity, clinical presentation and implications for cardiovascular risk. Barnes EV, Narain S, Naranjo A, et al. Lupus. 2005;14:576–582. doi: 10.1191/0961203305lu2157oa. [DOI] [PubMed] [Google Scholar]
- 206.Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. Roman MJ, Shanker BA, Davis A, et al. N Engl J Med. 2003;349:2399–2406. doi: 10.1056/NEJMoa035471. [DOI] [PubMed] [Google Scholar]
- 207.Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. Esdaile JM, Abrahamowicz M, Grodzicky T, et al. Arthritis Rheum. 2001;44:2331–2337. doi: 10.1002/1529-0131(200110)44:10<2331::aid-art395>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 208.Natural history of hypercholesterolemia in systemic lupus erythematosus. Bruce IN, Urowitz MB, Gladman DD, Hallett DC. https://pubmed.ncbi.nlm.nih.gov/10529129/ J Rheumatol. 1999;26:2137–2143. [PubMed] [Google Scholar]
- 209.Lipid profiles in patients with systemic lupus erythematosus. Leong KH, Koh ET, Feng PH, Boey ML. https://pubmed.ncbi.nlm.nih.gov/7966068/ J Rheumatol. 1994;21:1264–1267. [PubMed] [Google Scholar]
- 210.Weight change associated with corticosteroid therapy in adolescents with systemic lupus erythematosus. Manaboriboon B, Silverman ED, Homsanit M, Chui H, Kaufman M. Lupus. 2013;22:164–170. doi: 10.1177/0961203312469260. [DOI] [PubMed] [Google Scholar]
- 211.Low-dose aspirin as primary prophylaxis for cardiovascular events in systemic lupus erythematosus: a long-term retrospective cohort study. Iudici M, Fasano S, Gabriele Falcone L, Pantano I, La Montagna G, Migliaresi S, Valentini G. Rheumatology (Oxford) 2016;55:1623–1630. doi: 10.1093/rheumatology/kew231. [DOI] [PubMed] [Google Scholar]
- 212.Hydroxychloroquine in systemic lupus erythematosus (SLE) Ponticelli C, Moroni G. Expert Opin Drug Saf. 2017;16:411–419. doi: 10.1080/14740338.2017.1269168. [DOI] [PubMed] [Google Scholar]
- 213.Systemic lupus erythematosus (SLE) [ May; 2023 ]. 2022. https://emedicine.medscape.com/article/332244-overview https://emedicine.medscape.com/article/332244-overview
- 214.Systemic lupus erythematosus and myasthenia gravis: a rare association. Nagarajan M, Maasila AT, Dhanapriya J, et al. Indian J Nephrol. 2019;29:62–64. doi: 10.4103/ijn.IJN_12_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Myasthenia gravis. [ May; 2023 ]. 2023. https://emedicine.medscape.com/article/1171206-overview https://emedicine.medscape.com/article/1171206-overview
- 216.Systemic lupus erythematosus and secondary antiphospholipid syndrome after thymectomy for myasthenia gravis - a case report. Miskovic R, Plavsic A, Peric-Popadic A, Raskovic S, Bogic M. Open Access Maced J Med Sci. 2015;3:439–442. doi: 10.3889/oamjms.2015.096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Systemic lupus erythematosus-myasthenia gravis overlap syndrome: presentation and treatment depend on prior thymectomy. Minchenberg SB, Chaparala G, Oaks Z, Banki K, Perl A. Clin Immunol. 2018;194:100–104. doi: 10.1016/j.clim.2018.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Development of myasthenia gravis in systemic lupus erythematosus. Kigawa N, Pineau C, Clarke AE, et al. Eur J Case Rep Intern Med. 2014;1 [Google Scholar]
- 219.Myasthenia gravis associated with lupus nephritis. Lee H, Chen W, Su K, Chou C, Tsai C. Lupus. 2008;17:860–861. doi: 10.1177/0961203308093463. [DOI] [PubMed] [Google Scholar]
- 220.The chemokine CXCL13 is a key molecule in autoimmune myasthenia gravis. Meraouna A, Cizeron-Clairac G, Panse RL, Bismuth J, Truffault F, Tallaksen C, Berrih-Aknin S. Blood. 2006;108:432–440. doi: 10.1182/blood-2005-06-2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.The role of chemokines in inflammation. Proost P, Wuyts A, van Damme J. Int J Clin Lab Res. 1996;26:211–223. doi: 10.1007/BF02602952. [DOI] [PubMed] [Google Scholar]
- 222.Roles of GM-CSF in the pathogenesis of autoimmune diseases: an update. [ May; 2023 ];Lotfi N, Thome R, Rezaei N, Zhang GX, Rezaei A, Rostami A, Esmaeil N. Front Immunol. 2019 10:1265. doi: 10.3389/fimmu.2019.01265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Systemic lupus erythematosus and myasthenia gravis. Hrycek A. https://pubmed.ncbi.nlm.nih.gov/19776704/ Pol Arch Med Wewn. 2009;119:582–585. [PubMed] [Google Scholar]
- 224.The association of systemic lupus erythematosus and myasthenia gravis: a series of 17 cases, with a special focus on hydroxychloroquine use and a review of the literature. Jallouli M, Saadoun D, Eymard B, et al. J Neurol. 2012;259:1290–1297. doi: 10.1007/s00415-011-6335-z. [DOI] [PubMed] [Google Scholar]
- 225.Appearance of systemic lupus erythematosus in patients with myasthenia gravis following thymectomy: two case reports. Park MJ, Kim YA, Lee SS, Kim BC, Kim MK, Cho KH. J Korean Med Sci. 2004;19:134–136. doi: 10.3346/jkms.2004.19.1.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Myasthenia gravis and associated diseases. Tanovska N, Novotni G, Sazdova-Burneska S, et al. Open Access Maced J Med Sci. 2018;6:472–478. doi: 10.3889/oamjms.2018.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.The course of myasthenia gravis with systemic lupus erythematosus. Bekircan-Kurt CE, Tuncer Kurne A, Erdem-Ozdamar S, Kalyoncu U, Karabudak R, Tan E. Eur Neurol. 2014;72:326–329. doi: 10.1159/000365568. [DOI] [PubMed] [Google Scholar]
- 228.High prevalence of systemic lupus erythematosus in 78 myasthenia gravis patients: a clinical and serologic study. Sthoeger Z, Neiman A, Elbirt D, et al. Am J Med Sci. 2006;331:4–9. doi: 10.1097/00000441-200601000-00004. [DOI] [PubMed] [Google Scholar]
- 229.Telitacicept: first approval. Dhillon S. Drugs. 2021;81:1671–1675. doi: 10.1007/s40265-021-01591-1. [DOI] [PubMed] [Google Scholar]
- 230.Telitacicept, a novel humanized, recombinant TACI-Fc fusion protein, for the treatment of systemic lupus erythematosus. Fan Y, Gao D, Zhang Z. Drugs Today (Barc) 2022;58:23–32. doi: 10.1358/dot.2022.58.1.3352743. [DOI] [PubMed] [Google Scholar]
- 231.International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS) Miyakis S, Lockshin MD, Atsumi T, et al. J Thromb Haemost. 2006;4:295–306. doi: 10.1111/j.1538-7836.2006.01753.x. [DOI] [PubMed] [Google Scholar]
- 232.Antiphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients. Cervera R, Piette JC, Font J, et al. Arthritis Rheum. 2002;46:1019–1027. doi: 10.1002/art.10187. [DOI] [PubMed] [Google Scholar]
- 233.Primary antiphospholipid syndrome: functional outcome after 10 years. Erkan D, Yazici Y, Sobel R, Lockshin MD. https://pubmed.ncbi.nlm.nih.gov/11128669/ J Rheumatol. 2000;27:2817–2821. [PubMed] [Google Scholar]
- 234.Epidemiology of the antiphospholipid antibody syndrome. Petri M. J Autoimmun. 2000;15:145–151. doi: 10.1006/jaut.2000.0409. [DOI] [PubMed] [Google Scholar]
- 235.Detection of coronary artery disease and the role of traditional risk factors in the Hopkins Lupus Cohort. Petri M. Lupus. 2000;9:170–175. doi: 10.1191/096120300678828226. [DOI] [PubMed] [Google Scholar]
- 236.The lupus anticoagulant is a risk factor for myocardial infarction (but not atherosclerosis): Hopkins Lupus Cohort. Petri M. Thromb Res. 2004;114:593–595. doi: 10.1016/j.thromres.2004.07.012. [DOI] [PubMed] [Google Scholar]
- 237.Antiphospholipid antibodies: anticardiolipin and the lupus anticoagulant in systemic lupus erythematosus (SLE) and in non-SLE disorders. Prevalence and clinical significance. Love PE, Santoro SA. Ann Intern Med. 1990;112:682–698. doi: 10.7326/0003-4819-112-9-682. [DOI] [PubMed] [Google Scholar]
- 238.Risk for venous thrombosis related to antiphospholipid antibodies in systemic lupus erythematosus--a meta-analysis. Wahl DG, Guillemin F, de Maistre E, Perret C, Lecompte T, Thibaut G. Lupus. 1997;6:467–473. doi: 10.1177/096120339700600510. [DOI] [PubMed] [Google Scholar]
- 239.Pregnancy outcome in 396 pregnancies in patients with SLE in Saudi Arabia. Al Arfaj AS, Khalil N. Lupus. 2010;19:1665–1673. doi: 10.1177/0961203310378669. [DOI] [PubMed] [Google Scholar]
- 240.Pregnancy outcome in systemic lupus erythematosus complicated by anti-phospholipid antibodies. Mecacci F, Bianchi B, Pieralli A, et al. Rheumatology (Oxford) 2009;48:246–249. doi: 10.1093/rheumatology/ken458. [DOI] [PubMed] [Google Scholar]
- 241.A systematic review and meta-analysis of pregnancy outcomes in patients with systemic lupus erythematosus and lupus nephritis. Smyth A, Oliveira GH, Lahr BD, Bailey KR, Norby SM, Garovic VD. Clin J Am Soc Nephrol. 2010;5:2060–2068. doi: 10.2215/CJN.00240110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Increased risk for heart valve disease associated with antiphospholipid antibodies in patients with systemic lupus erythematosus: meta-analysis of echocardiographic studies. Zuily S, Regnault V, Selton-Suty C, et al. Circulation. 2011;124:215–224. doi: 10.1161/CIRCULATIONAHA.111.028522. [DOI] [PubMed] [Google Scholar]
- 243.Pulmonary hypertension in antiphospholipid syndrome. Zuily S, Wahl D. Curr Rheumatol Rep. 2015;17:478. doi: 10.1007/s11926-014-0478-8. [DOI] [PubMed] [Google Scholar]
- 244.Increased risk of livedo reticularis associated with antiphospholipid antibodies in patients with systemic lupus erythematosus: a systematic review and meta-analysis (Abstract) DeFlippis E, Wahl D, Zuily S. https://acrabstracts.org/abstract/increased-risk-of-livedo-reticularis-associated-with-antiphospholipid-antibodies-in-patients-with-systemic-lupus-erythematosus-a-systematic-review-and-meta-analysis/ Arthritis Rheum. 2015;67:2182. doi: 10.1177/09612033221126852. [DOI] [PubMed] [Google Scholar]
- 245.The clinical significance of antiphospholipid antibodies in systemic lupus erythematosus. Ünlü O, Zuily S, Erkan D. Eur J Rheumatol. 2016;3:75–84. doi: 10.5152/eurjrheum.2015.0085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.The role of antiphospholipid autoantibodies in the cognitive deficits of patients with systemic lupus erythematosus. Coín MA, Vilar-López R, Peralta-Ramírez I, Hidalgo-Ruzzante N, Callejas-Rubio JL, Ortego-Centeno N, Pérez-García M. Lupus. 2015;24:875–879. doi: 10.1177/0961203315572717. [DOI] [PubMed] [Google Scholar]
- 247.Analysis of risk factors for the development of thrombotic complications in antiphospholipid antibody positive lupus patients. Tarr T, Lakos G, Bhattoa HP, Shoenfeld Y, Szegedi G, Kiss E. Lupus. 2007;16:39–45. doi: 10.1177/0961203306074767. [DOI] [PubMed] [Google Scholar]
- 248.Risk factors for thrombosis and primary thrombosis prevention in patients with systemic lupus erythematosus with or without antiphospholipid antibodies. Tektonidou MG, Laskari K, Panagiotakos DB, Moutsopoulos HM. Arthritis Rheum. 2009;61:29–36. doi: 10.1002/art.24232. [DOI] [PubMed] [Google Scholar]
- 249.Influence of aspirin on the clinical outcomes of 103 anti-phospholipid antibodies-positive patients. Hereng T, Lambert M, Hachulla E, et al. Lupus. 2008;17:11–15. doi: 10.1177/0961203307084724. [DOI] [PubMed] [Google Scholar]
- 250.[Two cases of systemic lupus erythematosus accompanied by antiphospholipid syndrome nephropathy without immune complex nephritis] Komiya T, Okamura M, Kawakami K, et al. https://pubmed.ncbi.nlm.nih.gov/12607972/ Nihon Jinzo Gakkai Shi. 2002;44:817–822. [PubMed] [Google Scholar]
- 251.Renal involvement in primary antiphospholipid syndrome and its response to immunosuppressive therapy. Korkmaz C, Kabukcuoğlu S, Isiksoy S, Yalçin AU. Lupus. 2003;12:760–765. doi: 10.1191/0961203303lu461oa. [DOI] [PubMed] [Google Scholar]
- 252.Massive proteinuria as a main manifestation of primary antiphospholipid syndrome. Levy Y, George J, Ziporen L, et al. Pathobiology. 1998;66:49–52. doi: 10.1159/000027995. [DOI] [PubMed] [Google Scholar]
- 253.[Low-molecular heparins in the treatment of APS-nephropathy in primary and secondary antiphospholipid syndrome] Kozlovskaia NL, Shakhnova EA, Kushnir VV, Shilov EM. https://pubmed.ncbi.nlm.nih.gov/15532374/ Ter Arkh. 2004;76:35–40. [PubMed] [Google Scholar]
- 254.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. Bertsias GK, Tektonidou M, Amoura Z, et al. Ann Rheum Dis. 2012;71:1771–1782. doi: 10.1136/annrheumdis-2012-201940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Inhibition of the mTORC pathway in the antiphospholipid syndrome. Canaud G, Bienaimé F, Tabarin F, et al. N Engl J Med. 2014;371:303–312. doi: 10.1056/NEJMoa1312890. [DOI] [PubMed] [Google Scholar]
- 256.Effectiveness of nonpharmacologic interventions for decreasing fatigue in adults with systemic lupus erythematosus: a systematic review. del Pino-Sedeño T, Trujillo-Martín MM, Ruiz-Irastorza G, Cuellar-Pompa L, de Pascual-Medina AM, Serrano-Aguilar P. Arthritis Care Res (Hoboken) 2016;68:141–148. doi: 10.1002/acr.22675. [DOI] [PubMed] [Google Scholar]
- 257.Towards a practical management of fatigue in systemic lupus erythematosus. Mertz P, Schlencker A, Schneider M, Gavand PE, Martin T, Arnaud L. Lupus Sci Med. 2020;7 doi: 10.1136/lupus-2020-000441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Balneotherapy, a complementary non-pharmacological approach for non-inflammatory complaints in systemic lupus erythematosus: a pilot study. Fritsch K, Nagy G, Szekanecz Z, Szűcs G, Kovacs L, Bender T. In Vivo. 2022;36:3010–3017. doi: 10.21873/invivo.13046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Effect of training on health outcome including fatigue, depression and quality of life in patients with systemic lupus erythromatosus. Youssef MK. Beni-Suef Univ J Basic Appl Sci. 2021;10:90. [Google Scholar]
- 260.Impact of psycho-educational therapy on disease activity, quality of life, psychological status, treatment satisfaction and adherence in systemic lupus erythematosus patients. Sakr BR, Seif EM, Kamel RM, Eleishi HH. Egypt Rheumatol. 2022;44:313–317. [Google Scholar]