Startseite 2025 Chinese guidelines for the diagnosis and treatment of systemic lupus erythematosus
Artikel Open Access

2025 Chinese guidelines for the diagnosis and treatment of systemic lupus erythematosus

  • Jiuliang Zhao , Shangzhu Zhang , Qian Wang , Xinping Tian , Yaolong Chen , Mengtao Li EMAIL logo und Xiaofeng Zeng EMAIL logo
Veröffentlicht/Copyright: 4. Oktober 2025
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Systemic lupus erythematosus (SLE) is a complex autoimmune disease that poses significant challenges in diagnosis and treatment. In recent years, advances in basic and clinical research related to SLE have led to the emergence of new diagnostic and therapeutic approaches, as well as the continuous updates of international clinical guidelines. Consequently, existing guidelines no longer fully align with current clinical practice in China. Initiated by the National Clinical Research Center for Dermatologic and Immunologic Diseases and the Chinese SLE Treatment and Research Group, and in collaboration with the Chinese Rheumatology Association and Chinese Association of Rheumatology and Immunology Physicians, an expert panel has comprehensively evaluated the latest research evidence and integrated domestic clinical experience. In accordance with the GRADE framework, the “2020 Chinese Guidelines for the Diagnosis and Treatment of Systemic Lupus Erythematosus” have undergone systematic revision. The updated guidelines provide detailed evidence-based recommendations addressing 12 critical clinical concerns prioritized by frontline rheumatologists in China. The revision aims to optimize the scientific rigor of SLE clinical management and enhance patient-centered healthcare services.

Background

Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by multi-organ involvement, relapsing-remitting course, and the presence of autoantibodies. Without timely treatment, SLE can cause irreversible end-organ damage and fatal outcomes. The pathogenesis of SLE is multifactorial, with contributions from genetic susceptibility, sex-hormonal influences, and environmental exposures (such as viral and bacterial infections).[1, 2, 3] SLE prevalence varies significantly across regions, with global incidence ranging from 1.4 to 11 per 100, 000 individuals. Based on analyses of China’s national health insurance database and the Chinese Rheumatism Data Center (CRDC), the estimated incidence in mainland China is approximately 14.09 per 100, 000,[4,5] with a female-to-male ratio of 10–12: 1.[6, 7, 8] The development of SLE therapeutics has driven substantial improvements in survival outcomes. Epidemiological data reveal a progressive increase in 5-year survival rates from 50%-60% (1950 s) to over 90% (1990 s), with stabilization observed in the 2008–2016 period (95% in high-income vs. 92% in low-and middle-income countries).[9, 10, 11] Consequently, SLE has transitioned from an acute, highly fatal disease to a chronic, manageable condition.

Leading international organizations, including the American College of Rheumatology (ACR), the European League Against Rheumatism (EULAR), the British Society for Rheumatology (BSR), and the Asia Pacific League of Associations for Rheumatology (APLAR), have developed their respective guidelines for the diagnosis and treatment of SLE,[12, 13, 14] Similarly, the Chinese Rheumatology Association released two editions of SLE guidelines in 2010 and 2020, [15,16] which have significantly enhanced the scientific rigor and standardization of clinical decision-making. However, existing guidelines face limitations in guiding current SLE clinical practice in China due to the following reasons: (1) Data from the Chinese SLE Treatment and Research Group (CSTAR) registry cohort demonstrates that the disease onset, clinical manifestations, and prognosis of SLE patients in China differ from those in Western populations.[6,17,18] (2) International SLE management recommendations may not align with China’s clinical realities. (3) With continuous advancements in both SLE therapeutics (including novel agents) and evidence-based guideline methodology, the Chinese 2020 SLE guidelines no longer adequately address current clinical needs. To address these gaps, the National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID) (Peking Union Medical College Hospital), in collaboration with the CSTAR, the Chinese Rheumatology Association, the Chinese Association of Rheumatology and Immunology Physicians, the Rheumatology Rehabilitation Branch of Chinese Association of Rehabilitation Medicine, and the Chinese Research Hospital Association of Rheumatology and Immunology Professional Committee, has developed the “2025 Chinese Guidelines for the Diagnosis and Treatment of Systemic Lupus Erythematosus (revised edition) “(hereafter referred to as this guideline). This guideline development process adhered to internationally recognized evidence-based medicine standards, while comprehensively considering China’s unique clinical practice and healthcare system characteristics.

Guideline Development Methods

Guideline Initiators and Expert Panel

This guideline was initiated by the NCRC-DID (Peking Union Medical College Hospital) and the CSTAR, in collaboration with the Chinese Rheumatology Association, the Chinese Association of Rheumatology and Immunology Physicians, the Rheumatology Rehabilitation Branch of Chinese Association of Rehabilitation Medicine, and the Chinese Research Hospital Association of Rheumatology and Immunology Professional Committee. The development of this guideline was launched on August 21st, 2024, and was finalized on December 21st, 2024.

Guideline Working Group

A multidisciplinary working group was established, comprising 75 experts in rheumatology, nephrology, dermatology, obstetrics, radiology, and evidence-based medicine. Methodological support was provided by the Evidence-Based Medicine Center of Lanzhou University, the Lanzhou University GRADE (The Grading of Recommendations Assessment, Development and Evaluation) Center, and the Evidence-Based Evaluation and Guideline Innovation Unit of the Chinese Academy of Medical Sciences. All working group members completed conflict-of-interest disclosure forms, confirming no direct interest conflicts related to this guideline.

Guideline Registration and Protocol Development

The guideline was registered at the International Practice Guidelines Registry Platform (http://www.guidelines-registry.org; registration number: PREPARE-2024CN1085). Its development adhered to the World Health Organization (WHO) Handbook for Guideline Development (2014),[19] the Basic Methods and Procedures for Developing/Revising Clinical Practice Guidelines issued by the Chinese Medical Association (2016),[20] and incorporated the Appraisal of Guidelines for Research and Evaluation II (AGREE II) instrument[21] and the Reporting Items for Practice Guidelines in Healthcare (RIGHT) standards.[22,23]

Guideline Users and Target Population

This guideline is intended for rheumatologists, nephrologists, dermatologists, obstetricians, clinical pharmacists, radiologists, and other healthcare professionals involved in SLE diagnosis and management. The recommendations apply to patients with confirmed SLE.

Selection and Prioritization of Clinical Questions

Through systematic reviews of published SLE guidelines and systematic reviews, supplemented by interviews with rheumatology experts, the working group initially identified 30 clinical questions. An online survey assessed their importance, with two rounds of feedback from 83 participants. Twelve key clinical questions were ultimately selected for inclusion.

Evidence Retrieval

For each clinical question and outcome, the PICO (Population, Intervention, Comparison, Outcome) framework guided literature searches in: (1) MEDLINE, Cochrane Library, Web of Science, Embase, CBM (China Biology Medicine), Wanfang, and CNKI (China National Knowledge Infrastructure, from the establishment of the database to October 2024), prioritizing systematic reviews, meta-analyses, and network meta-analyses, followed by randomized controlled trials (RCTs), cohort studies, case-control studies, case series, and epidemiological surveys; (2) Official websites of the National Institute for Health and Care Excellence (NICE), the National Guideline Clearinghouse (NGC), the Scottish Intercollegiate Guidelines Network (SIGN), ACR, EULAR, and APLAR for SLE-related studies and guidelines; (3) Supplementary searches in Epistemonikos, UpToDate, DynaMed, and Google Scholar.

Evidence Evaluation and Grading

The evidence team assessed bias risk using: A Measurement Tool to Assess Systematic Reviews (AMSTAR) for systematic reviews, meta-analysis, and network meta-analysis;[24] Cochrane Risk of Bias (ROB) tool for RCTs;[25] Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) for diagnostic studies;[26] Newcastle-Ottawa Scale (NOS) for observational studies.[27] Two independent reviewers conducted evaluations, with discrepancies resolved through discussion or third-party consultation. Evidence quality and recommendation strength were graded using the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) approach (Table 1).[28, 29, 30, 31, 32]

Table 1

Grading of Recommendations Assessment, Development, and Evaluation

GRADE Rating Description
Quality of Evidence
High (A) High confidence: The estimated effect lies close to the true value
Medium (B) Moderate confidence: The estimated effect is likely close to the true value, but may be substantially different
Low (C) Low confidence: The estimated effect may be substantially different from the true value
Very Low (D) Very low confidence: The estimated effect is likely substantially different from the true value
Strength of Recommendation
Strong (1) Clearly demonstrates that benefits outweigh harms or vice versa
Weak (2) Uncertain balance of benefits and harms, or evidence shows comparable benefits and harms regardless of quality

Recommendation Formulation

Based on the synthesized evidence (including domestic and international studies) provided by the evidence evaluation team, and after comprehensive consideration of Chinese patients’ preferences and values, intervention costs, and benefit-risk profiles, the expert panel drafted initial recommendations specific to China’s clinical practice. A Delphi survey was conducted on December 11th, 2024, which collected 30 feedback suggestions. Face-to-face consensus meetings held on November 22nd and December 19th, 2024, further refined the recommendations. Final consensus was achieved for all recommendations (defined as ≥75% expert agreement).

Guideline Update

The working group will continuously monitor emerging evidence and the applicability of current recommendations. The recommendations of this guideline are planned to be updated within 3–5 years in accordance with the methodologies in International Guidelines.[33]

Question 1: How to diagnose SLE?

Recommendation 1: The 2012 Systemic Lupus International Collaborating Clinics (SLICC) classification criteria or the 2019 EULAR/ACR SLE classification criteria are recommended for the diagnosis of patients with suspected SLE (1B). For patients with atypical clinical presentations or diagnostic difficulties, consultation with or referral to a rheumatology specialist is recommended (2C).

To improve the sensitivity and specificity of SLE classification criteria, the 2019 EULAR/ACR criteria were developed. These include one entry criterion and 18 criteria across 10 domains. Each criterion must exclude causes such as infection, malignancy, or drug. Criteria do not have to occur simultaneously. Within each domain, only the highest weighted criterion is counted toward the total scores. Patients who meet the entry criterion and receive scores of 10 or more are classified as SLE.[34] Validation studies showed that the 2019 EULAR/ACR, the 2012 SLICC, and the 1997 ACR criteria had sensitivities of 96%, 97%, and 83%, and specificities of 93%, 84%, and 93%, respectively, with the 2019 EULAR/ACR criteria demonstrating the optimal balance.[34] However, in adult SLE patients, the 2012 SLICC criteria showed superior sensitivity (100%) and specificity (75%) compared with the 2019 EULAR/ACR criteria (93% sensitivity, 73% specificity) and the 1997 ACR criteria (83% sensitivity, 82% specificity).[35] Thus, this guideline recommends using both the 2019 EULAR/ACR and the 2012 SLICC classification criteria for SLE diagnosis in China. To avoid missing diagnoses in persistently antinuclear (ANA)-negative individuals, the 2012 SLICC criteria should be applied concurrently.

SLE shares overlapping clinical and laboratory features with primary Sjögren’s syndrome, rheumatoid arthritis, and mixed connective tissue disease. Detailed clinical evaluation and autoantibody testing are critical for differentiation. The 2019 EULAR/ACR criteria significantly reduce misdiagnosis. A retrospective study (352 SLE patients vs. 385 disease controls) from China found both the 2012 SLICC and the 2019 EULAR/ACR criteria effective for early diagnosis, with the latter performing better.[36] A cross-sectional study from another country revealed that only 23% of SLE cases diagnosed by primary care physicians met the 1997 ACR criteria (fulfilled 4 or more criteria), compared to 79% of cases diagnosed by rheumatologists.[37] This highlights the importance of the involvement of rheumatology specialists in SLE diagnosis. A review published in The Lancet in 2024 emphasized the challenges primary care physicians face when diagnosing SLE and recommended early referral to rheumatologists.[38]

Question 2: What are the treatment principles and goals for SLE?

Recommendation 2: The principles of SLE treatment emphasize early intervention and a Treat-to-Target (T2T) strategy (1C). Short-term goals include controlling disease activity and achieving clinical remission or at least low disease activity (1C). Long-term goals include maintaining remission, preventing flares, minimizing drug toxicity, reducing organ damage, improving quality of life, and lowering mortality (1C).

The average delay in diagnosing SLE in China is 10.8 months, [39] which is associated with increased disease activity and organ damage.[40] A meta-analysis confirmed that active disease worsens quality of life [41] and increases mortality,[42] underscoring the need for early intervention.

The Treat-to-Target (T2T) strategy involves adjusting therapy timely to achieve defined, feasible, and clinically meaningful targets. The 2021 DORIS (Definitions Of Remission In SLE) defines clinical remission [43] (Table 2). If remission is unattainable, the Asia-Pacific Lupus Collaboration (APLC)-proposed Lupus Low Disease Activity State (LLDAS) serves as an alternative.[44]

Table 2

Definitions of SLE Clinical Remission and Low Disease Activity

Treatment Target Clinical Remission[43] Low Disease Activity[44]
Definition • Clinical SLEDAI (cSLEDAI) = 0 • SLEDAI-2K ≤4
• Physician Global Assessment (PGA) ≤0.5 • PGA ≤1
• Permitted therapies: antimalarials (e.g., hydroxychloroquine), lowdose glucocorticoids (prednisone ≤5 mg/d or equivalents), and/or stable-dose immunosuppressants/biologics • Permitted therapies: antimalarials, low-dose glucocorticoids (prednisone ≤7.5 mg/d or equivalents), and/or stable-dose immunosuppressants/biologics

A systematic review of 41 studies (17, 270 patients) found that 42.4%-88% of patients achieved remission within one year, and 21.1%-70% sustained remission for five years or longer. [45] Another systemic review indicated that remission (OR = 0.49–0.75) and LLDAS (OR = 0.19–0.88) significantly reduce the SLICC/ACR Damage Index (SDI), correlating with lower risks of organ damage, flares, mortality, and other adverse outcomes (such as death, severe infections, and hospitalization risks) compared to uncontrolled disease activity. [46] Even after achieving remission/LLDAS, close monitoring is critical for preventing disease flare and optimizing treatment. Maintaining these states for more than 3–6 months reduces the risks of flares and organ damage, with extended maintenance further improving outcomes. [47,48]

Question 3: How to assess SLE disease activity and organ damage?

Recommendation 3: It is recommended to use SLE disease activity index (SLEDAI2000) score combined with the clinician’s comprehensive assessment to evaluate SLE disease activity (2C). Assessment of disease activity is recommended at least once a month for patients with active SLE and once every 3 to 6 months for patients with stable SLE (2D). Disease flares should be treated as active SLE (2D).

Multiple tools are available for SLE activity assessment,[49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59] all requiring integration of past medical history, physical examination, and laboratory tests. The physician’s personal preference and expertise, the cost of evaluation (whether a computer is needed and the expenses of examination), and the time required all influence the choice of evaluation tools.[60] SLEDAI-2000 scores range from 0 to 105, while British Isles Lupus Assessment Group (BILAG)-2004 scores are divided into 5 categories: A, B, C, D and E. Compared with BILAG-2004, SLEDAI-2000 takes less time for assessment and is easier to use, which is preferred.[61,62]

Disease activity can be graded based on SLEDAI-2000. At present, there are mainly four criteria for grading disease activity,[12,13,43,44] and the EULAR’s criteria are recommended, which define mild activity as SLEDAI-2000 ≤6, moderate activity as SLEDAI-2000 7–12, and severe activity as SLEDAI-2000 >12.[12] Because a higher SLEDAI-2000 is associated with increased risks of organ damage (HR = 1.18, 95% CI: 1.02–1.37) and death (HR= 1.14, 95% CI: 1.02–1.22),[63] regular monitoring of disease activity and organ damage is needed. Since evaluating disease activity based solely on SLEDAI-2000 and BILAG-2004 has certain limitations, it is also necessary to combine the Physician’s Global Assessment (PGA) with reference to the patient’s clinical manifestations to improve the accurance of the evaluation.

The Systemic Lupus Erythematosus Disease Activity Score (SLE-DAS) was developed by Jesus et al in 2019. The aim is to address the low sensitivity of SLEDAI-2000 in capturing changes in disease activity through a continuous scoring method, while maintaining high specificity and clinical usability.[54] SLE-DAS includes 17 clinical and laboratory indicators, such as hemolytic anemia, cardiopulmonary manifestations, and gastrointestinal involvement, which are absent in SLEDAI-2000. SLE-DAS is calculated using a mathematical formula, with a strong correlation with PGA and SLEDAI-2000 (r = 0.875 and r = 0.943, respectively). In validation study, SLE-DAS was significantly more sensitive to disease improvement and deterioration than SLEDAI-2000, with a greater ability to predict damage accumulation. The calculation of SLE-DAS is relatively complex and relies on formula calculations. According to the score, the disease activity can be divided into mild activity (SLE-DAS ≤2.08), moderate activity (2.08<SLE-DAS≤7.64), and severe activity (SLE-DAS >7.64).[64]

The Systemic Lupus International Collaborating Clinics (SLICC) Damage Index (SDI) is the only internationally recognized and validated assessment standard for SLE organ damage, which scores 12 organ systems independently and is an effective tool for evaluating organ damage in clinical practice. SDI provides a basis for better predicting the prognosis of SLE patients.[65]

There is a lack of evidence regarding the frequency of monitoring disease activity in SLE patients. The UK SLE guidelines recommend evaluating disease activity at least once every 1–3 months in patients with active disease, while the Spanish SLE guidelines recommend evaluations at least once every 3–4 months in the first year. For SLE patients with stable or low disease activity, both the UK and Spanish SLE guidelines recommend an evaluation every 6–12 months.[13,66] Based on the above international guideline recommendations, this guideline recommends assessing disease activity at least once a month for patients with active SLE (83.33% consensus), and once every 3–6 months for patients with stable SLE (94.44% consensus).

In addition, the frequency of clinical monitoring should be adjusted according to the progression of the disease and the intensity of treatment. SLE flares should be treated as active disease, and the disease activity evaluation should be performed at least once a month until SLE is stable.

Question 4: How to use glucocorticoids in the treatment of SLE?

Recommendation 4: Glucocorticoid is the foundational medication for the treatment of SLE (1A). An individualized glucocorticoid regimen should be formulated based on disease activity, the involved organs, and its severity, at the lowest effective dose (1B). For mild SLE, low-dose glucocorticoids (≤10 mg/d prednisone or equivalent) should be considered if hydroxychloroquine or nonsteroidal anti-inflammatory drugs (NSAIDs) are ineffective. For moderate SLE, glucocorticoids (0.5–1 mg/kg/d prednisone or equivalent) combined with immunosuppressants are recommended (2C). For severe SLE, high-dose glucocorticoids (≥1 mg/kg/d prednisone or equivalent) combined with immunosuppressants should be used (2C). For life-threatening organ involvement, pulse glucocorticoid therapy combined with immunosuppressants is recommended (1B). Clinicians should closely monitor disease activity and adjust glucocorticoid doses accordingly. For patients with long-term stable disease, the maintenance dose should be ≤5 mg/d prednisone or equivalent, with an attempt to discontinue if possible (1B).

Glucocorticoids play a crucial role in SLE treatment and are the most commonly used agents for induction therapy, widely recommended in both domestic and international guidelines.[12,13,67, 68, 69] Glucocorticoid regimens should be individualized based on disease activity, organ involvement, and severity, with dose adjustments guided by treatment response, duration, and adverse effects.

For mild SLE, low-dose glucocorticoids (≤10 mg/d prednisone or equivalent) may be considered if hydroxychloroquine or NSAIDs fail to control disease activity.[13,16,66,68] For patients with moderately active SLE, medium-dose glucocorticoids (0.5–1 mg/kg/d prednisone or equivalent) are recommended. If disease activity is not rapidly controlled with medium-dose glucocorticoids, combination therapy with immunosuppressants is advised to reduce cumulative steroid exposure and minimize the risk of long-term adverse effects.[13,16] For severe SLE, high-dose glucocorticoids (1 mg/kg/d prednisone or equivalent) combined with immunosuppressants are recommended, with dose adjustments after stabilization. Pulse glucocorticoid therapy may be used if necessary.[12,66,68] For life-threatening organ involvement, pulse glucocorticoid therapy (intravenous methylprednisolone 250–1000 mg/d for 3 days, repeatable at 5–30-day intervals if needed) combined with immunosuppressants is recommended. Following pulse therapy, oral prednisone (0.5–1 mg/kg/d or equivalent) should be given for 4–8 weeks, with dose modified according to patient response. Compared with high-dose oral glucocorticoids, pulse therapy achieves faster disease control, allows quicker tapering, and does not significantly increase the risk of adverse effects.

Dosage adjustment and tapering of glucocorticoids should be based on disease activity and the occurrence of steroid-related adverse effects. The tapering process must be gradual and carefully monitored to avoid abrupt withdrawal. For stable patients, early dose reduction is recommended, with the goal of maintaining ≤5 mg/d prednisone or equivalent, and discontinuation if feasible. Real-world studies show that patients maintained on <5 mg/d prednisone or equivalent have less damage accumulation, while those never tapered below 5 mg/d face higher risk of steroid-related adverse effects.[69] A global multicenter prospective observational cohort study demonstrated that in SLE patients meeting the criteria for modified serologic activity (elevated anti-dsDNA and/or hypocomplementemia) with clinically quiescent disease (SACQ), glucocorticoid tapering did not increase the risk of disease flares and exhibited a protective effect against damage accumulation.[70] A meta-analysis indicated that glucocorticoid discontinuation may slightly increases the risk of flares (mainly mild flares) but reduces the incidence of SDI progression.[71]

Glucocorticoid-related adverse effects occur in >30% of patients. The most common short-term adverse reactions are gastrointestinal disturbances, excitation, palpitations, and insomnia, while long-term adverse reactions include osteoporosis, osteonecrosis of the femoral head, secondary infection, glaucoma, and cataracts.[72] The occurrence of adverse effects from glucocorticoid therapy is positively correlated with dosage. Therefore, clinicians should aim to prescribe the minimal effective dose required for disease control, concurrently avoiding potential risks stemming from inadequate dosage or inappropriate administration.

Question 5: How to use hydroxychloroquine in the treatment of SLE?

Recommendation 5: Long-term hydroxychloroquine is recommended as the mainstay of treatment for patients with SLE (1A). For patients taking hydroxychloroquine, ocular risk assessment is recommended. For high-risk patients, an annual ophthalmologic examination is recommended. Low-risk patients are recommended to undergo an ophthalmologic examination annually from the fifth year of medication (2C).

Hydroxychloroquine (HCQ) is a cornerstone therapy for patients with SLE. Long-term use of HCQ reduces disease activity, decreases the risk of organ damage and thrombosis, and improves lipid profiles and survival.[73, 74, 75] The recommended dose is 5 mg/kg/d (real body weight), not exceeding 400 mg/d. The dosage may be individualized based on the patient’s risk of flares and retinal toxicity. A recent follow-up study of 342 SLE patients found that using HCQ at ≤5 mg/kg/d, compared to >5 mg/kg/d, significantly increased the risk of disease relapse (OR = 1.98, 95% CI: 1.03–3.79).[76] An additional study of the SLICC cohort showed that both dose reduction (HR = 1.20, 95% CI: 1.04–1.38) and discontinuation (HR = 1.56, 95% CI: 1.31–1.86) of HCQ significantly elevated the risk of flares. [77] High-risk factors for HCQ-related retinal toxicity include long-term and/or high-dose ( >5 mg/kg/d) use, hepatic or renal impairment, concurrent use of tamoxifen, a history of retinal or macular disease, and age over 45 years.[78, 79, 80] Among individuals administered HCQ at ≤5 mg/kg/d, the risk of toxicity is under 1% over 5 years and under 2% over 10 years, but it rises to almost 20% after 20 years. However, for patients who remain toxicity-free after 20 years of use, the future probability of developing toxicity is only 4%.[81] Therefore, for patients without high-risk factors, a baseline ophthalmologic examination is recommended, followed by annual screenings (such as visual field testing and spectral-domain optical coherence tomography [SD-OCT]) starting after five years of treatment to monitor for drug-induced retinal toxicity.[81,82] In contrast, patients at high risk for retinal toxicity should undergo annual ophthalmologic examinations both before and during HCQ treatment to enable early detection and intervention.

Contraindications for HCQ include hypersensitivity to HCQ or its derivatives and a history of retinopathy caused by similar drugs. Recent studies suggest that HCQ may be associated with QT interval prolongation[83, 84, 85] and cardiac conduction blocks.[86] Therefore, patients with preexisting cardiac conditions or those taking concomitant medications (such as Class I and III antiarrhythmic agents, tricyclic antidepressants, antipsychotics, and certain anti-infective drugs such as macrolides and fluoroquinolones) should be monitored for arrhythmic risk.[85] Additionally, case reports have suggested a potential association between long-term HCQ use and cardiomyopathy,[86, 87, 88, 89] though the exact causality and specific risk factors require further investigation.

Question 6: How to use immunosuppressants and biologics in the treatment of SLE?

Question 6.1: How to use immunosuppressants in the treatment of SLE?

Recommendation 6.1: Immunosuppressants are recommended for patients who do not respond well to glucocorticoids in combination with HCQ or who are unable to taper glucocorticoids to a maintenance dose (≤5 mg/d of prednisone or an equivalent dose of other glucocorticoids) (1B). Immunosuppressants are recommended at the time of initial treatment for patient with organ involvement (2B).

The use of immunosuppressants reduces the cumulative dose of glucocorticoids and prevents disease flares.[90] In patients with refractory (poor response to conventional therapy) or flared SLE, immunosuppressants can reduce glucocorticoids dosage, control disease activity and improve clinical remission rates.[91,92] For patients with severe SLE with major organ involvement, high-dose cyclophosphamide (CTX) pulse therapy should be considered.[12]

For SLE patients with active skin lesions who show inadequate response to local treatments (such as topical glucocorticoids or tacrolimus ointment) and hydroxychloroquine, oral glucocorticoids combined with immunosuppressants can be added. Methotrexate (MTX) and mycophenolate mofetil (MMF) have comparable efficacy,[93] and thalidomide and lenalidomide may be considered for patients who do not respond to other treatments.[94,95] For lupus nephritis (LN) patients undergoing initial treatment (induction phase), compared to glucocorticoid monotherapy, the combination of immunosuppressants significantly improves clinical remission rates and reduces treatment failure. Therefore, immunosuppressants or multitarget therapy should be considered from the beginning of treatment.[96, 97, 98] For SLE patients with organ involvement, the choice of immunosuppressants should be based on a comprehensive assessment of clinical manifestations, fertility issues, drug safety, and cost (Table 3).

Table 3

Indications, Advantages and Common and Significant Adverse Reactions of Different Immunosuppressants

Immunosuppressants Main Applicable Populations Advantages Common Adverse Reactions
Mycophenolate Mofetil (MMF) Moderate to severe SLE[91] In patients with moderate to severe lupus nephritis, mycophenolate mofetil is an effective treatment in both the induction and maintenance phases, and in reducing the flare rate.[109] It can also be used for the induction and maintenance treatment of mild to moderate neuropsychiatric lupus.[110] For patients with mild to moderate SLE without organ involvement, MMF can help reduce the risk of severe flares and the occurrence of lupus nephritis.[111] The most common adverse reactions are gastrointestinal discomfort. Some patients develop infections, myelosuppression and liver damage.[112] Due to the teratogenicity, pregnancy can only be attempted at least 6 weeks after discontinuation.[113]
Cyclophosphamide (CTX) Moderate to severe lupus nephritis, neuropsychiatric lupus and SLE with immune thrombocytopenia[90,97,114] It is an effective treatment in the induction and maintenance phases for patients with moderate to severe lupus nephritis, and is an effective immunosuppressive agent for the treatment of neuropsychiatric lupus and hematological disorders.[97,114] The common adverse reactions include gastrointestinal discomfort, nausea, and vomiting. Liver damage and myelosuppression are the main adverse reactions. It has well-documented reproductive toxicity and may increase the risk of malignancy.[90,115,116] Glutathione S-transferase Pi 1 (GSTP1) gene polymorphism (rs1695, 313A > G) may be useful in predicting adverse reactions.[117]
Leflunomide (LEF) Proliferative lupus nephritis[118,119] It is effective and well tolerated in some patients with proliferative lupus nephritis.[119] It can be used for the maintenance treatment of lupus nephritis.[120] LEF can cause liver damage, hypertension, leukopenia, infections, and other complications. Due to its teratogenicity, it is recommended to discontinue LEF for 24 months before attempting pregnancy. Alternatively, cholestyramine (8 g orally, three times daily for 11 days) can be used for drug elimination. If two blood drug concentration tests, taken two weeks apart, both show levels <0.02 mg/L, LEF can be considered cleared, and pregnancy may be attempted.[121]
Methotrexate (MTX) Mild to moderate SLE without renal involvement[90] It is effective in improving skin, arthritis and overall condition in patients with SLE.[90,122,123] The most common adverse reactions are gastrointestinal discomfort, such as nausea and vomiting. Hematological adverse reactions such as anemia, leukopenia and liver damage are also common.[123]
Tacrolimus (TAC) Proliferative lupus nephritis, refractory lupus nephritis and SLE with immune thrombocytopenia[109] It is an effective treatment in the induction and maintenance phases for patients with lupus nephritis. It can also reduce flares.[109,127] It can be used for the treatment of refractory lupus nephritis, especially in those with proteinuria.[124,125] Compared with other immunosuppressants or glucocorticoids, it has a low risk of causing serious infections.[128] It can be used during pregnancy.[129,130] The common adverse reactions include gastrointestinal discomfort, and some patients may experience kidney and liver damage. In patients with impaired liver function, the TAC dosage should be reduced. During treatment, renal toxicity, blood glucose, serum uric acid, and blood pressure should be monitored.[127,131]
Cyclosporine (CsA) Lupus nephritis and immune thrombocytopenia Cyclosporine in combination with other immunosuppressants may be used to treat lupus nephritis especially type V that does not respond to standard therapy.[135] It is effective in some patients with hematological involvement,[132,134] and can be used during pregnancy.[129,130] The common adverse reactions include kidney damage, liver damage, increased blood pressure, gastrointestinal discomfort, and infections.[134]
Azathioprine (AZA) Moderate SLE[113] It can be used as the maintenance treatment for SLE. It has a low risk of severe infection. It is safe during pregnancy.[113] The common adverse reactions are myelosuppression and liver damage.[113] Testing for thiopurine methyltransferase activity is necessary.
Sirolimus Moderate SLE[101] It can be added for SLE patients with inadequate response to standard therapy. It can improve arthritis, rashes, and thrombocytopenia.[100,101] It can be used as the maintenance therapy for lupus nephritis.[104,105] The common adverse reactions are upper respiratory tract infections, gastrointestinal reactions, menstrual irregularities, oral ulcers, cytopenia, rash, and lipid abnormalities.[101,103]

Sirolimus, also known as rapamycin, was first reported in 2006 as a potential treatment for active SLE patients who have inadequate responses to conventional immunosuppressive therapy. Sirolimus treatment at a dose of 2 mg/d decreases disease activity and allows glucocorticoid tapering.[99] A prospective, single-arm, open-label, single-center clinical trial in 2018 showed that 55% of patients experienced a significant reduction in disease activity (SLEDAI-2000 and BILAG-2004), and 66% had a response according to the SLE Responder Index (SRI) after one year of treatment. [100]

A prospective real-world study conducted in China included 49 SLE patients with persistent disease activity despite standard immunosuppressive therapy. Following oral sirolimus treatment, SLEDAI-2000 scores significantly decreased, prednisone dosage was reduced, and serological markers (complement levels and anti-dsDNA antibodies) improved. Specifically, among LN patients, 41.2% achieved renal remission.[101] A real-world cohort study from CSTAR demonstrated that sirolimus had comparable eficacy to tacrolimus in patients with active SLE.[102] A meta-analysis further confirmed that sirolimus treatment leads to reductions in SLEDAI-2000, BILAG-2004 scores, and prednisone dosage in patients with active SLE.[103] Additionally, small-scale non-randomized controlled studies suggested that sirolimus may be used as the maintenance treatment for LN,[104,105] refractory SLE with immune thrombocytopenia,[106,107] and SLE with concomitant antiphospholipid syndrome.[108]

Question 6.2: How to use biologics in the treatment of SLE?

Recommendation 6.2: For patients who fail to achieve treatment targets or experience flares despite receiving glucocorticoid and conventional immunosuppressant therapy, biologic agents may be considered (1B).

In recent years, an increasing number of biologics have been used to treat SLE, some of which have been approved for SLE treatment in China. Currently, biologics for SLE include agents primarily targeting B cells, such as belimumab, telitacicept, rituximab, and obinutuzumab, as well as the type I interferon receptor antagonist anifrolumab, and the C5 complement inhibitor eculizumab (Table 4).

Table 4

Indications, Advantages, and Major Adverse Reactions of Different Biologic Agents

Biologic Agent Target FDA or CFDA Approval Status Usage Recommendations
Belimumab Soluble B lymphocyte stimulator (BLyS) Yes (FDA and CFDA approval) It is the first biological agent approved for SLE treatment. It can significantly increase SRI-4 response rates, reduce disease activity and flares, and help reduce glucocorticoid and immunosuppressant dosage. It is recommended for combination therapy in patients with active SLE or LN.
Telitacicept BLyS and a proliferation-inducing ligand (APRIL) Yes (CFDA approval) It can significantly improve SRI-4 response rates, ameliorate SLE disease activity and proteinuria levels, and facilitate glucocorticoid and immunosuppressant tapering.
Rituximab CD20 No It is effective for some patients with severe refractory SLE and LN, and can improve symptoms of neuropsychiatric lupus and hematologic involvement. It is recommended for patients with poor response to initial treatment.
Obinutuzumab CD20 No As a fully humanized CD20-targeted antibody, it is effective for LN patients, LN patients unresponsive to rituximab and SLE patients without renal involvement. It can improve renal remission rates and sustain renal responses.
Anifrolumab type I interferon receptor Yes (FDA approval) It can be used to treat moderate-to-severe SLE, improving SRI-4 and BICLA response rates, enhance complete renal response in active class III/IV LN, and reducing glucocorticoid dosage and flare rates. It is recommended for patients who have difficulty tapering glucocorticoids.
Eculizumab C5 No It has been reported to be effective in some cases of refractory LN, thrombotic microangiopathy secondary to SLE, and antiphospholipid syndrome secondary to SLE

Belimumab is a human immunoglobulin G1 monoclonal antibody that inhibits the binding of soluble B lymphocyte stimulator (BLyS) to B cells. By blocking the binding of soluble BLyS to its receptors on B cells, it inhibits B-cell proliferation and is the first biologic approved globally for SLE treatment. The phase III trials BLISS-52, BLISS-76, and a study primarily involving Chinese patients in Northeast Asia, demonstrated that belimumab significantly increased the SLE Responder Index 4 (SRI-4) response rate, reduced SLE disease activity and risk of severe flares, and exhibited a favorable overall safety profile.[136, 137, 138] Another international multicenter phase III trial (BLISS-LN) showed that adding belimumab in conjunction with standard care improved renal response rates and significantly reduced the probability of kidney-related events or death [139] in patients with active LN. A post hoc analysis of BLISS-LN revealed that belimumab in conjunction with standard therapy can reduce the risks of renal flare and decrease in estimated glomerular filtration rate (eGFR) in LN patients.[140] Real-world studies have also confirmed that belimumab improved various SLE clinical manifestations, including renal, cutaneous, and joint involvement, and was associated with glucocorticoid dose reductions.[141, 142, 143] The 2023 EULAR guidelines recommend belimumab for SLE patients who do not respond to hydroxychloroquine (either alone or combined with glucocorticoids), or who are unable to reduce glucocorticoids to maintenance doses, or who have active proliferative LN.[12] The 2024 KDIGO guidelines recommend that patients with repeated kidney flares or at high-risk for progression to kidney failure due to severe chronic kidney disease should receive a triple immunosuppressive regimen of glucocorticoids combined with mycophenolate mofetil and belimumab, or glucocorticoids combined with reduced-dose cyclophosphamide and belimumab, which may also be continued as maintenance therapy.[144] Additionally, studies have shown that belimumab is effective in treating active cutaneous manifestations.[12]

Telitacicept is a dual-target biologic against BLyS and a proliferation-inducing ligand (APRIL) that was independently developed and manufactured in China. Telitacicept received approval from the National Medical Products Administration in March 2021 for the treatment of SLE. Phase IIb trials showed telitacicept in conjunction with standard therapy significantly improved SRI-4 response, reduced disease activity and glucocorticoids dose.[145] Phase III data demonstrated a SRI-4 response of 82.6% at 52 weeks.[146] Real-world studies reported a SRI-4 response of 80% by week 45, with LN patients showing significantly reduced proteinuria and glucocorticoid dosage reduction.[147] Retrospective analyses confirmed that telitacicept decreased disease activity, increased LLDAS achievement, reduced glucocorticoid dosage, and improved both renal and hematologic parameters.[148,149]

Rituximab, an anti-CD20 monoclonal antibody, showed efficacy in refractory SLE despite failing to meet its primary efficacy endpoint in EXPLORER and LUNAR trials.[150,151] Observational studies and meta-analyses supported its efficacy in severe refractory SLE.[152,153] EULAR 2023 guidelines recommend rituximab for refractory LN,[12] and KDIGO 2024 guidelines recommend it for active LN unresponsive to initial therapy.[144] The 2021 Chinese LN guidelines recommend it for proliferative LN refractory to conventional immunosuppressants.[154] Additionally, rituximab can rapidly alleviate central nervous system symptoms in patients with refractory neuropsychiatric lupus, including cognitive impairment, neurological symptoms, and epilepsy. It has also been shown to improve corresponding imaging findings.[155] A systematic review study showed that rituximab treatment for neuropsychiatric lupus achieved response rates as high as 73%-100%.[110] A multicenter retrospective cohort study demonstrated that rituximab yielded an initial treatment response rate of 86% in SLE patients with hematologic involvement, with specific response rates of 91% for immune thrombocytopenia, 87.5% for autoimmune hemolytic anemia, and 60% for Evans syndrome.[156] A meta-analysis confirmed an improved complete remission rate in SLE patients with thrombocytopenia without increased infection risk.[157] Therefore, 2023 EULAR guidelines recommend to consider rituximab for acute treatment of severe autoimmune thrombocytopenia.[12]

Obinutuzumab is a fully humanized type II anti-CD20 monoclonal antibody that exhibits stronger B-cell cytotoxic effects compared to rituximab. The phase II NOBILITY trial investigating obinutuzumab in active LN demonstrated a significantly higher complete renal response rate at week 52 compared to placebo, with sustained renal improvement persisting through week 104.[158] The phase 3 REGENCY trial further confirmed a superior complete renal response rate at week 76 for obinutuzumab combined with standard therapy versus placebo, particularly in patients with higher baseline disease activity (UPCR ≥3 g/g, hypocomplementemia, high-titer anti-dsDNA antibodies, class IV LN, or mixed class V LN).[159] Obinutuzumab also demonstrated efficacy in rituximab-refractory LN and non-renal SLE, with additional benefits in reducing overall disease activity.[160]

Anifrolumab, a type I interferon receptor antagonist, has been approved by the U.S. Food and Drug Administration for moderate-to-severe SLE. The phase 2b MUSE trial showed that anifrolumab could significantly increase the proportion of patients achieving SRI-4 response, reduce oral glucocorticoid dose, improve the Composite Lupus Assessment (BICLA) response based on BILAG, and alleviate joint swelling or tenderness and rash compared to placebo. Notably, the anifrolumab group exhibited significantly lower SLE flare rates.[161] Post-hoc analysis of MUSE revealed patients of anifrolumab group achieved LLDAS faster and maintained LLDAS longer than those of placebo group.[162] Although the phase 3 TULIP-1 trial did not meet its primary endpoint of SRI-4 response, anifrolumab significantly outperformed placebo in glucocorticoid tapering, CLASI score improvement, joint symptom relief, and BICLA response rates.[163] TULIP-2 confirmed greater BICLA response rates in anifrolumab group, with more patients achieving prednisone ≤7.5 mg/d and ≥50% CLASI score reduction versus placebo.[164] The TULIP-LN trial demonstrated enhanced complete renal responses in active class III/IV LN patients receiving intensive anifrolumab therapy.[165] Based on the aforementioned research findings, the 2023 EULAR guidelines recommend adding anifrolumab for SLE patients who are unresponsive to hydroxychloroquine (either monotherapy or combined with glucocorticoids) or who are unable to taper glucocorticoids to maintenance doses.[12]

Eculizumab, a recombinant humanized monoclonal antibody selectively inhibiting complement C5, is indicated for paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS) in adults and children. Case reports suggested its potential efficacy in refractory LN, thrombotic microangiopathy secondary to SLE, and antiphospholipid syndrome secondary to SLE.[166, 167, 168, 169, 170, 171] A systematic review reported a 93% response rate in LN-associated thrombotic microangiopathy patients treated with eculizumab.[172]

Question 7: How to treat SLE with organ and systemic involvement?

Question 7.1: How to treat lupus nephritis?

Recommendation 7.1: For Class I and Class II LN with low-grade proteinuria that below the nephrotic range, treatment should be based on extrarenal manifestations (2C). For Class II LN with nephrotic range proteinuria, glucocorticoids and/or conventional immunosuppressants are recommended (2B). For active Class III or IV LN, with or without class V, induction therapy with glucocorticoids plus any one of the following is recommended:mycophenolate mofetil (MMF)(1B) or intravenous cyclophosphamide (IV-CTX)(1B), which may be combined with belimumab (1B); orcalcineurin inhibitors (CNI) (1C); orMMF and CNI (1C). As for maintenance therapy, MMF or azathioprine (AZA) is recommended (1B). If MMF or AZA is contraindicated or not tolerated, CNI or leflunomide may be considered (1C). For pure Class V LN, induction therapy should be stratified based on urinary protein levels (1B). For LN patients with persistent disease activity or inadequate response to initial standard-of-care therapy, addition of rituximab (RTX) may be considered (1B).

The treatment principles and goals for LN are generally consistent with those for SLE, and renal pathology type and disease activity are fundamental to formulate LN treatment plans. The LN pathological classification is primarily based on the 2003 International Society of Nephrology/Renal Pathology Society (ISN/RPS) recommendations[173] and the 2018 ISN/RPS revisions to LN pathological classification and the National Institutes of Health (NIH) activity/chronicity index scoring system.[174]

In current initial treatment regimens for LN, glucocorticoids are universally used. For patients with proliferative LN (with or without class V), it is recommended to start with intravenous methylprednisolone at a dose of 250–500 mg/d for 1–3 days, followed by oral prednisone ≤0.5 mg/kg/d.[12,144] In cases with severe active lesions accompanied by acute kidney injury, the methylprednisolone pulse dose may be increased to 500–1000 mg/d, or the treatment duration may be extended. For class II and class V LN, oral glucocorticoid therapy is typically used, with the dose adjusted based on the severity of proteinuria, renal function, and extrarenal manifestations.

For Class I LN and Class II LN with proteinuria that below the nephrotic range, since renal damage is mild, treatment should be based on extrarenal manifestations of SLE.

For Class II LN with nephrotic range proteinuria, lupus podocytopathy should be considered. For lupus podocytopathy with mild glomerular lesions or mesangial proliferation, glucocorticoid monotherapy and glucocorticoid combined with immunosuppressant therapy have similar remission rates. For cases with focal segmental glomerulosclerosis (FSGS) lesions, the complete remission rate with glucocorticoid monotherapy is only 16.7%, while glucocorticoids combined with conventional immunosuppressants can increase the remission rate to 33.3%. Since the flare rate with glucocorticoid monotherapy is significantly higher than that with combination therapy (89.5% vs. 35.7%, P < 0.001),[175] glucocorticoids combined with conventional immunosuppressants are recommended.

Class III/IV LN, with or without Class V LN, is a progressive disease requiring prompt and effective treatment to suppress renal immune-inflammatory lesions. A meta-analysis showed that for the induction therapy of proliferative LN, MMF and IV-CTX had similar complete remission rates (OR = 1.44, 95% CI: 1.00–2.06), as did CNI and IV-CTX (OR = 1.74, 95% CI: 1.09–2.79). As for the maintenance therapy, MMF had a lower risk of flares than AZA (OR = 0.53, 95% CI: 0.31–0.90), while CNI (OR = 0.64, 95% CI: 0.22–1.88) and IV-CTX (OR = 1.68, 95% CI: 0.51–5.51) showed no statistical difference in flare risk compared to AZA.[97] A Chinese multicenter prospective open-label study included 270 LN patients who achieved remission with the induction therapy with glucocorticoids plus IV-CTX. These patients were randomly assigned to receive maintenance therapy with glucocorticoids plus either leflunomide (LEF) at a does of 20 mg/d or AZA at a target dose of 100 mg/d. At 36 months, there was no significant difference in rates of flares (15.7% vs. 17.8%) or adverse effects between the two groups.[120]

The triple immunosuppressive regimen of glucocorticoids, MMF and CNI is particularly suitable for LN patients with extensive podocyte injury causing massive proteinuria. The multi-target regimen including fixed-dose tacrolimus (TAC), reduced-dose MMF, and glucocorticoids significantly reduces proteinuria and maintains stable renal function. A multicenter RCT of 368 LN patients with active LN and baseline serum creatinine (SCr) ≤3 mg/dL showed that multi-target regimen had significantly higher cumulative complete response rates than IV-CTX (46% vs. 26%, P < 0.001) at 24 weeks, especially for Class IV+V LN (45% vs. 27%), with similar adverse event rates.[176] Another study comparing multi-target maintenance therapy after multi-target induction therapy versus AZA maintenance therapy after IV-CTX induction therapy found no significant difference in renal flare rates (5.47% vs. 7.62%, P = 0.74) at 24-month, but AZA maintenance therapy has a significantly higher rate of adverse events (44.4% vs. 16.4%, P < 0.01).[177] A meta-analysis of 8 trials (801 LN patients) showed multi-target induction therapy had superior complete response rates versus IV-CTX (RR [risk ratio] = 1.94, 95% CI: 1.61–2.33; P < 0.00001), particularly for Class IV (RR = 1.52), Class V (RR = 4.24) and Class IV+V LN (RR = 2.29). Multi-target therapy has lower rates of gastrointestinal symptoms, liver dysfunction, leukopenia and menstrual irregularities, but it increases the risk of new-onset hypertension. Notably, CNI should be used cautiously in patients with severe chronic kidney damage.[178]

The BLISS-LN study enrolled 448 LN patients who were randomly assigned to receive either belimumab or placebo in combination with standard therapy (MMF or CTX followed by AZA). At week 104, the belimumab treatment group showed significantly higher rates of primary renal response (43% vs. 32%; OR = 1.6; 95% CI: 1.0–2.3; P = 0.03) and complete renal response (30% vs. 20%; OR = 1.7; 95% CI: 1.1–2.7; P = 0.02) compared with the placebo group.[139] In the East Asian subgroup of 142 LN patients, belimumab showed higher rates of primary renal response (53% vs. 37%; OR = 1.76; 95% CI: 0.88–3.51) and complete renal response rate (35% vs. 25%; OR = 1.73; 95% CI: 0.80–3.74) at 104 weeks, with significantly better primary response rate at 52 weeks (belimumab-to-placebo 62% to 37%; OR = 2.74; 95% CI: 1.33–5.64).[179] A post-hoc analysis found that belimumab improved outcomes in both newly-diagnosed and flared LN,[180] significantly reducing renal-related events, death and LN flare risk, especially in patients with proliferative LN with UPCR <3 g/g.[140] For patients with class III/IV + V LN who respond to belimumab during the induction therapy phase, combination treatment with belimumab may be continued during the maintenance phase.[144]

For Class V LN, also referred as membranous LN, the initial treatment depends on the severity of proteinuria. For patients with proteinuria <1 g/24 h, the prognosis is generally favorable, and no specific immunosuppressive therapy targeting renal involvement is required. In these cases, treatment should be formulated based on extrarenal manifestations of SLE. General renal protective measures including renin-angiotensin-aldosterone system inhibitors (RAASi) and SGLT2 inhibitors are also recommended.[181] Persistent high-grade proteinuria represents a higher risk for developing end-stage kidney disease (ESKD), cardiovascular events, and thrombotic complications in Class V LN, necessitating conventional immunosuppressive therapy to control proteinuria. Therefore, for patients with 24-hour urinary protein levels of 1.0–3.5 g, treatment with glucocorticoids combined with conventional immunosuppressants is recommended, while for those with 24-hour urinary protein levels >3.5 g/24 h, a regimen combining glucocorticoids with MMF plus CNI is preferred as the first-line therapy. During the maintenance phase, options include MMF, CNI, CNI combined with MMF, AZA, and tripterygium glycosides. A subgroup analysis of a Chinese multi-center study demonstrated that multi-target therapy showed significantly higher complete remission rates compared to IV-CTX (33% vs. 8%, P = 0.01) in Class V LN, with no significant difference in adverse event rates.[176]

RTX effectively depletes B cells and controls disease activity, serving as an important treatment option for refractory LN. A meta-analysis incorporating 31 studies involving 1, 112 SLE/LN patients showed that RTX combination therapy achieved overall, complete, and partial remission rates of 72%, 46%, and 32% respectively, and also reduced disease activity scores and glucocorticoid dosage.[153] Obinutuzumab depletes B cells more effectively, significantly improves complete renal response rates, and remains effective even in LN patients with secondary non-response to rituximab.[159,160]

In addition to optimizing immunosuppressive therapy, the management of LN should also address non-immune factors contributing to disease progression. These include a low-salt diet (sodium intake <5 g/d), avoidance of high-protein intake, smoking cessation, moderate exercise, weight reduction (BMI, body mass index ≤25 kg/m2), preferential use of RAASi to achieve target blood pressure (<120/70 mmHg), reduction of proteinuria, and delay of chronic kidney disease progression.[182]

Question 7.2 How to treat severe thrombocytopenia in SLE?

Recommendation 7.2: The primary treatment goal for SLE-associated immune thrombocytopenia is to minimize bleeding risk. In the acute phase, high-dose glucocorticoids (including intravenous methylprednisolone pulses) or intravenous immunoglobulin (IVIG) may be considered (1A). For patients with inadequate response, glucocorticoids combined with rituximab or conventional immunosuppressants are recommended (2B). Thrombopoietin receptor agonists (TPO-RAs) can be used for life-threatening bleeding events or emergency surgery (1A).

Immune-mediated thrombocytopenia is a common hematologic manifestation of SLE, defined as a platelet count < 100×109/L on two consecutive tests.[34] However, alternative etiologies must be excluded, including viral infections (e.g., Epstein-Barr virus [EBV], cytomegalovirus [CMV]), thrombotic events, hypersplenism, drug-induced thrombocytopenia (e.g., heparin), and thrombotic microangiopathy.

Disease severity should be assessed based on platelet count and bleeding symptoms.[183,184] The therapeutic objective is to maintain a safe platelet count to prevent bleeding, rather than normalize platelet counts. Typically, when the platelet count is greater than 50×109/L, the bleeding risk is relatively low. Active treatment is warranted if the platelet count is ≤ 30×109/L and/or the bleeding score is ≥2 points. For severe thrombocytopenia with a platelet count <10×109/L or a bleeding score >5 points, the platelet count should be rapidly increased to a safe level.[185] Currently, high-quality evidence and guidelines/expert consensus for SLE-associated thrombocytopenia are limited. Management strategies are largely extrapolated from primary immune thrombocytopenia (ITP) guidelines.

A retrospective cohort study involving 18 antinuclear antibody-positive ITP patients showed that glucocorticoid mono-therapy achieves a complete response (CR) rate of 22.2%, with a cumulative flare-free duration of 3.4 months.[186] A prospective multicenter randomized trial conducted in China enrolled 195 adult ITP patients, comparing high-dose dexamethasone (HD-DXM group: 40 mg/d for 4 consecutive days) versus prednisone (PDN group: 1 mg/kg for 4 weeks with tapering). The HD-DXM group demonstrated superior overall initial response rate (82.1% vs. 67.4%, P = 0.044) and complete response rate (50.5% vs. 26.8%, P = 0.001).[187]

A systematic review incorporating 22 RCTs and 3 cohort studies (n = 1, 989) compared low-dose (LD) versus high-dose (HD) IVIG for pediatric ITP. Results showed no significant differences in the response rate (RR = 0.99, 95% CI: 0.96–1.02) or durable remission rate (defined as platelet count maintained within the normal range for at least 3 months without flares or additional drug treatment; RR= 0.97, 95% CI: 0.89–1.07). The two groups also exhibited similar results of time to platelet count elevation and time to hemostasis. Moreover, combined use with glucocorticoid (dexamethasone or methylprednisolone) could improve the therapeutic effect.[188]

For patients with inadequate response to high-dose glucocorticoid and/or IVIG, RTX may be considered. A multi-center retrospective study included 71 SLE patients with immune cytopenia. Before receiving RTX, these patients had received 3.1 ± 1.3 types of treatments, including glucocorticoids (100%) and hydroxychloroquine (88.5%). Among them, 44 patients with concurrent ITP received rituximab in addition to continued glucocorticoid combined with traditional immunosuppressant therapy, and the overall response rate (complete remission rate + partial remission rate) of these patients is 91%.[156]

Glucocorticoids combined with conventional immunosuppressants may prolong flare-free survival in patients. In a retrospective cohort study of 47 SLE patients with immune thrombocytopenia, after acute phase treatment with prednisone at 1 mg/kg/d, the patients received combination therapy with CTX, AZA, or RTX as induction therapy, respectively. CTX induction therapy was associated with a lower flare-free survival compared to RTX (CTX 43.6 weeks vs. RTX 51.8 weeks, P = 0.040) or AZA (CTX 43.6 weeks vs. AZA 51.2 weeks, P = 0.024).[189] A retrospective cohort study of antinuclear antibody (ANA)-positive ITP patients revealed that the combination therapy of glucocorticoids and AZA achieved a significantly higher complete response rate (60.0% vs. 22.2%, P = 0.038) and a trend toward improved overall response rate (86.7% vs. 55.6%, P = 0.070). The combination therapy group also had a significantly prolonged cumulative flare-free survival (median: 7.8 vs. 3.4 months, P = 0.038). Multivariate analysis confirmed that combination therapy was independently associated with longer cumulative flare-free survival.[186]

In a multicenter, open-label, RCT of 120 immune thrombocytopenia patients with a maximum follow-up duration of 2 years, glucocorticoids combined with MMF showed a lower failure rate (RR = 0.41, 95% CI: 0.21–0.80; P = 0.008) and a greater complete response rate (platelet count >100×109/L: 91.5% vs. 63.9%, P < 0.001) compared to glucocorticoid monotherapy. No significant differences were observed in bleeding events or treatment-related adverse effects (including infections) between the two groups.[190]

Combined therapy of glucocorticoids and tacrolimus (1 mg twice daily) was evaluated in a retrospective study of 20 SLE patients with immune thrombocytopenia. At 1 month, 3 patients (15%) achieved complete response, 14 (75%) achieved partial response (PR), and 3 (15%) were non-responders. At 3 months, all patients showed significant platelet improvements, with CR rate increasing to 25% (5 patients) and PR rate to 75%. At 6 months, all patients achieved sustained PR or CR without flare (CR: 75%).[126]

A retrospective study of 43 SLE-ITP patients (refractory to initial glucocorticoid/ immunosuppressants) compared between RTX and CsA with a follow-up period of at least 6 months. CsA could still induce remission in patients with prior treatment failure, but the remission rate was lower than that of patients treated with RTX.[191]

A Chinese single-arm, open-label trial of 14 SLE patients with refractory thrombocytopenia treated with sirolimus for 6 months reported an overall response rate of 71.4% and CR rate of 64.3%.[106]

For patients with poor response to the above-mentioned medications, or in emergency situations requiring rapid platelet elevation (e.g., life-threatening bleeding events or emergency surgery), thrombopoietin agents (e.g., recombinant human TPO [rhTPO], TPO receptor agonists [TPO-RA]) may be used. However, high-quality evidence for SLE-associated thrombocytopenia remains limited, and clinical practice mainly refers to ITP protocols. A multicenter open-label RCT in China comparing RTX+rhTPO versus RTX monotherapy in glucocorticoid-refractory/relapsed ITP demonstrateed that the combined therapy had a superior overall response rate (79.2% vs. 71.1%, P = 0.36) and complete response rate (45.4% vs. 23.7%, P = 0.026) compared with monotherapy.[192] TPO-RAs including eltrombopag olamine and romiplostim have been approved for refractory ITP with favorable safety and efficacy profiles, and are recommended as second-line options in guidelines.

Splenectomy may be considered for SLE-ITP patients who cannot be effectively controlled by glucocorticoids and the aforementioned treatments or have persistent flares, though thrombocytopenia recurrence can still occur after splenectomy.[193]

Question 7.3: How to diagnose and treat antiphospholipid syndrome (APS) in SLE?

Recommendation 7.3: It is recommended to refer to the 2023 ACR/EULAR APS classification criteria for diagnosis (1A). For SLE patients with thrombotic APS, long-term oral vitamin K antagonist anticoagulation therapy is recommended after the first arterial or unprovoked venous thrombotic event (1A). For patients with persistent positivity of antiphospholipid antibodies (APLs) but no thrombotic events, low-dose aspirin (75–100 mg/d) may be considered for primary prophylaxis (2B). For APLs-positive pregnant patients, low-dose aspirin and/or low molecular weight heparin (LMWH) are recommended, with stratified treatment based on prior pregnancy morbidity history, thrombotic history, and types of APLs (2B).

The APLs included in the APS classification criteria mainly consist of anticardiolipin antibody (ACL), anti-β2 glycoprotein I antibody, and lupus anticoagulant (LA). Multiple large-scale prospective cohort studies, including CSTAR, have shown that approximately 30%–45% of SLE patients are APLs positive. APLs can affect vascular endothelial cell function, activate platelets, activate the complement system, promote inflammation and oxidative stress, and interfere with the coagulation and fibrinolytic systems, ultimately leading to clinical manifestations in SLE patients such as arterial/venous thrombotic events, microangiopathy, pregnancy morbidity, and cardiac valve damage. The 2023 ACR/EULAR APS classification criteria[194] have been validated in multiple cohort studies to have very high specificity (95%–99%), and it is recommended to use these criteria in combination with clinical manifestations for the diagnosis of SLE with APS. However, the sensitivity of these criteria is only 81%–84%.[195,196] Therefore, clinical diagnosis of SLE with APS requires comprehensive consideration of the patient’s overall clinical presentation and laboratory findings.

Long-term adequate anticoagulation remains the cornerstone of thrombotic APS treatment. A randomized and double-blind controlled trial published in 2003 involving 114 thrombotic APS patients demonstrated that long-term oral warfarin anticoagulation with a target international normalized ratio (INR) of 2.0–3.0 had comparable efficacy in thrombotic recurrence rates and similar bleeding risks to high-intensity warfarin anticoagulation (INR 3.1–4.0).[197] For high-risk thrombotic APS patients, particularly those with arterial thrombosis, warfarin is recommended over direct oral anticoagulants (DOACs). An open-label randomized controlled trial published in 2018 compared rivaroxaban (20 mg/d) with warfarin (INR 2.0–3.0) in high-risk thrombotic APS patients. This study is the first to demonstrate that the rivaroxaban group has a significantly higher thrombotic recurrence rate (12%, including 4 ischemic strokes and 3 myocardial infarctions) compared to the warfarin group.[198] A meta-analysis of DOACs in thrombotic APS published in 2023 showed that the use of DOAC was associated with an increased risk of arterial thrombotic events (OR = 5.43), particularly stroke (OR = 10.74), and an overall increase in arterial/venous thrombotic events (OR = 4.46).[199] For APLspositive SLE patients without prior thrombotic events but with high-risk APL profiles (LA positivity, two APLs [any combination of LA, ACL, or anti-β2 glycoprotein I antibodies], or triple APLs positivity), low-dose aspirin (75–100 mg/d) should be considered for primary prophylaxis (Table 5).

Table 5

Treatment Recommendations for SLE with APL Positivity

Patient Population Recommendation Evidence Level Recommendation Grade
Thrombotic Event Management
APLs-positive SLE patients without symptoms Aspirin (75–100 mg/d) for primary prophylaxis 2a B
First unprovoked venous thrombosis Long-term anticoagulation with vitamin K antagonist (INR 2.0–3.0) 2b B
First arterial thrombosis in confirmed APS Vitamin K antagonist preferred over aspirin alone; individualized INR targets (2.0–3.0 or 3.0–4.0) based on bleeding/thrombosis risk; considering warfarin plus aspirin 2b C
Triple-APLs-positive patients with thrombosis Rivaroxaban not recommended 1b B
Pregnancy Morbidity Management
APL-positive SLE patients without prior thrombosis/pregnancy morbidity Aspirin (75–100 mg/d) 1b B
APL-positive SLE patients with prior thrombosis Aspirin combine with therapeutic-dose LMWH 1b B
APL-positive SLE patients with recurrent early miscarriage meeting the APS classification criteria Aspirin combine with prophylactic-dose LMWH 2b B
APL-positive SLE patients with late miscarriage or pathological pregnancy related to placental insufficiency meeting the APS classification criteria Aspirin combine with therapeutic-dose LMWH 2b B

A small subset of APS patients may develop catastrophic APS (CAPS), characterized by multiple (≥3) organ thromboses within one week, involving critical organs such as the brain, kidneys, liver, or heart, leading to organ failure and death. The incidence of CAPS is approximately 1.0%, but the mortality rate is as high as 50%–70%. The pathogenesis may involve acute thrombotic storms and inflammatory cascades. High-quality evidence for the treatment of SLE with CAPS is lacking, but early diagnosis and aggressive treatment are critical.[200,201] Early intervention for infections and avoidance of anticoagulation interruption or dose reduction may help prevent CAPS. First-line treatment for CAPS includes heparin anticoagulation combined with glucocorticoids, plasma exchange, and/or intravenous immunoglobulin,[202] along with active identification and management of triggers such as infections or malignancies. Case reports and small-scale studies suggested that refractory CAPS may be treated with rituximab (B-cell depletion) or eculizumab (complement pathway blockade), but large-scale controlled studies are still lacking.[203]

APLs positivity is an independent risk factor for pregnancy morbidity in SLE patients.[204] Multiple cohort studies on APS-related pregnancy outcomes have demonstrated that APLspositive patients have significantly higher rates of pregnancy complications, including recurrent early miscarriage, intrauterine fetal death, preeclampsia/eclampsia, gestational hypertension, fetal growth restriction, and preterm birth. During the perinatal period, SLE patients with persistent APLs positivity should be stratified based on prior thrombotic history, pregnancy morbidity history, and types of APLs to develop individualized treatment plans, with low-dose aspirin and/or LMWH recommended. A systematic review of 11 studies (including 9 RCTs and 2 quasi-RCTs) involving 1672 APLs-positive women (including SLE patients) showed that treatment with aspirin and heparin (unfractionated heparin or LMWH) during pregnancy significantly increased live birth rates compared to placebo or aspirin alone (RR = 1.27, 95% CI: 1.09–1.49), with no serious adverse effects or teratogenicity.[205]

Question 7.4: How to treat neuropsychiatric SLE (NPSLE)?

Recommendation 7.4: For confirmed active NPSLE, glucocorticoids combined with immunosuppressants are recommended (1B). For patients with persistent APLs or cerebrovascular disease, combined antiplatelet and/or anticoagulant therapy should be considered (2B).

The treatment principles and goals of NPSLE should follow the general principles and goals of SLE treatment. The treatment regimen includes glucocorticoids combined with immunosuppressants, antiplatelet/anticoagulant therapy, and symptomatic management.

For induction therapy of active NPSLE, glucocorticoids combined with immunosuppressants are currently recommended.[12] For patients with mild to moderate NPSLE (e.g., mild cognitive dysfunction and headache without definitive organic neurological damage), the recommended glucocorticoid dose is prednisone 0.5–1 mg/kg/d or equivalent. For severe NPSLE with inflammatory manifestations (e.g., myelopathy, acute confusional state, and status epilepticus), early high-dose methylprednisolone pulse therapy (250–1000 mg/d intravenously for 3–5 days) is recommended.[206]

A systematic review reported that immunosuppressants used for NPSLE treatment include CTX, MMF, and AZA.[110] In severe NPSLE patients, high-dose methylprednisolone pulse therapy combined with intravenous CTX improved psychiatric symptoms, with a total improvement rate of 94.7%, significantly superior to methylprednisolone pulse therapy alone (46.2%).[207] A post-hoc subgroup analysis of an RCT comparing MMF and IV-CTX in non-renal SLE patients showed no statistical difference in achieving NPSLE remission between the two treatments. [208] After NPSLE reaches remission, maintenance therapy with low-dose glucocorticoids (≤5 mg/d prednisone or equivalent) combined with MMF or AZA is recommended.[209]

Multiple clinical studies and systematic reviews have reported that rituximab (monotherapy or combined with CTX) is effective for refractory NPSLE.[155,210,211] A post-hoc analysis of two phase 3 studies on belimumab involving 45 mild NPSLE patients demonstrated that belimumab improved headache symptoms. [212] However, a pooled analysis of five clinical studies on belimumab showed no clear protective effect against NPSLE flares. [213] The therapeutic effects of telitacicept and anifrolumab in NPSLE remain unproven.

Observational studies have identified antimalarials as protective factors against NPSLE, particularly against epileptic seizures.[214] Intrathecal injection of dexamethasone and MTX for NPSLE treatment is associated with higher survival rates and flare-free survival rates. In the subgroup of NPSLE patients with elevated cerebrospinal fluid protein levels, intrathecal injection of dexamethasone and MTX significantly improved prognosis (P < 0.001) ,[215] but attention should still be paid to the neurotoxicity of MTX in sensitive individuals. A systematic review indicateed that IVIG and plasma exchange may be effective for inducing clinical remission of NPSLE.[110]

For NPSLE patients with persistent APLs positivity, both Chinese and international guidelines recommend antiplatelet therapy as primary prophylaxis to reduce thrombotic events.[216] For thrombotic cerebrovascular disease, standard-intensity vitamin K antagonist therapy (target INR 2.0–3.0) should be initiated as secondary prevention.

For NPSLE patients with epilepsy, treatment with valproate, lamotrigine, or levetiracetam is recommended. Diazepam is the first-line treatment for terminating status epilepticus.[217] Antianxiety/antidepressant medications and psychological interventions can effectively improve psychiatric symptoms in NPSLE patients.[218] Meanwhile, prevention and identification of nervous system infections, blood pressure management, and metabolic factor monitoring are also essential.

Question 8: What other therapeutic measures are available for SLE?

Recommendation 8: Plasma exchange or immunoadsorption may be considered as adjunctive therapy for severe or refractory SLE (2C). Intravenous immunoglobulin (IVIG) may be added for refractory SLE or SLE complicated with infections (2D).

Plasma exchange and immunoadsorption (clearance of circulating immunoglobulin and immunocomplexes) may provide short-term clinical improvements in severe or refractory SLE and serve as an adjuvant to first-line therapies.[110,219] A real-world retrospective study suggested that whole blood exchange therapy helped increase hemoglobin levels more rapidly in refractory autoimmune hemolytic anemia.[220] A meta-analysis of 18 RCTs (1990–2020, n= 457, Chinese SLE patients) comparing immunoadsorption (IAS) versus non-IAS showed that IAS combined with medications reduced serum IgG, creatinine, anti-nuclear antibodies, urinary protein, and tumor necrosis factor-alpha (TNF-α) levels, while increased levels of complement C3 and C4. However, IAS groups had higher risks of fever or chills, hypotension, and bleeding.[221]

IVIG may benefit patients with refractory SLE or SLE complicated with infections.[222] For LN unresponsive to induction therapy, IVIG demonstrated response rates of 60%–70% (excluding class V LN).[223]

Janus kinase inhibitors (JAKi), by blocking the JAK-STAT signaling pathway, disrupt cytokine-mediated signaling, thereby mitigating inflammation and immune dysregulation. JAKi hold significant potential for SLE treatment, particularly in patients refractory to conventional therapies. A meta-analysis of JAKi for SLE demonstrated superior efficacy over placebo, especially in improving musculoskeletal and mucocutaneous manifestations. JAKi-treated patients showed higher rates of achieving SRI-4 (risk ratio [RR] = 1.18, 95% CI: 1.07–1.31; P = 0.001), BICLA response (RR= 1.16, 95% CI: 1.02–1.31; P = 0.02), LLDAS attainment (RR = 1.28, 95% CI: 1.07–1.54; P = 0.008), and remission of arthritis and rash in SLEDAI-2000 (RR= 1.09, 95% CI: 1.00–1.18; P = 0.04), with similar adverse event rates to placebo (RR= 1.01, 95% CI: 0.97–1.04; P = 0.65).[224] A phase II RCT (BRAVE I) of baricitinib (4 mg) in SLE revealed that 70 out of 104 patients (67%) achieved remission of arthritis and rash at week 24 (OR = 1.8 vs. placebo, 95% CI: 1.0–3.3; P = 0.0414), which significantly improved symptoms in patients with active SLE inadequately controlled by standard therapy, whereas baricitinib at a dose of 2 mg showed no benefits. The phase III SLE-BRAVE-I trial published in The Lancet in 2023 confirmed a superior SRI-4 response rate with baricitinib at a dose of 4 mg (P = 0.016).[225] However, another phase III RCT published in The Lancet in 2023 failed to replicate these findings.[226] Case reports and case series highlighted the efficacy of tofacitinib in SLE-associated cutaneous lesions, alopecia, and arthritis,[227, 228, 229] and the efficacy of baricitinib in cutaneous manifestations and alopecia.[230, 231, 232] Emerging data suggest that novel JAKi (e.g., deucravacitinib[233] and filgotinib[234]) may also have efficacy for SLE, warranting further clinical exploration. Collectively, JAKi represent a potential therapeutic option, particularly for SLE with prominent cutaneous or musculoskeletal involvement.

Additional study suggests that tripterygium glycosides combined with conventional therapy may reduce lupus activity and improve overall response rate.[235] However, the overall incidence of reproductive toxicity in patients using tripterygium glycosides is 17.9% (95% CI: 14.1%–22.5%),[236] necessitating caution in clinical practice.

In recent years, genetically engineered precision cell therapies, particularly chimeric antigen receptor T cells (CAR-T), have shown preliminary success in treating refractory SLE. Case reports and case series indicated that CD19-targeted autologous CAR-T cells can effectively eliminate pathogenic autoantibodies, markedly reduce proteinuria levels, and improve renal function in refractory LN, thereby achieving low disease activity or clinical remission.[237, 238, 239] In refractory SLE-associated thrombocytopenia, CAR-T therapy has demonstrated significant platelet count increase, enabling glucocorticoids and immunosuppressants tapering or discontinuation.[240] A phase I trial of dual-targeting CD19 and B-cell maturation antigen (BCMA) autologous CAR-T cells in refractory SLE patients also reported favorable efficacy and safety profiles.[241] However, challenges such as complex manufacturing processes, high treatment costs, uncertainties regarding long-term efficacy, safety, and optimal therapeutic protocols necessitate further high-quality clinical investigations.

Question 9: How to select and use non-pharmacological therapies for SLE?

Recommendation 9: Non-pharmacological therapies for SLE patients aim to improve health-related quality of life and should follow these principles: (1) moderate exercise; (2) social and psychological interventions; (3) avoidance of common hazardous substances; (4) sun protection; (5) smoking cessation; and (6) vitamin D supplementation (1C).

Non-pharmacological therapies for SLE patients aim to improve health-related quality of life. Exercise interventions were shown to reduce depression (standardized mean difference [SMD] = -0.40, 95% CI: -0.71 to-0.09) and fatigue (mean difference [MD] = -0.52, 95% CI: -0.91 to-0.13).[242,243] Psychological interventions have been found to decrease anxiety (SMD = -0.95, 95% CI: -1.57 to-0.34), mental stress (SMD = -0.63, 95% CI: -1.02 to-0.23), and depression (SMD = -1.14, 95% CI: -1.84 to-0.44), and also aid in the control of disease activity (SMD = -0.34, 95% CI: -0.57 to-0.11).[244] The skin is the most commonly affected organ in SLE, and certain cosmetics may contain substances that exacerbate lupus activity.[245] Additionally, SLE patients should avoid exposure to hair dyes and eyebrow dyes.[246,247] Ultraviolet (UV) light exposure can trigger SLE flares, and sun protection measures (e.g., sunscreen) can prevent UV-induced skin irritation and reduce skin inflammation,[248, 249, 250] thereby decreasing the risk of disease flares. Compared to non-smokers, smokers have a higher risk of SLE (OR = 1.49, 95% CI: 1.06 to2.08), a higher score of SLEDAI (15.6 ±7.8 vs. 9.0 ±5.8), and a poorer response to pharmacological treatments (OR = 0.53, 95% CI: 0.31 to0.93).[251,252] Osteoporosis is a major comorbidity in SLE patients, and their serum vitamin D levels are significantly lower than those in healthy individuals. Therefore, vitamin D supplementation may alleviate inflammation and reduce disease activity in SLE patients.[253,254]

Question 10: How to prevent and control infections in SLE?

Recommendation 10: Timely assessment of infection risk in SLE patients is essential, and infections should be promptly prevented, identified, and managed (1B).

In China, the proportion of SLE patients dying from infections has been increasing annually, and infections have become the leading cause of death in SLE patients, with over 50% of patients dying due to infections.[255] Key risk factors for infections in SLE patients include methylprednisolone pulse therapy (HR = 1.5, 95% CI: 1.1 to 2.1), the doses of glucocorticoid exceeding prednisone 7.5 mg/d (OR = 2.7, 95% CI: 1.3 to 5.3), disease activity (HR = 1.0, 95% CI: 1.0 to 1.0), long disease duration (HR = 1.2, 95% CI: 1.1 to 1.3), involvement of the hematological system (OR = 2.5, 95% CI: 1.3 to 4.9), and serositis (OR= 2.8, 95% CI: 1.3 to 5.7). Hydroxychloroquine has been shown to reduce the risk of severe infections (HR = 0.7, 95% CI: 0.5 to 1.0).[128,256] The serum C-reactive protein ≥46.8 mg/L, procalcitonin ≥0.53 μg/L, and lymphocyte count ≤1.0×109/L suggest a high likelihood of infection,[257,258] which warrants timely assessment, prompt identification, and active treatment, along with measures to prevent infection.

Question 11: How to manage SLE patients during the peri-gestational period?

Recommendation 11: SLE patients should prepare for pregnancy after meeting the necessary conditions and completing pre-pregnancy counseling and risk assessment. Pregnancy can be considered in SLE patients when meeting the following criteria: (1) The disease has been stable for at least 6 months; (2) oral prednisone ≤15 mg/d (or equivalent doses of non-fluorinated glucocorticoids); (3) discontinuation of teratogenic drugs for the required duration; (4) 24-hour urine protein <0.5 g/d; and (5) no signs of vital organ damage (2B). For pregnant SLE patients, an individualized follow-up plan should be developed by a multidisciplinary team, including rheumatologists and obstetricians, to closely monitor the disease activity, and fetal growth and development (1C). If there is no contraindication, hydroxychloroquine (HCQ) should be used throughout pregnancy (1B).

To reduce pregnancy complications in women with SLE and achieve favorable pregnancy outcomes, adequate preparation before conception and strict monitoring of the disease during pregnancy are necessary. Women with SLE who have severe organ dysfunction and/or damage should be informed about the risks associated with pregnancy.[259,260] Compared to patients with non-active LN, patients with active LN have significantly higher risks of disease flares during pregnancy (OR = 2.04, 95% CI: 1.21 to 3.45), preeclampsia (OR = 2.62, 95% CI: 1.36 to 5.05), fetal loss (OR = 4.90, 95% CI: 1.54 to 15.59), and preterm birth (OR = 4.26, 95% CI: 2.19 to 8.31).[261] Patients who have been in remission for more than 6 months before pregnancy, with urine protein <0.5 g/d, no renal failure, and discontinuation of teratogenic immunosuppressants for more than 1 year, have markedly higher rates of full-term delivery (76.47% vs. 23.08%) and live births (80.39% vs. 30.77%) compared to patients with active disease during the 6 months before pregnancy. They also have significantly lower risks of gestational hypertension (17.65% vs. 23.08%) and preeclampsia or eclampsia (9.80% vs. 15.38%).[262]

Pre-pregnancy counseling is crucial for successful pregnancies in SLE patients, as planned pregnancies reduce the risks of disease flares and adverse pregnancy outcomes compared to unintended pregnancies.[263,264] SLE can increase the risk of adverse pregnancy outcomes,[265] so strict disease control is essential before pregnancy. A multidisciplinary team (including at least rheumatologists and obstetricians) should perform pre-pregnancy assessment, strictly evaluate indications and contraindications for pregnancy, and closely monitor disease activity throughout pregnancy to achieve favorable maternal and fetal outcomes.[266,267] Specific measures for multidisciplinary management of SLE pregnancies by rheumatologists and obstetricians are detailed in the “2022 Chinese guideline for the management of pregnancy and reproduction in systemic lupus erythematosus”.[268]

Close monitoring of disease activity and fetal growth and development in SLE patients during pregnancy is critical for favorable maternal and fetal outcomes. Screening risk factors associated with pregnancy-related complications, such as anti-phospholipid antibodies, should be conducted before pregnancy. During pregnancy, strict monitoring of disease activity, placental function, and fetal growth and development is essential.[267,269, 270, 271, 272, 273]

HCQ can reduce the risks of disease activity, disease flares, and adverse fetal outcomes such as preterm birth in SLE patients. Continuous HCQ therapy is associated with a lower incidence of SLE flares during and after pregnancy. Therefore, HCQ should be used continuously throughout pregnancy if no contraindications exist.

Question 12: How to manage vaccination in SLE patients?

Recommendation 12: Vaccination is an effective measure to prevent infections in SLE patients. The feasibility and timing of vaccination for adult SLE patients should be determined based on disease activity, current treatment regimen, type of vaccines, and patient preferences (2D).

Infections remain one of the leading causes of death in SLE patients. Glucocorticoids and cytotoxic immunosuppressive agents can interfere patients’ immune responses to viral and bacterial pathogens, increasing the risk of infections. Current evidence indicates that SLE patients generally show good immunogenicity and tolerance to vaccines, which can decrease the rates of emergency visits and hospitalization due to infections, as well as the occurrence of invasive infections. No evidence up to now suggests that vaccination increases the risk of disease flares. Therefore, multiple international guidelines recommend that rheumatologists should conduct a comprehensive assessment of vaccination status for SLE patients annually.[274,275] The vaccination should be scheduled based on current disease activity, current glucocorticoid dosage, use of immunosuppressive agents or biologics (especially B-cell-depleting therapies), and patient preferences. In general, SLE patients receiving glucocorticoids and immunosuppressive agents can receive inactivated vaccines, but live-attenuated vaccines are not recommended.

Currently, vaccines recommended for SLE patients include influenza vaccines, pneumococcal vaccines, hepatitis B vaccines, human papillomavirus vaccines, and herpes zoster vaccines. All of these vaccines available in China are inactivated or polysaccharide/recombinant protein formulations. Detailed vaccination indications and immunosuppressive drug adjustments are detailed in Table 6.

Table 6

Vaccination for SLE Patients

Vaccination Eligible Population Adjustment of Immunosuppressive Therapy
Influenza Vaccine Patients aged ≥65 years, and patients aged 18–65 years who are receiving immunosuppressive therapy. Discontinue MTX for 2 weeks after vaccination.
Pneumococcal Vaccine Patients aged <65 years who are receiving immunosuppressive therapy. If receiving glucocorticoids equivalent to prednisone ≥20 mg/d, delay vaccination until the dose is reduced to < 20 mg/d.
Hepatitis B Vaccine Patients susceptible to hepatitis B virus. Delay rituximab administration for 2 weeks after vaccination.
HPV Vaccine Patients aged 9–45 years who are receiving immunosuppressive therapy and have not been previously vaccinated for HPV.
Recombinant Herpes Zoster Vaccine Patients aged >18 years who are receiving immunosuppressive therapy.
  1. a

    For patients receiving glucocorticoids equivalent to prednisone <20 mg/d, if vaccination is critical and discontinuation of glucocorticoids pose high risks of disease flares or adrenal insufficiency, continuation of low-dose glucocorticoids may be considered. bFor patients receiving methotrexate ≤0.4 mg/kg/week or azathioprine ≤3 mg/kg/d (low-level immunosuppressive therapy), if vaccination is critical and discontinuation of immunosuppressants pose high risks of disease flares, the duration of drug discontinuation may be shortened. HPV, human papillomavirus.

Lead Experts

Xiaofeng Zeng (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Mengtao Li (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College).

Lead Methodology Expert

Yaolong Chen (Innovation Unit of Evidence-Based Evaluation and Guidelines, Chinese Academy of Medical Sciences; Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University).

Steering Committee

Xiaofeng Zeng (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Mengtao Li (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Yaolong Chen (Innovation Unit of Evidence-Based Evaluation and Guidelines, Chinese Academy of Medical Sciences; Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University); Xinping Tian (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Jiuliang Zhao (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College).

Evidence Evaluation Group and Secretariat

Yinghua Chen (Department of Nephrology, Eastern Theater General Hospital); Yisha Li (Department of Rheumatology and Immunology, Xiangya Hospital, Central South University); Tingting Liu (Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine); Shangzhu Zhang (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Ting Zhang (Department of Rheumatology and Immunology, The Second Affiliated Hospital, Zhejiang University School of Medicine); Mengmeng Zhao (Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University); Yangzhong Zhou (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Ling Wang (School of Population Medicine and Public Health, Peking Union Medical College); Yuanyuan Yao (School of Basic Medical Sciences, Lanzhou University); Ye Wang (School of Public Health, Lanzhou University); Zhewei Li (School of Public Health, Lanzhou University); Bingyi Wang (School of Basic Medical Sciences, Lanzhou University).

Expert Panel (Listed in Alphabetical Order by Surname)

Shuhong Chi (Department of Rheumatology and Immunology, General Hospital of Ningxia Medical University); Zhanyun Da (Department of Rheumatology and Immunology, Affiliated Hospital of Nantong University); Shengming Dai (Department of Rheumatology and Immunology, Shanghai Sixth People’s Hospital); Xinwang Duan (Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University); Lingli Dong (Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology); Xuebing Feng (Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Nanjing University Medical School); Jinsong Gao (Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Lan He (Department of Rheumatology and Immunology, The First Affiliated Hospital of Xi’an Jiaotong University); Wenhui Huang (Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guangzhou Medical University); Lindi Jiang (Department of Rheumatology and Immunology, Zhongshan Hospital, Fudan University); Zhenyu Jiang (Department of Rheumatology and Immunology, The First Hospital of Jilin University); Hongzhong Jin (Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Caifeng Li (Department of Rheumatology and Immunology, Beijing Children’s Hospital, Capital Medical University); Fen Li (Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University); Hongbin Li (Department of Rheumatology and Immunology, Affiliated Hospital of Inner Mongolia Medical University); Mengtao Li (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Juan Li (Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University); Xiaomei Li (Department of Rheumatology and Immunology, The First Affiliated Hospital of University of Science and Technology of China); Jin Lin (Department of Rheumatology and Immunology, The First Affiliated Hospital, Zhejiang University); Shudian Lin (Department of Rheumatology and Immunology, Hainan General Hospital); Shunping Lin (Department of Rheumatology and Immunology, Fujian Medical University Union Hospital); Zhiming Lin (Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-sen University); Dongzhou Liu (Department of Rheumatology and Immunology, Shenzhen People’s Hospital); Shengyun Liu (Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University); Yi Liu (Department of Rheumatology and Immunology, West China Hospital, Sichuan University); Hui Luo (Department of Rheumatology and Immunology, Xiangya Hospital, Central South University); Liangjing Lyu (Department of Rheumatology and Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine); Hanyou Mo (Department of Rheumatology and Immunology, The First Affiliated Hospital of Guangxi Medical University); Haili Shen (Department of Rheumatology and Immunology, The Second Hospital of Lanzhou University); Xiaofei Shi (Department of Rheumatology and Immunology, The First Affiliated Hospital of Henan University of Science and Technology); Qiang Shu (Department of Rheumatology, Qilu Hospital of Shandong University); Yijun Song (Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Yin Su (Department of Rheumatology and Immunology, Peking University People’s Hospital); Lingyun Sun (Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Nanjing University Medical School); Wenfeng Tan (Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanjing Medical University); Xinping Tian (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Caihong Wang (Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University); Jing Wang (Department of Rheumatology and Immunology, Yunnan Provincial First People’s Hospital); Qian Wang (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Yongfu Wang (Department of Rheumatology and Immunology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology); Wei Wei (Department of Rheumatology and Immunology, Tianjin Medical University General Hospital); Zhenbiao Wu (Department of Rheumatology and Immunology, The Second Affiliated Hospital of Air Force Medical University); Lijun Wu (Department of Rheumatology and Immunology, People’s Hospital of Xinjiang Uygur Autonomous Region; Xinjiang Clinical Research Center for Rheumatoid Arthritis); Jian Xu (Department of Rheumatology and Immunology, The First Affiliated Hospital of Kunming Medical University); Jing Xue (Department of Rheumatology and Immunology, The Second Affiliated Hospital, Zhejiang University School of Medicine); Chengde Yang (Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine); Min Yang (Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University); Niansheng Yang (Department of Rheumatology and Immunology, The First Affiliated Hospital of Sun Yat-sen University); Pingting Yang (Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University); Qibing Xie (Department of Rheumatology and Immunology, West China Hospital, Sichuan University); Jiashun Zeng (Department of Rheumatology and Immunology, Affiliated Hospital of Guizhou Medical University); Xiaofeng Zeng (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Liyun Zhang (Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences); Miaojia Zhang (Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanjing Medical University); Xiao Zhang (Department of Rheumatology and Immunology, The Eighth Affiliated Hospital of Sun Yat-sen University); Wen Zhang (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Zhuoli Zhang (Department of Rheumatology and Immunology, Peking University First Hospital); Dongbao Zhao (Department of Rheumatology and Immunology, The First Affiliated Hospital of Naval Medical University); Jiuliang Zhao (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Yan Zhao (Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College); Yi Zhao (Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University); Jing Zhu (Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital).


Address for correspondence: Mengtao Li, Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.

Funding statement: The Chinese National Key Technology R & D Program, Ministry of Science and Technology (2021YFC2501300); Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (CIFMS)(2021-I2M-1-005); Beijing Municipal Science & Technology Commission (Z201100005520027); National High Level Hospital Clinical Research Funding (2022-PUMCH-D-009).

Acknowledgements

None.

  1. Author contributions

    Jiuliang Zhao: Conceptualization, Writing—Original draft preparation, Writing—Reviewing and Editing. Shangzhu Zhang: Writing—Original draft preparation. Qian Wang: Supervision. Xinping Tian: Writing—Reviewing and Editing, Supervision. Yaolong Chen: Supervision. Mengtao Li: Supervision, Project administration. Xiaofeng Zeng: Supervision, Project administration.

  2. Ethical approval

    Not applicable.

  3. Informed consent

    Not applicable.

  4. Conflict of interest

    Jiuliang Zhao and Qian Wang are Editorial Board Members of the journal; Xinping Tian is the Executive Editor-in-Chief; Mengtao Li is an Associate Editor-in-Chief; and Xiaofeng Zeng is the Editor-in-Chief. This article was subjected to the standard procedures of the journal, with peer review process independent of these editors and their research groups. All other authors declare no conflicts of interest.

  5. Use of large language models, AI and machine learning tools

    None declared.

  6. Data availability statement

    No additional data is available.

  7. Additional disclosure

    The guideline of Chinese-language version was published in Chinese Medical Journal (Chinese Version) in June 2025. This English-language version is published after minor revisions with permission by the original authors and the Chinese Medical Journal.

References

[1] Durcan L, O’Dwyer T, Petri M. Management strategies and future directions for systemic lupus erythematosus in adults. Lancet. 2019;393:2332–2343.10.1016/S0140-6736(19)30237-5Suche in Google Scholar PubMed

[2] Fava A, Petri M. Systemic lupus erythematosus: Diagnosis and clinical management. J Autoimmun. 2019;96:1–13.10.1016/j.jaut.2018.11.001Suche in Google Scholar PubMed PubMed Central

[3] Lisnevskaia L, Murphy G, Isenberg D. Systemic lupus erythematosus. Lancet,. 2014;384:1878–1888.10.1016/S0140-6736(14)60128-8Suche in Google Scholar PubMed

[4] Barber MRW, Drenkard C, Falasinnu T, et al. Global epidemiology of systemic lupus erythematosus. Nat Rev Rheumatol. 2021;17:515–532.10.1038/s41584-021-00668-1Suche in Google Scholar PubMed PubMed Central

[5] Li M, Li C, Cao M, et al. Incidence and prevalence of systemic lupus erythematosus in urban China, 2013–2017: A nationwide population-based study. Sci Bull (Beijing). 2024;69:3089–3097.10.1016/j.scib.2024.04.075Suche in Google Scholar PubMed

[6] Li M, Zhang W, Leng X, et al. Chinese SLE Treatment and Research group (CSTAR) registry: I. Major clinical characteristics of Chinese patients with systemic lupus erythematosus. Lupus. 2013;22:1192–1199.10.1177/0961203313499086Suche in Google Scholar PubMed

[7] Zhang S, Su J, Li X, et al. Chinese SLE Treatment and Research group (CSTAR) registry: V. gender impact on Chinese patients with systemic lupus erythematosus. Lupus. 2015;24:1267–1275.10.1177/0961203315585813Suche in Google Scholar PubMed

[8] Wang Z, Li M, Zhao J, et al. 220 Clinical characteristics and remission of patients with systemic lupus erythematosus in china: results from SLE treatment and research group (CSTAR) registry with a real-time collecting system. Lupus Sci Med. 2019;6(Suppl 1):A164.1-A164.10.1136/lupus-2019-lsm.220Suche in Google Scholar

[9] Merrell M, Shulman LE. Determination of prognosis in chronic disease, illustrated by systemic lupus erythematosus. J Chronic Dis. 1955;1:12–32.10.1016/0021-9681(55)90018-7Suche in Google Scholar PubMed

[10] Borchers AT, Keen CL, Shoenfeld Y, et al. Surviving the butterfly and the wolf: mortality trends in systemic lupus erythematosus. Autoimmun Rev. 2004;3:423–453.10.1016/j.autrev.2004.04.002Suche in Google Scholar PubMed

[11] Tektonidou MG, Lewandowski LB, Hu J, et al. Survival in adults and children with systemic lupus erythematosus: a systematic review and Bayesian meta-analysis of studies from 1950 to 2016. Ann Rheum Dis. 2017;76:2009–2016.10.1136/annrheumdis-2017-211663Suche in Google Scholar PubMed

[12] Fanouriakis A, Kostopoulou M, Andersen J, et al. EULAR recommendations for the management of systemic lupus erythematosus: 2023 update. Ann Rheum Dis. 2024;83:15–29.10.1136/ard-2024-225617Suche in Google Scholar PubMed

[13] Gordon C, Amissah-Arthur MB, Gayed M, et al. The British Society for Rheumatology guideline for the management of systemic lupus erythematosus in adults. Rheumatology (Oxford). 2018;57:e1–e45.10.1093/rheumatology/kex286Suche in Google Scholar PubMed

[14] Mok CC, Hamijoyo L, Kasitanon N, et al. The Asia-Pacific League of Associations for Rheumatology consensus statements on the management of systemic lupus erythematosus. Lancet Rheumatol. 2021;3:e517–e531.10.1016/S2665-9913(21)00009-6Suche in Google Scholar PubMed

[15] Chinese Rheumatology Association. [Guidelines for the Diagnosis and Treatment of Systemic Lupus Erythematosus]. Chin J Rheumatol. 2010;14:342–346.Suche in Google Scholar

[16] Chinese Rheumatology Association; National Clinical Research Center for Dermatologic and Immunologic Diseases; Chinese Systemic Lupus Erythematosus Treatment and Research Group. [2020 Chinese guidelines for the diagnosis and treatment of systemic lupus erythematosus]. Zhonghua Nei Ke Za Zhi. 2020;59:172–185.Suche in Google Scholar

[17] Wu CY, Li CF, Wu QJ, et al. Chinese Systemic Lupus Erythematosus Treatment and Research Group Registry IX: Clinical Features and Survival of Childhood-Onset Systemic Lupus Erythematosus in China. Chin Med J (Engl). 2017;130:1276–1282.10.4103/0366-6999.206346Suche in Google Scholar PubMed PubMed Central

[18] Li M, Wang Z, Wang Y, et al. THU0281 Long-term prognosis and predictingfactors of chinese patients with systemic lupus erythematosus: a multi-center cohort study from cstar registry. Ann Rheum Dis. 2017;76:310.10.1136/annrheumdis-2017-eular.5791Suche in Google Scholar

[19] World Health Organization. WHO Handbook for Guideline Development. 2nd ed. 2014. Available at: https://www.who.int/publications/i/item/9789241548960. Accessed on Aug. 15, 2025.Suche in Google Scholar

[20] Jiang Z, Zhan S, Jia X. [Basic methods and procedures for formulating/revising the Clinical Diagnosis and Treatment Guidelines]. Natl Med J China. 2016;96:250-253.Suche in Google Scholar

[21] Brouwers MC, Kho ME, Browman GP, et al. AGREE II: advancing guideline development, reporting, and evaluation in health care. Prev Med. 2010;51:421–424.10.1016/j.ypmed.2010.08.005Suche in Google Scholar PubMed

[22] Chen Y, Yang K, Marušic A, et al. A Reporting Tool for Practice Guidelines in Health Care: The RIGHT Statement. Ann Intern Med. 2017;166:128–132.10.7326/M16-1565Suche in Google Scholar PubMed

[23] Chen Y, Wang X, Wang Q. [Follow the reporting guidance to improve the quality of guidelines report.] Zhonghua Nei Ke Za Zhi. 2018;57:168–170.Suche in Google Scholar

[24] Shea BJ, Grimshaw JM, Wells GA, et al. Development of AM-STAR: a measurement tool to assess the methodological quality of systematic reviews. BMC Med Res Methodol. 2007;7:10.10.1186/1471-2288-7-10Suche in Google Scholar PubMed PubMed Central

[25] Higgins JP, Altman DG, Gøtzsche PC, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.10.1136/bmj.d5928Suche in Google Scholar PubMed PubMed Central

[26] Whiting PF, Rutjes AW, Westwood ME, et al. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155:529–536.10.7326/0003-4819-155-8-201110180-00009Suche in Google Scholar PubMed

[27] Wells GA, Shea B, O’Connell D, et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. 2000.Suche in Google Scholar

[28] Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol. 2011;64:383–394.10.1016/j.jclinepi.2010.04.026Suche in Google Scholar PubMed

[29] Chen YL, Yao L, Norris S, et al. [Application of GRADE in systematic reviews: necessity, frequently-asked questions and concerns]. Chin J Evid Based Med. 2013; 13:1401–1404.Suche in Google Scholar

[30] Yao L, Chen YL, Du L, et al. [Application of GRADE in systematic reviews of diagnostic accuracy tests: a case analysis.] Chin J Evid Based Med. 2014;14:1407–1412.Suche in Google Scholar

[31] Chen YL, Yao L, Du L, et al. [Rationales, methods, challenges and development tendency of using GRADE in systematic reviews of diagnostic accuracy tests.] Chin J Evid Based Med 2014;14:1402–1406.Suche in Google Scholar

[32] Yang N, Xiao SJ, Zhou Q, et al. [An introduction of principles and methods of applying GRADE to network meta-analysis.] Chin J Evid Based Med. 2016;16:598–603.Suche in Google Scholar

[33] Vernooij RW, Alonso-Coello P, Brouwers M, et al. Reporting Items for Updated Clinical Guidelines: Checklist for the Reporting of Updated Guidelines (CheckUp). PLoS Med. 2017;14:e1002207.10.1371/journal.pmed.1002207Suche in Google Scholar PubMed PubMed Central

[34] Aringer M, Costenbader K, Daikh D, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis. 2019;78:1151–1159.10.1136/annrheumdis-2018-214819Suche in Google Scholar PubMed

[35] Dahlström Ö, Sjöwall C. The diagnostic accuracies of the 2012 SLICC criteria and the proposed EULAR/ACR criteria for systemic lupus erythematosus classification are comparable. Lupus. 2019;28:778–782.10.1177/0961203319846388Suche in Google Scholar PubMed PubMed Central

[36] Wentian L, Ying Z, Chenghua W, et al. Utility of the ACR-1997, SLICC-2012 and EULAR/ACR-2019 classification criteria for systemic lupus erythematosus: a single-centre retrospective study. Lupus Sci Med. 2022;9:e000718.10.1136/lupus-2022-000718Suche in Google Scholar

[37] McDougall JA, Helmick CG, Lim SS, et al. Differences in the diagnosis and management of systemic lupus erythematosus by primary care and specialist providers in the American Indian/Alaska Native population. Lupus. 2018;27:1169–1176.10.1177/0961203318763529Suche in Google Scholar PubMed

[38] Hoi A, Igel T, Mok CC, et al. Systemic lupus erythematosus. Lancet. 2024;403:2326–2338.10.1016/S0140-6736(24)00398-2Suche in Google Scholar PubMed

[39] Li M, Wang Y, Zhao J, et al. Chinese SLE Treatment and Research Group (CSTAR) Registry 2009–2019: Major Clinical Characteristics of Chinese Patients with Systemic Lupus Erythematosus. Rheumatol Immunol Res. 2021;2:43–47.10.2478/rir-2021-0001Suche in Google Scholar PubMed PubMed Central

[40] Kernder A, Richter JG, Fischer-Betz R, et al. Delayed diagnosis adversely affects outcome in systemic lupus erythematosus: Cross sectional analysis of the LuLa cohort. Lupus. 2021;30:431–438.10.1177/0961203320983445Suche in Google Scholar PubMed PubMed Central

[41] Shi Y, Li M, Liu L, et al. Relationship between disease activity, organ damage and health-related quality of life in patients with systemic lupus erythematosus: A systemic review and meta-analysis. Autoimmun Rev. 2021;20:102691.10.1016/j.autrev.2020.102691Suche in Google Scholar PubMed

[42] Murimi-Worstell IB, Lin DH, Nab H, et al. Association between organ damage and mortality in systemic lupus erythematosus: a systematic review and meta-analysis. BMJ Open. 2020;10:e031850.10.1136/bmjopen-2019-031850Suche in Google Scholar PubMed PubMed Central

[43] van Vollenhoven RF, Bertsias G, Doria A, et al. 2021 DORIS definition of remission in SLE: final recommendations from an international task force. Lupus Sci Med. 2021;8:e000538.10.1136/lupus-2021-000538corr1Suche in Google Scholar PubMed PubMed Central

[44] Franklyn K, Lau CS, Navarra SV, et al. Definition and initial validation of a Lupus Low Disease Activity State (LLDAS). Ann Rheum Dis. 2016;75:1615–1621.10.1136/annrheumdis-2015-207726Suche in Google Scholar PubMed

[45] Yang Z, Cheng C, Wang Z, et al. Prevalence, Predictors, and Prognostic Benefits of Remission Achievement in Patients With Systemic Lupus Erythematosus: A Systematic Review. Arthritis Care Res (Hoboken). 2022;74:208–218.10.1002/acr.24464Suche in Google Scholar PubMed

[46] Kostopoulou M, Mukhtyar CB, Bertsias G, et al. Management of systemic lupus erythematosus: a systematic literature review informing the 2023 update of the EULAR recommendations. Ann Rheum Dis. 2024;83:1489–1501.10.1136/ard-2023-225319Suche in Google Scholar PubMed PubMed Central

[47] Pitsigavdaki S, Nikoloudaki M, Garantziotis P, et al. Pragmatic targets for moderate/severe SLE and their implications for clinical care and trial design: sustained DORIS or LLDAS for at least 6 months is sufficient while their attainment for at least 24 months ensures high specificity for damage-free progression. Ann Rheum Dis. 2024;83:464–474.10.1136/ard-2023-224919Suche in Google Scholar PubMed PubMed Central

[48] Golder V, Kandane-Rathnayake R, Li N, et al. Association of sustained lupus low disease activity state with improved outcomes in systemic lupus erythematosus: a multinational prospective cohort study. Lancet Rheumatol. 2024;6:e528–e536.10.1016/S2665-9913(24)00121-8Suche in Google Scholar PubMed

[49] Bae SC, Koh HK, Chang DK, et al. Reliability and validity of systemic lupus activity measure-revised (SLAM-R) for measuring clinical disease activity in systemic lupus erythematosus. Lupus. 2001;10:405–409.10.1191/096120301678646146Suche in Google Scholar PubMed

[50] Bombardier C, Gladman DD, Urowitz MB, et al. Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum. 1992;35:630–640.10.1002/art.1780350606Suche in Google Scholar PubMed

[51] Dafna D Gladman, Dominique Ibañez, Murray B Urowitz. Systemic lupus erythematosus disease activity index 2000. J Rheumatol. 2002;29:288–291.Suche in Google Scholar

[52] J Guzmán, M H Cardiel, A Arce-Salinas, et al. Measurement of disease activity in systemic lupus erythematosus. Prospective validation of 3 clinical indices. J Rheumatol. 1992;19:1551–1558.Suche in Google Scholar

[53] Isenberg DA, Rahman A, Allen E, et al. BILAG 2004. Development and initial validation of an updated version of the British Isles Lupus Assessment Group’s disease activity index for patients with systemic lupus erythematosus. Rheumatology (Oxford). 2005;44:902–906.10.1093/rheumatology/keh624Suche in Google Scholar PubMed

[54] Jesus D, Matos A, Henriques C, et al. Derivation and validation of the SLE Disease Activity Score (SLE-DAS): a new SLE continuous measure with high sensitivity for changes in disease activity. Ann Rheum Dis. 2019;78:365–371.10.1136/annrheumdis-2018-214502Suche in Google Scholar PubMed

[55] Liang MH, Socher SA, Larson MG, et al. Reliability and validity of six systems for the clinical assessment of disease activity in systemic lupus erythematosus. Arthritis Rheum. 1989;32:1107–1118.10.1002/anr.1780320909Suche in Google Scholar PubMed

[56] Mikdashi J, Nived O. Measuring disease activity in adults with systemic lupus erythematosus: the challenges of administrative burden and responsiveness to patient concerns in clinical research. Arthritis Res Ther. 2015;17:183.10.1186/s13075-015-0702-6Suche in Google Scholar PubMed PubMed Central

[57] Petri M, Kim MY, Kalunian KC, et al. Combined oral contraceptives in women with systemic lupus erythematosus. N Engl J Med. 2005;353:2550–2558.10.1056/NEJMoa051135Suche in Google Scholar PubMed

[58] Symmons DP, Coppock JS, Bacon PA, et al. Development and assessment of a computerized index of clinical disease activity in systemic lupus erythematosus. Members of the British Isles Lupus Assessment Group (BILAG). Q J Med. 1988;69:927–937.Suche in Google Scholar

[59] Touma Z, Gladman DD, Su J, et al. A novel lupus activity index accounting for glucocorticoids: SLEDAI-2K glucocorticoid index. Rheumatology (Oxford). 2018;57:1370–1376.10.1093/rheumatology/key103Suche in Google Scholar PubMed

[60] Keeling SO, Alabdurubalnabi Z, Avina-Zubieta A, et al. Canadian Rheumatology Association Recommendations for the Assessment and Monitoring of Systemic Lupus Erythematosus. J Rheumatol. 2018;45:1426–1439.10.3899/jrheum.171459Suche in Google Scholar PubMed

[61] América G Uribe, Luis M Vilá, Gerald McGwin Jr, et al. The Systemic Lupus Activity Measure-revised, the Mexican Systemic Lupus Erythematosus Disease Activity Index (SLE-DAI), and a modified SLEDAI-2K are adequate instruments to measure disease activity in systemic lupus erythematosus. J Rheumatol. 2004;31:1934–1940.Suche in Google Scholar

[62] Touma Z, Urowitz MB, Ibañez D, et al. SLEDAI-2K 10 days versus SLEDAI-2K 30 days in a longitudinal evaluation. Lupus. 2011;20:67–70.10.1177/0961203310385163Suche in Google Scholar PubMed

[63] Keeling SO, Vandermeer B, Medina J, et al. Measuring Disease Activity and Damage with Validated Metrics: A Systematic Review on Mortality and Damage in Systemic Lupus Erythematosus. J Rheumatol. 2018;45:1448–1461.10.3899/jrheum.171310Suche in Google Scholar PubMed

[64] Jesus D, Larosa M, Henriques C, et al. Systemic Lupus Erythematosus Disease Activity Score (SLE-DAS) enables accurate and user-friendly definitions of clinical remission and categories of disease activity. Ann Rheum Dis. 2021;80:1568–1574.10.1136/annrheumdis-2021-220363Suche in Google Scholar PubMed

[65] Gladman D, Ginzler E, Goldsmith C, et al. The development and initial validation of the Systemic Lupus International Collaborating Clinics/American College of Rheumatology damage index for systemic lupus erythematosus. Arthritis Rheum. 1996;39:363–369.10.1002/art.1780390303Suche in Google Scholar PubMed

[66] Trujillo-Martín MM, Rúa-Figueroa Fernández de Larrinoa I, Ruíz-Irastorza G, et al. [Clinical practice guidelines for systemic lupus erythematosus: Recommendations for general clinical management]. Med Clin (Barc). 2016;146:413.e1-14.10.1016/j.medcle.2016.06.033Suche in Google Scholar

[67] Chinese SLE Treatment and Research Group. [Expert Consensus on the Rational Use of Glucocorticoids in Patients With Systemic Lupus Erythematosus.] Zhong Hua Nei Ke Za Zhi. 2014;53:502–504.Suche in Google Scholar

[68] Fanouriakis A, Kostopoulou M, Alunno A, et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann Rheum Dis. 2019;78:736–745.10.1136/annrheumdis-2019-215089Suche in Google Scholar PubMed

[69] Floris A, Chessa E, Sebastiani GD, et al. Glucocorticoid tapering and associated outcome in patients with newly diagnosed systemic lupus erythematosus: the real-world GULP prospective observational study. RMD Open. 2022;8:e002701.10.1136/rmdopen-2022-002701Suche in Google Scholar PubMed PubMed Central

[70] Katsumata Y, Inoue E, Harigai M, et al. Risk of flare and damage accrual after tapering glucocorticoids in modified serologically active clinically quiescent patients with systemic lupus erythematosus: a multinational observational cohort study. Ann Rheum Dis. 2024;83:998–1005.10.1136/ard-2023-225369Suche in Google Scholar PubMed

[71] Ji L, Xie W, Zhang Z. Low-dose glucocorticoids should be withdrawn or continued in systemic lupus erythematosus? A systematic review and meta-analysis on risk of flare and damage accrual. Rheumatology (Oxford). 2021;60:5517–5526.10.1093/rheumatology/keab149Suche in Google Scholar PubMed

[72] Sarnes E, Crofford L, Watson M, et al. Incidence and US costs of corticosteroid-associated adverse events: a systematic literature review. Clin Ther. 2011;33:1413–1432.10.1016/j.clinthera.2011.09.009Suche in Google Scholar PubMed

[73] Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, et al. Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review. Ann Rheum Dis. 2010;69:20–28.10.1136/ard.2008.101766Suche in Google Scholar PubMed

[74] Wang F, Zhang W, Wang S, et al. Protective effects of antimalarials in Chinese patients with systemic lupus erythematosus. Ann Rheum Dis. 2019;78:e80.10.1136/annrheumdis-2018-213819Suche in Google Scholar PubMed

[75] Babary H, Liu X, Ayatollahi Y, et al. Favorable effects of hydroxychloroquine on serum low density lipid in patients with systemic lupus erythematosus: A systematic review and meta-analysis. Int J Rheum Dis. 2018;21:84–92.10.1111/1756-185X.13159Suche in Google Scholar PubMed

[76] Jorge AM, Mancini C, Zhou B, et al. Hydroxychloroquine Dose per Ophthalmology Guidelines and the Risk of Systemic Lupus Erythematosus Flares. JAMA. 2022;328:1458–1460.10.1001/jama.2022.13591Suche in Google Scholar PubMed PubMed Central

[77] Almeida-Brasil CC, Hanly JG, Urowitz M, et al. Flares after hydroxychloroquine reduction or discontinuation: results from the Systemic Lupus International Collaborating Clinics (SLICC) inception cohort. Ann Rheum Dis. 2022;81:370–378.10.1136/annrheumdis-2021-221295Suche in Google Scholar PubMed PubMed Central

[78] Melles RB, Marmor MF. The risk of toxic retinopathy in patients on long-term hydroxychloroquine therapy. JAMA Ophthalmol. 2014;132:1453–1460.10.1001/jamaophthalmol.2014.3459Suche in Google Scholar PubMed

[79] Mukwikwi ER, Pineau CA, Vinet E, et al. Retinal Complications in Patients with Systemic Lupus Erythematosus Treated with Antimalarial Drugs. J Rheumatol. 2020;47:553–556.10.3899/jrheum.181102Suche in Google Scholar PubMed

[80] Huang X, Lin HE, Zheng R, et al. Literature Review of Hydroxychloroquine-induced Ocular Toxicity. Chinese J Pharmacoepidemiol. 2019;28:59–63.Suche in Google Scholar

[81] Marmor MF, Kellner U, Lai TY, et al. Recommendations on Screening for Chloroquine and Hydroxychloroquine Retinopathy (2016 Revision). Ophthalmology. 2016;123:1386–1394.10.1016/j.ophtha.2016.01.058Suche in Google Scholar PubMed

[82] Yusuf IH, Foot B, Galloway J, et al. The Royal College of Ophthalmologists recommendations on screening for hydroxychloroquine and chloroquine users in the United Kingdom: executive summary. Eye (Lond). 2018;32:1168–1173.10.1038/s41433-018-0136-xSuche in Google Scholar PubMed PubMed Central

[83] Hooks M, Bart B, Vardeny O, et al. Effects of hydroxychloroquine treatment on QT interval. Heart Rhythm. 2020;17:1930–1935.10.1016/j.hrthm.2020.06.029Suche in Google Scholar PubMed PubMed Central

[84] Mercuro NJ, Yen CF, Shim DJ, et al. Risk of QT Interval Prolongation Associated With Use of Hydroxychloroquine With or Without Concomitant Azithromycin Among Hospitalized Patients Testing Positive for Coronavirus Disease 2019 (CO-VID-19). JAMA Cardiol. 2020;5:1036–1041.10.1001/jamacardio.2020.1834Suche in Google Scholar PubMed PubMed Central

[85] Nguyen LS, Dolladille C, Drici MD, et al. Cardiovascular Toxicities Associated With Hydroxychloroquine and Azithromycin: An Analysis of the World Health Organization Pharmacovigilance Database. Circulation. 2020;142:303–305.10.1161/CIRCULATIONAHA.120.048238Suche in Google Scholar PubMed PubMed Central

[86] Chatre C, Roubille F, Vernhet H, et al. Cardiac Complications Attributed to Chloroquine and Hydroxychloroquine: A Systematic Review of the Literature. Drug Saf. 2018;41:919–931.10.1007/s40264-018-0689-4Suche in Google Scholar PubMed

[87] Desmarais J, Rosenbaum JT, Costenbader KH, et al. American College of Rheumatology White Paper on Antimalarial Cardiac Toxicity. Arthritis Rheumatol. 2021;73:2151–2160.10.1002/art.41934Suche in Google Scholar PubMed

[88] Goldman A, Bomze D, Dankner R, et al. Cardiovascular adverse events associated with hydroxychloroquine and chloroquine: A comprehensive pharmacovigilance analysis of pre-COVID-19 reports. Br J Clin Pharmacol. 2021;87:1432–1442.10.1111/bcp.14546Suche in Google Scholar PubMed

[89] Yogasundaram H, Putko BN, Tien J, et al. Hydroxychloroquine-induced cardiomyopathy: case report, pathophysiology, diagnosis, and treatment. Can J Cardiol. 2014;30:1706–1715.10.1016/j.cjca.2014.08.016Suche in Google Scholar PubMed

[90] Pego-Reigosa JM, Cobo-Ibáñez T, Calvo-Alén J, et al. Efficacy and safety of nonbiologic immunosuppressants in the treatment of nonrenal systemic lupus erythematosus: a systematic review. Arthritis Care Res (Hoboken). 2013;65:1775–1785.10.1002/acr.22035Suche in Google Scholar PubMed

[91] Tselios K, Gladman DD, Su J, et al. Mycophenolate Mofetil in Nonrenal Manifestations of Systemic Lupus Erythematosus: An Observational Cohort Study. J Rheumatol. 2016;43:552–558.10.3899/jrheum.150779Suche in Google Scholar PubMed

[92] Choi CB, Won S, Bae SC. Outcomes of multitarget therapy using mycophenolate mofetil and tacrolimus for refractory or relapsing lupus nephritis. Lupus. 2018;27:1007–1011.10.1177/0961203318758505Suche in Google Scholar PubMed

[93] Keyes E, Jobanputra A, Feng R, et al. Comparative responsiveness of cutaneous lupus erythematosus patients to methotrexate and mycophenolate mofetil: A cohort study. J Am Acad Dermatol. 2022;87:447–448.10.1016/j.jaad.2021.09.017Suche in Google Scholar PubMed PubMed Central

[94] Chasset F, Tounsi T, Cesbron E, et al. Efficacy and tolerance profile of thalidomide in cutaneous lupus erythematosus: A systematic review and meta-analysis. J Am Acad Dermatol. 2018;78:342–350.10.1016/j.jaad.2017.09.059Suche in Google Scholar PubMed

[95] Aitmehdi R, Arnaud L, Francès C, et al. Long-term efficacy and safety outcomes of lenalidomide for cutaneous lupus erythematosus: A multicenter retrospective observational study of 40 patients. J Am Acad Dermatol. 2021;84:1171–1174.10.1016/j.jaad.2020.11.014Suche in Google Scholar PubMed

[96] Jiang N, Jin S, Yu C, et al. Efficacy and safety of immunosuppressive agents for adults with lupus nephritis: a systematic review and network meta-analysis. Front Immunol. 2023;14:1232244.10.3389/fimmu.2023.1232244Suche in Google Scholar PubMed PubMed Central

[97] Palmer SC, Tunnicliffe DJ, Singh-Grewal D, et al. Induction and Maintenance Immunosuppression Treatment of Proliferative Lupus Nephritis: A Network Meta-analysis of Randomized Trials. Am J Kidney Dis. 2017;70:324–336.10.1053/j.ajkd.2016.12.008Suche in Google Scholar PubMed

[98] Tang KT, Tseng CH, Hsieh TY, et al. Induction therapy for membranous lupus nephritis: a systematic review and network meta-analysis. Int J Rheum Dis. 2018;21:1163–1172.10.1111/1756-185X.13321Suche in Google Scholar PubMed

[99] Fernandez D, Bonilla E, Mirza N, et al. Rapamycin reduces disease activity and normalizes T cell activation-induced calcium fluxing in patients with systemic lupus erythematosus. Arthritis Rheum. 2006;54:2983–2988.10.1002/art.22085Suche in Google Scholar PubMed PubMed Central

[100] Lai ZW, Kelly R, Winans T, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet. 2018;391:1186–1196.10.1016/S0140-6736(18)30485-9Suche in Google Scholar PubMed PubMed Central

[101] Peng L, Wu C, Hong R, et al. Clinical efficacy and safety of sirolimus in systemic lupus erythematosus: a real-world study and meta-analysis. Ther Adv Musculoskelet Dis. 2020;12:1759720.10.1177/1759720X20953336Suche in Google Scholar PubMed PubMed Central

[102] Jiang N, Li M, Zhang H, et al. Sirolimus versus tacrolimus for systemic lupus erythematosus treatment: results from a real-world CSTAR cohort study. Lupus Sci Med. 2022;9:e000617.10.1136/lupus-2021-000617Suche in Google Scholar PubMed PubMed Central

[103] Ji L, Xie W, Zhang Z. Efficacy and safety of sirolimus in patients with systemic lupus erythematosus: A systematic review and meta-analysis. Semin Arthritis Rheum. 2020;50:1073–1080.10.1016/j.semarthrit.2020.07.006Suche in Google Scholar PubMed

[104] Yap DYH, Tang C, Chan GCW, et al. Longterm Data on Sirolimus Treatment in Patients with Lupus Nephritis. J Rheumatol. 2018;45:1663–1670.10.3899/jrheum.180507Suche in Google Scholar PubMed

[105] Piranavan P, Perl A. Improvement of renal and non-renal SLE outcome measures on sirolimus therapy - A 21-year follow-up study of 73 patients. Clin Immunol. 2021;229:108781.10.1016/j.clim.2021.108781Suche in Google Scholar PubMed PubMed Central

[106] Wu C, Wang Q, Xu D, et al. Sirolimus for patients with connective tissue disease-related refractory thrombocytopenia: a single-arm, open-label clinical trial. Rheumatology (Oxford). 2021;60:2629–2634.10.1093/rheumatology/keaa645Suche in Google Scholar PubMed

[107] Du H, Su W, Su J, et al. Sirolimus for the treatment of patients with refractory connective tissue disease-related thrombocytopenia: a pilot study. Rheumatology (Oxford). 2024;63:79–84.10.1093/rheumatology/kead160Suche in Google Scholar PubMed

[108] Canaud G, Bienaimé F, Tabarin F, et al. Inhibition of the mTORC pathway in the antiphospholipid syndrome. N Engl J Med. 2014;371:303–312.10.1056/NEJMoa1312890Suche in Google Scholar PubMed

[109] Lee YH, Song GG. Comparative efficacy and safety of tacrolimus, mycophenolate mofetil, azathioprine, and cyclophosphamide as maintenance therapy for lupus nephritis : A Bayesian network meta-analysis of randomized controlled trials. Z Rheumatol. 2017;76:904–912.10.1007/s00393-016-0186-zSuche in Google Scholar PubMed

[110] Papachristos DA, Oon S, Hanly JG, et al. Management of inflammatory neurologic and psychiatric manifestations of systemic lupus erythematosus: A systematic review. Semin Arthritis Rheum. 2021;51:49–71.10.1016/j.semarthrit.2020.12.004Suche in Google Scholar PubMed

[111] You Y, Zhou Z, Wang F, et al. Mycophenolate Mofetil and New-Onset Systemic Lupus Erythematosus: A Randomized Clinical Trial. JAMA Netw Open. 2024;7:e2432131.10.1001/jamanetworkopen.2024.32131Suche in Google Scholar PubMed PubMed Central

[112] Mok CC. Mycophenolate mofetil for non-renal manifestations of systemic lupus erythematosus: a systematic review. Scand J Rheumatol. 2007;36:329–337.10.1080/03009740701607042Suche in Google Scholar PubMed

[113] Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, et al. Entericcoated mycophenolate sodium versus azathioprine in patients with active systemic lupus erythematosus: a randomised clinical trial. Ann Rheum Dis. 2017;76:1575–1582.10.1136/annrheumdis-2016-210882Suche in Google Scholar PubMed

[114] Fernandes Moça Trevisani V, Castro AA, Ferreira Neves Neto J, et al. Cyclophosphamide versus methylprednisolone for treating neuropsychiatric involvement in systemic lupus erythematosus. Cochrane Database Syst Rev. 2013;201:3.10.1002/14651858.CD002265.pub3Suche in Google Scholar PubMed PubMed Central

[115] Blumenfeld Z, Mischari O, Schultz N, et al. Gonadotropin releasing hormone agonists may minimize cyclophosphamide associated gonadotoxicity in SLE and autoimmune diseases. Semin Arthritis Rheum. 2011;41:346–352.10.1016/j.semarthrit.2011.05.008Suche in Google Scholar PubMed

[116] Marder W, McCune WJ, Wang L, et al. Adjunctive GnRH-a treatment attenuates depletion of ovarian reserve associated with cyclophosphamide therapy in premenopausal SLE patients. Gynecol Endocrinol. 2012;28:624–627.10.3109/09513590.2011.650752Suche in Google Scholar PubMed PubMed Central

[117] Gong JY, Peng SY, Xing K, et al. Evaluating the role of GSTP1 genetic polymorphism (rs1695, 313A>G) as a predictor in cyclophosphamide-induced toxicities. Medicine (Baltimore). 2021;100:e24423.10.1097/MD.0000000000024423Suche in Google Scholar PubMed PubMed Central

[118] Cao H, Rao Y, Liu L, et al. The Efficacy and Safety of Leflunomide for the Treatment of Lupus Nephritis in Chinese Patients: Systematic Review and Meta-Analysis. PLoS One. 2015;10:e0144548.10.1371/journal.pone.0144548Suche in Google Scholar PubMed PubMed Central

[119] Zhang M, Qi C, Zha Y, et al. Leflunomide versus cyclophosphamide in the induction treatment of proliferative lupus nephritis in Chinese patients: a randomized trial. Clin Rheumatol. 2019;38:859–867.10.1007/s10067-018-4348-zSuche in Google Scholar PubMed

[120] Fu Q, Wu C, Dai M, et al. Leflunomide versus azathioprine for maintenance therapy of lupus nephritis: a prospective, multi-centre, randomised trial and long-term follow-up. Ann Rheum Dis. 2022;81:1549–1555.10.1136/ard-2022-222486Suche in Google Scholar PubMed PubMed Central

[121] Østensen M, Khamashta M, Lockshin M, et al. Anti-inflammatory and immunosuppressive drugs and reproduction. Arthritis Res Ther. 2006;8:209.10.1186/ar1957Suche in Google Scholar PubMed PubMed Central

[122] Hannon CW, McCourt C, Lima HC, et al. Interventions for cutaneous disease in systemic lupus erythematosus. Cochrane Database Syst Rev. 2021;3:CD007478.10.1002/14651858.CD007478.pub2Suche in Google Scholar PubMed PubMed Central

[123] Sakthiswary R, Suresh E. Methotrexate in systemic lupus erythematosus: a systematic review of its efficacy. Lupus. 2014;23:225–235.10.1177/0961203313519159Suche in Google Scholar PubMed

[124] Yap DY, Ma MK, Mok MM, et al. Long-term data on tacrolimus treatment in lupus nephritis. Rheumatology (Oxford). 2014;53:2232–2237.10.1093/rheumatology/keu265Suche in Google Scholar PubMed

[125] Mok CC, To CH, Yu KL, et al. Combined low-dose mycophenolate mofetil and tacrolimus for lupus nephritis with suboptimal response to standard therapy: a 12-month prospective study. Lupus. 2013;22:1135–1141.10.1177/0961203313502864Suche in Google Scholar PubMed

[126] Li Y, Feng X. Efficacy and safety of tacrolimus in systemic lupus erythematosus patients with refractory thrombocytopenia: a retrospective study. Lupus. 2018;27:60–65.10.1177/0961203317711011Suche in Google Scholar PubMed

[127] Zheng Z, Zhang H, Peng X, et al. Effect of Tacrolimus vs Intravenous Cyclophosphamide on Complete or Partial Response in Patients With Lupus Nephritis: A Randomized Clinical Trial. JAMA Netw Open. 2022;5:e224492.10.1001/jamanetworkopen.2022.4492Suche in Google Scholar PubMed PubMed Central

[128] Singh JA, Hossain A, Kotb A, et al. Risk of serious infections with immunosuppressive drugs and glucocorticoids for lupus nephritis: a systematic review and network meta-analysis. BMC Med. 2016;14:137.10.1186/s12916-016-0673-8Suche in Google Scholar PubMed PubMed Central

[129] Tarter L, Bermas BL. Expert Perspective on a Clinical Challenge: Lupus and Pregnancy. Arthritis Rheumatol. 2024;76:321–331.10.1002/art.42756Suche in Google Scholar PubMed

[130] Jiang Y, Tao M, Chen J, et al. Calcineurin inhibitors in the treatment of systemic lupus erythematosus during pregnancy: A narrative review with emphasis on efficacy and safety. Eur J Obstet Gynecol Reprod Biol. 2024;294:148–155.10.1016/j.ejogrb.2023.12.039Suche in Google Scholar PubMed

[131] Chen W, Tang X, Liu Q, et al. Short-term outcomes of induction therapy with tacrolimus versus cyclophosphamide for active lupus nephritis: A multicenter randomized clinical trial. Am J Kidney Dis. 2011;57:235–244.10.1053/j.ajkd.2010.08.036Suche in Google Scholar PubMed

[132] Jesus D, Rodrigues M, da Silva JAP, et al. Multitarget therapy of mycophenolate mofetil and cyclosporine A for induction treatment of refractory lupus nephritis. Lupus. 2018;27:1358–1362.10.1177/0961203318758508Suche in Google Scholar PubMed

[133] Moroni G, Doria A, Mosca M, et al. A randomized pilot trial comparing cyclosporine and azathioprine for maintenance therapy in diffuse lupus nephritis over four years. Clin J Am Soc Nephrol. 2006;1:925–932.10.2215/CJN.02271205Suche in Google Scholar PubMed

[134] Yang TH, Wu TH, Chang YL, et al. Cyclosporine for the treatment of lupus nephritis in patients with systemic lupus erythematosus. Clin Nephrol. 2018;89:277–285.10.5414/CN109325Suche in Google Scholar PubMed

[135] Young Ho Lee, Gwan Gyu Song. Comparative Efficacy and Safety of Tacrolimus, Cyclosporin A, Mycophenolate Mofetil, Cyclophosphamide, and Corticosteroids as Induction Therapy for Membranous Lupus Nephritis: A Network Meta-Analysis. Pharmacology. 2022;107:439–445.10.1159/000525066Suche in Google Scholar PubMed

[136] Furie R, Petri M, Zamani O, et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63:3918–3930.10.1002/art.30613Suche in Google Scholar PubMed PubMed Central

[137] Navarra SV, Guzmán RM, Gallacher AE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377:721–731.10.1016/S0140-6736(10)61354-2Suche in Google Scholar PubMed

[138] Zhang F, Bae SC, Bass D, et al. A pivotal phase III, randomised, placebo-controlled study of belimumab in patients with systemic lupus erythematosus located in China, Japan and South Korea. Ann Rheum Dis. 2018;77:355–363.10.1136/annrheumdis-2017-211631Suche in Google Scholar PubMed PubMed Central

[139] Furie R, Rovin BH, Houssiau F, et al. Two-Year, Randomized, Controlled Trial of Belimumab in Lupus Nephritis. N Engl J Med. 2020;383:1117–1128.10.1056/NEJMoa2001180Suche in Google Scholar PubMed

[140] Rovin BH, Furie R, Teng YKO, et al. A secondary analysis of the Belimumab International Study in Lupus Nephritis trial examined effects of belimumab on kidney outcomes and preservation of kidney function in patients with lupus nephritis. Kidney Int. 2022;101:403–413.10.1016/j.kint.2021.08.027Suche in Google Scholar PubMed

[141] Collins CE, Cortes-Hernández J, Garcia MA, et al. Real-World Effectiveness of Belimumab in the Treatment of Systemic Lupus Erythematosus: Pooled Analysis of Multi-Country Data from the OBSErve Studies. Rheumatol Ther. 2020;7:949–965.10.1007/s40744-020-00243-2Suche in Google Scholar PubMed PubMed Central

[142] Gatto M, Saccon F, Andreoli L, et al. Durable renal response and safety with add-on belimumab in patients with lupus nephritis in real-life setting (BeRLiSS-LN). Results from a large, nationwide, multicentric cohort. J Autoimmun. 2021;124:102729.10.1016/j.jaut.2021.102729Suche in Google Scholar PubMed

[143] Zen M, Gatto M, Depascale R, et al. Early and Late Response and Glucocorticoid-Sparing Effect of Belimumab in Patients with Systemic Lupus Erythematosus with Joint and Skin Manifestations: Results from the Belimumab in Real Life Setting Study-Joint and Skin (BeRLiSS-JS). J Pers Med. 2023;13:691.10.3390/jpm13040691Suche in Google Scholar PubMed PubMed Central

[144] Kidney Disease: Improving Global Outcomes (KDIGO) Lupus Nephritis Work Group. KDIGO 2024 Clinical Practice Guideline for the management of LUPUS NEPHRITIS. Kidney Int. 2024;105:S1-S69.10.1016/j.kint.2023.09.002Suche in Google Scholar PubMed

[145] Wu D, Li J, Xu D, et al. Telitacicept in patients with active systemic lupus erythematosus: results of a phase 2b, randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2024;83:475–487.10.1136/ard-2023-224854Suche in Google Scholar PubMed PubMed Central

[146] WU D, LI J, XU D, et al. Telitacicept, a Human Recombinant Fusion Protein Targeting B Lymphocyte Stimulator (BlyS) and a Proliferation-Inducing Ligand (APRIL), in Systemic Lupus Erythematosus (SLE): Results of a Phase 3 Study. ACR Convergence 2022; 2022. Available at: https://acrabstracts.org/abstract/telitacicept-a-human-recombinant-fusion-protein-targeting-b-lymphocyte-stimulator-blys-and-a-proliferation-inducing-ligand-april-in-systemic-lupus-erythematosus-sle-resultsof-a-phase-3-study/. Accessed on Aug 15, 2025.Suche in Google Scholar

[147] Chen R, Fu R, Lin Z, et al. The efficacy and safety of telitacicept for the treatment of systemic lupus erythematosus: a real life observational study. Lupus. 2023;32:94–100.10.1177/09612033221141253Suche in Google Scholar PubMed

[148] Cheng J, Peng Y, Wu Q, et al. Eficacy and safety of telitacicept therapy in systemic lupus erythematosus with hematological involvement. Clin Rheumatol. 2024;43:2229–2236.10.1007/s10067-024-06992-7Suche in Google Scholar PubMed

[149] Huang X, Lin F, Chen H. Eficacy and safety of telitacicept in patients with lupus nephritis: a single-center, real-world retrospective study. Clin Exp Nephrol. 2024;28:902–909.10.1007/s10157-024-02501-xSuche in Google Scholar PubMed

[150] Merrill JT, Neuwelt CM, Wallace DJ, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010;62:222–233.10.1002/art.27233Suche in Google Scholar PubMed PubMed Central

[151] Rovin BH, Furie R, Latinis K, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012;64:1215–1226.10.1002/art.34359Suche in Google Scholar PubMed

[152] Weidenbusch M, Römmele C, Schröttle A, et al. Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol Dial Transplant. 2013;28:106–111.10.1093/ndt/gfs285Suche in Google Scholar PubMed

[153] Alshaiki F, Obaid E, Almuallim A, et al. Outcomes of rituximab therapy in refractory lupus: A meta-analysis. Eur J Rheumatol. 2018;5:118–126.10.5152/eurjrheum.2018.17096Suche in Google Scholar PubMed PubMed Central

[154] Zang H, Yang N, Lu J, et al. [Recommendations for the diagnosis and management of lupus nephritis in China.] Zhong Hua Nei Ke Za Zhi. 2021; 60::784–790.Suche in Google Scholar

[155] Tokunaga M, Saito K, Kawabata D, et al. Efficacy of rituximab (anti-CD20) for refractory systemic lupus erythematosus involving the central nervous system. Ann Rheum Dis. 2007;66:470–475.10.1136/ard.2006.057885Suche in Google Scholar PubMed PubMed Central

[156] Serris A, Amoura Z, Canouï-Poitrine F, et al. Efficacy and safety of rituximab for systemic lupus erythematosus-associated immune cytopenias: A multicenter retrospective cohort study of 71 adults. Am J Hematol. 2018;93:424–429.10.1002/ajh.24999Suche in Google Scholar PubMed

[157] Chugh S, Darvish-Kazem S, Lim W, et al. Rituximab plus standard of care for treatment of primary immune thrombocytopenia: a systematic review and meta-analysis. Lancet Haematol. 2015;2:e75–81.10.1016/S2352-3026(15)00003-4Suche in Google Scholar PubMed

[158] Furie RA, Aroca G, Cascino MD, et al. B-cell depletion with obinutuzumab for the treatment of proliferative lupus nephritis: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2022;81:100–107.10.1136/annrheumdis-2021-220920Suche in Google Scholar PubMed PubMed Central

[159] Furie RA, Rovin BH, Garg JP, et al. Efficacy and Safety of Obinutuzumab in Active Lupus Nephritis. N Engl J Med. 2025;392:1471–1483.10.1056/NEJMoa2410965Suche in Google Scholar PubMed

[160] Arnold J, Dass S, Twigg S, et al. Efficacy and safety of obinutuzumab in systemic lupus erythematosus patients with secondary non-response to rituximab. Rheumatology (Oxford). 2022;61:4905–4909.10.1093/rheumatology/keac150Suche in Google Scholar PubMed

[161] Furie R, Khamashta M, Merrill JT, et al. Anifrolumab, an Anti-Interferon-α Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus. Arthritis Rheumatol. 2017;69:376–386.10.1002/art.39962Suche in Google Scholar PubMed PubMed Central

[162] Morand EF, Trasieva T, Berglind A, et al. Lupus Low Disease Activity State (LLDAS) attainment discriminates responders in a systemic lupus erythematosus trial: post-hoc analysis of the Phase IIb MUSE trial of anifrolumab. Ann Rheum Dis. 2018;77:706–713.10.1136/annrheumdis-2017-212504Suche in Google Scholar PubMed PubMed Central

[163] Furie RA, Morand EF, Bruce IN, et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TU-LIP-1): a randomised, controlled, phase 3 trial. Lancet Rheumatol. 2019;1:e208–e219.10.1016/S2665-9913(19)30076-1Suche in Google Scholar PubMed

[164] Morand EF, Furie R, Tanaka Y, et al. Trial of Anifrolumab in Active Systemic Lupus Erythematosus. N Engl J Med. 2020;382:211–221.10.1056/NEJMoa1912196Suche in Google Scholar PubMed

[165] Jayne D, Rovin B, Mysler EF, et al. Phase II randomised trial of type I interferon inhibitor anifrolumab in patients with active lupus nephritis. Ann Rheum Dis. 2022;81:496–506.10.1136/annrheumdis-2021-221478Suche in Google Scholar PubMed PubMed Central

[166] Bermea RS, Sharma N, Cohen K, et al. Use of Eculizumab in Atypical Hemolytic Uremic Syndrome, Complicating Systemic Lupus Erythematosus. J Clin Rheumatol. 2016;22:320–323.10.1097/RHU.0000000000000423Suche in Google Scholar PubMed

[167] de Holanda MI, Pôrto LC, Wagner T, et al. Use of eculizumab in a systemic lupus erythemathosus patient presenting thrombotic microangiopathy and heterozygous deletion in CFHR1-CFHR3. A case report and systematic review. Clin Rheumatol. 2017;36:2859–2867.10.1007/s10067-017-3823-2Suche in Google Scholar PubMed

[168] Sciascia S, Radin M, Yazdany J, et al. Expanding the therapeutic options for renal involvement in lupus: eculizumab, available evidence. Rheumatol Int. 2017;37:1249–1255.10.1007/s00296-017-3686-5Suche in Google Scholar PubMed

[169] Kello N, Khoury LE, Marder G, et al. Secondary thrombotic microangiopathy in systemic lupus erythematosus and antiphospholipid syndrome, the role of complement and use of eculizumab: Case series and review of literature. Semin Arthritis Rheum. 2019;49:74–83.10.1016/j.semarthrit.2018.11.005Suche in Google Scholar PubMed

[170] Marinho A, Delgado Alves J, Fortuna J, et al. Biological therapy in systemic lupus erythematosus, antiphospholipid syndrome, and Sjögren’s syndrome: evidence- and practice-based guidance. Front Immunol. 2023;14:1117699.10.3389/fimmu.2023.1117699Suche in Google Scholar PubMed PubMed Central

[171] Smith J, Hans V, Yacyshyn E, et al. Systemic lupus erythematosus presenting with atypical hemolytic uremic syndrome: a case report and review of the literature. Rheumatol Int. 2024;44:2213–2225.10.1007/s00296-024-05558-9Suche in Google Scholar PubMed

[172] Wright RD, Bannerman F, Beresford MW, et al. A systematic review of the role of eculizumab in systemic lupus erythematosus-associated thrombotic microangiopathy. BMC Nephrol. 2020;21:245.10.1186/s12882-020-01888-5Suche in Google Scholar PubMed PubMed Central

[173] Weening JJ, D’Agati VD, Schwartz MM, et al. The classification of glomerulonephritis in systemic lupus erythematosus revisited. Kidney Int. 2004;65:521–530.10.1111/j.1523-1755.2004.00443.xSuche in Google Scholar PubMed

[174] Bajema IM, Wilhelmus S, Alpers CE, et al. Revision of the International Society of Nephrology/Renal Pathology Society classification for lupus nephritis: clarification of definitions, and modified National Institutes of Health activity and chronicity indices. Kidney Int. 2018;93:789–796.10.1016/j.kint.2017.11.023Suche in Google Scholar PubMed

[175] Hu WX, Chen YH, Bao H, et al. Glucocorticoid with or without additional immunosuppressant therapy for patients with lupus podocytopathy: a retrospective single-center study. Lupus. 2015;24:1067–1075.10.1177/0961203315578766Suche in Google Scholar PubMed

[176] Liu Z, Zhang H, Liu Z, et al. Multitarget therapy for induction treatment of lupus nephritis: a randomized trial. Ann Intern Med. 2015;162:18–26.10.7326/M14-1030Suche in Google Scholar PubMed

[177] Zhang H, Liu Z, Zhou M, et al. Multitarget Therapy for Maintenance Treatment of Lupus Nephritis. J Am Soc Nephrol. 2017;28:3671–3678.10.1681/ASN.2017030263Suche in Google Scholar PubMed PubMed Central

[178] Deng J, Luo L, Zhu L, et al. Multitarget therapy versus intravenous cyclophosphamide in the induction treatment of lupus nephritis: a metaanalysis of randomized controlled trials. Turk J Med Sci. 2018;48:901–910.10.3906/sag-1804-57Suche in Google Scholar PubMed

[179] Yu X, Chen N, Xue J, et al. Efficacy and Safety of Belimum-ab in Patients With Lupus Nephritis: Subgroup Analyses of a Phase 3 Randomized Trial in the East Asian Population. Am J Kidney Dis. 2023;81:294–306.10.1053/j.ajkd.2022.06.013Suche in Google Scholar PubMed

[180] Anders HJ, Furie R, Malvar A, et al. Effect of belimumab on kidney-related outcomes in patients with lupus nephritis: post hoc subgroup analyses of the phase 3 BLISS-LN trial. Nephrol Dial Transplant. 2023;38:2733–2742.10.1093/ndt/gfad167Suche in Google Scholar PubMed PubMed Central

[181] The EMPA-KIDNEY Collaborative Group; Herrington WG, Staplin N, Wanner C, et al. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388:117–127.10.1056/NEJMoa2204233Suche in Google Scholar PubMed PubMed Central

[182] Lichtnekert J, Anders HJ. Lupus nephritis-related chronic kidney disease. Nat Rev Rheumatol. 2024;20:699–711.10.1038/s41584-024-01158-wSuche in Google Scholar PubMed

[183] Rodeghiero F, Stasi R, Gernsheimer T, et al. Standardization of terminology, definitions and outcome criteria in immune thrombocytopenic purpura of adults and children: report from an international working group. Blood. 2009;113:2386–2393.10.1182/blood-2008-07-162503Suche in Google Scholar PubMed

[184] Mehdi Khellaf, Marc Michel, Annette Schaeffer, et al. Assessment of a therapeutic strategy for adults with severe autoimmune thrombocytopenic purpura based on a bleeding score rather than platelet count. Haematologica. 2005;90:829–832.Suche in Google Scholar

[185] Cooper N, Ghanima W. Immune Thrombocytopenia. N Engl J Med,. 2019;381:945–955.10.1056/NEJMcp1810479Suche in Google Scholar PubMed

[186] Su J, Xu M, Dong Z, et al. Efficacy and safety of azathioprine plus prednisone versus prednisone alone as first-line treatment for antinuclear antibody-positive immune thrombocytopenia: a retrospective cohort study. Hematology. 2023;28:2196864.10.1080/16078454.2023.2196864Suche in Google Scholar PubMed

[187] Wei Y, Ji XB, Wang YW, et al. High-dose dexamethasone vs prednisone for treatment of adult immune thrombocytopenia: a prospective multicenter randomized trial. Blood. 2016;127:296–370.10.1182/blood-2015-07-659656Suche in Google Scholar PubMed

[188] Ren X, Zhang M, Zhang X, et al. Can low-dose intravenous immunoglobulin be an alternative to high-dose intravenous immunoglobulin in the treatment of children with newly diagnosed immune thrombocytopenia: a systematic review and meta-analysis. BMC Pediatr. 2024;24:199.10.1186/s12887-024-04677-3Suche in Google Scholar PubMed PubMed Central

[189] Cimé-Aké E, Barrera-Vargas A, Demichelis-Gómez R, et al. Description of therapeutic strategies in severe systemic lupus erythematosus-associated immune thrombocytopenia: a retrospective cohort study of response and relapse. Clin Rheumatol. 2024;43:2521–2532.10.1007/s10067-024-07031-1Suche in Google Scholar PubMed

[190] Bradbury CA, Pell J, Hill Q, et al. Mycophenolate Mofetil for First-Line Treatment of Immune Thrombocytopenia. N Engl J Med. 2021;385:885–895.10.1056/NEJMoa2100596Suche in Google Scholar PubMed

[191] Sun F, Chen J, Wu W, et al. Rituximab or cyclosporin in refractory immune thrombocytopenia secondary to connective tissue diseases: a real-world observational retrospective study. Clin Rheumatol. 2020;39:3099–3104.10.1007/s10067-020-05152-xSuche in Google Scholar PubMed

[192] Zhou H, Xu M, Qin P, et al. A multicenter randomized open-label study of rituximab plus rhTPO vs rituximab in corticosteroid-resistant or relapsed ITP. Blood. 2015;125:1541–1547.10.1182/blood-2014-06-581868Suche in Google Scholar PubMed PubMed Central

[193] Barron N, Arenas-Osuna J, Medina G, et al. Splenectomy in systemic lupus erythematosus and autoimmune hematologic disease: a comparative analysis. Clin Rheumatol. 2018;37:943–948.10.1007/s10067-018-3979-4Suche in Google Scholar PubMed

[194] Barbhaiya M, Zuily S, Naden R, et al. The 2023 ACR/EULAR Antiphospholipid Syndrome Classification Criteria. Arthritis Rheumatol. 2023;75:1687–1702.10.1002/art.42624Suche in Google Scholar PubMed

[195] Zhao Y, Huang C, Zhou Y, et al. Performance validation of the 2023 American College of Rheumatology/European League Against Rheumatism antiphospholipid syndrome classification criteria in an antiphospholipid syndrome cohort. J Thromb Haemost. 2024;22:1660–1674.10.1016/j.jtha.2024.02.019Suche in Google Scholar PubMed

[196] Yang Y, Jiang H, Tang Z, et al. Assessment of the 2023 ACR/EULAR antiphospholipid syndrome classification criteria in a Chinese cohort: Impact on clinical practice. J Autoimmun. 2024;146:103237.10.1016/j.jaut.2024.103237Suche in Google Scholar PubMed

[197] Crowther MA, Ginsberg JS, Julian J, et al. A comparison of two intensities of warfarin for the prevention of recurrent thrombosis in patients with the antiphospholipid antibody syndrome. N Engl J Med. 2003;349:1133–1138.10.1056/NEJMoa035241Suche in Google Scholar PubMed

[198] Pengo V, Denas G, Zoppellaro G, et al. Rivaroxaban vs warfarin in high-risk patients with antiphospholipid syndrome. Blood. 2018;132:1365–1371.10.1182/blood-2018-04-848333Suche in Google Scholar PubMed

[199] Khairani CD, Bejjani A, Piazza G, et al. Direct Oral Anticoagulants vs Vitamin K Antagonists in Patients With Antiphospholipid Syndromes: Meta-Analysis of Randomized Trials. J Am Coll Cardiol. 2023;81:16–30.10.1016/j.jacc.2022.10.008Suche in Google Scholar PubMed PubMed Central

[200] Erkan D. Expert Perspective: Management of Microvascular and Catastrophic Antiphospholipid Syndrome. Arthritis Rheumatol. 2021;73:1780–1790.10.1002/art.41891Suche in Google Scholar PubMed

[201] Huang C, Zhao Y, Tian X, et al. Early recognition of catastrophic antiphospholipid syndrome in patients with antiphospholipid syndrome based on a Chinese cohort study. Clin Exp Rheumatol. 2023;41:1017–1023.10.55563/clinexprheumatol/e0voi7Suche in Google Scholar PubMed

[202] Rodríguez-Pintó I, Espinosa G, Erkan D, et al. The effect of triple therapy on the mortality of catastrophic anti-phospholipid syndrome patients. Rheumatology (Oxford). 2018;57:1264–1270.10.1093/rheumatology/key082Suche in Google Scholar PubMed

[203] Yelnik CM, Miranda S, Mékinian A, et al. Patients with refractory catastrophic antiphospholipid syndrome respond inconsistently to eculizumab. Blood. 2020;136:2473–2477.10.1182/blood.2020007499Suche in Google Scholar PubMed

[204] Buyon JP, Kim MY, Guerra MM, et al. Predictors of Pregnancy Outcomes in Patients With Lupus: A Cohort Study. Ann Intern Med. 2015;163:153–163.10.7326/M14-2235Suche in Google Scholar PubMed PubMed Central

[205] Hamulyák EN, Scheres LJ, Marijnen MC, et al. Aspirin or heparin or both for improving pregnancy outcomes in women with persistent antiphospholipid antibodies and recurrent pregnancy loss. Cochrane Database Syst Rev. 2020;5:CD012852.10.1002/14651858.CD012852.pub2Suche in Google Scholar PubMed PubMed Central

[206] Magro-Checa C, Zirkzee EJ, Huizinga TW, et al. Management of Neuropsychiatric Systemic Lupus Erythematosus: Current Approaches and Future Perspectives. Drugs. 2016;76:459–483.10.1007/s40265-015-0534-3Suche in Google Scholar PubMed PubMed Central

[207] Barile-Fabris L, Ariza-Andraca R, Olguín-Ortega L, et al. Controlled clinical trial of IV cyclophosphamide versus IV methylprednisolone in severe neurological manifestations in systemic lupus erythematosus. Ann Rheum Dis. 2005;64:620–625.10.1136/ard.2004.025528Suche in Google Scholar PubMed PubMed Central

[208] Ginzler EM, Wofsy D, Isenberg D, et al. Nonrenal disease activity following mycophenolate mofetil or intravenous cyclophosphamide as induction treatment for lupus nephritis: findings in a multicenter, prospective, randomized, open-label, parallel-group clinical trial. Arthritis Rheum. 2010;62:211–221.10.1002/art.25052Suche in Google Scholar PubMed

[209] Legge AC, Hanly JG. Recent advances in the diagnosis and management of neuropsychiatric lupus. Nat Rev Rheumatol. 2024;20:712–728.10.1038/s41584-024-01163-zSuche in Google Scholar PubMed

[210] Ramos-Casals M, Soto MJ, Cuadrado MJ, et al. Rituximab in systemic lupus erythematosus: A systematic review of off-label use in 188 cases. Lupus. 2009;18:767–776.10.1177/0961203309106174Suche in Google Scholar PubMed

[211] Narváez J, Ríos-Rodriguez V, de la Fuente D, et al. Rituximab therapy in refractory neuropsychiatric lupus: current clinical evidence. Semin Arthritis Rheum. 2011;41:364–372.10.1016/j.semarthrit.2011.06.004Suche in Google Scholar PubMed

[212] Manzi S, Sánchez-Guerrero J, Merrill JT, et al. 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. Ann Rheum Dis. 2012;71:1833–1838.10.1136/annrheumdis-2011-200831Suche in Google Scholar PubMed PubMed Central

[213] Palazzo L, Lindblom J, Cetrez N, et al. Determinants of neuropsychiatric flares in patients with systemic lupus erythematosus: results from five phase III trials of belimumab. Rheumatology (Oxford). 2024;63:798–808.10.1093/rheumatology/kead249Suche in Google Scholar PubMed PubMed Central

[214] Andrade RM, Alarcón GS, González LA, et al. Seizures in patients with systemic lupus erythematosus: data from LUMINA, a multiethnic cohort (LUMINA LIV). Ann Rheum Dis. 2008;67:829–834.10.1136/ard.2007.077594Suche in Google Scholar PubMed PubMed Central

[215] Nie Y, Sun B, He X, et al. The influence of intrathecal injection of methotrexate and dexamethasone on neuropsychiatric systemic lupus erythematosus (NPSLE): a retrospective cohort study of 386 patients with NPSLE. Arthritis Res Ther. 2023;25:50.10.1186/s13075-023-03030-wSuche in Google Scholar PubMed PubMed Central

[216] Arnaud L, Mathian A, Ruffatti A, et al. Efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies: an international and collaborative meta-analysis. Autoimmun Rev. 2014;13:281–291.10.1016/j.autrev.2013.10.014Suche in Google Scholar PubMed

[217] Chinese Society of Neurology, Chinese Society of Electroencephalography and Epilepsy. [Chinese consensus for the anti-seizure medications polytherapy.] Chin J Neurol. 2024;57(2):108–117.Suche in Google Scholar

[218] Justiz-Vaillant AA, Gopaul D, Soodeen S, et al. Neuropsychiatric Systemic Lupus Erythematosus: Molecules Involved in Its Imunopathogenesis, Clinical Features, and Treatment. Molecules. 2024;29:747.10.3390/molecules29040747Suche in Google Scholar PubMed PubMed Central

[219] Chen Y, Hu Z, Cai S, et al. Efficacy of plasma exchange on top of standard immunosuppressive therapy in adult autoimmune inflammatory rheumatic diseases-associated macrophage activation syndrome, a single center real-world analysis. Semin Arthritis Rheum. 2022;55:152043.10.1016/j.semarthrit.2022.152043Suche in Google Scholar PubMed

[220] Jiang Y, Zhao HJ, Luo H, et al. Efficacy of Whole-Blood Exchange Transfusion in Refractory Severe Autoimmune Haemolytic Anaemia Secondary to Systemic Lupus Erythematosus: A Real-World Observational Retrospective Study. Front Immunol. 2022;13:861719.10.3389/fimmu.2022.861719Suche in Google Scholar PubMed PubMed Central

[221] Yang M, Liao C, Zhu Q, et al. Meta-analysis on the efficacy and safety of immunoadsorption for systemic lupus erythematosus among Chinese population. Clin Rheumatol. 2020;39:3581–3592.10.1007/s10067-020-05156-7Suche in Google Scholar PubMed

[222] Nieto-Aristizábal I, Martínez T, Urbano MA, et al. Treatment with intravenous immunoglobulins in systemic lupus erythematosus: a single-center experience with 63 patients. Lupus. 2019;28:1566–1570.10.1177/0961203319883680Suche in Google Scholar PubMed

[223] Cajamarca-Barón J, Buitrago-Bohórquez J, Mendoza Orozco JE, et al. Efficacy and safety of intravenous immunoglobulin in patients with lupus nephritis: A systematic review of the literature. Autoimmun Rev. 2022;21:103182.10.1016/j.autrev.2022.103182Suche in Google Scholar PubMed

[224] Wallace DJ, Furie RA, Tanaka Y, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2018;392:222–231.10.1016/S0140-6736(18)31363-1Suche in Google Scholar PubMed

[225] Morand EF, Vital EM, Petri M, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-I). Lancet. 2023;401:1001–1010.10.1016/S0140-6736(22)02607-1Suche in Google Scholar PubMed

[226] Petri M, Bruce IN, Dörner T, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-II). Lancet. 2023;401:1011–1019.10.1016/S0140-6736(22)02546-6Suche in Google Scholar PubMed

[227] Yang J, Li J, Shi L. Successful remission with tofacitinib in a patient with refractory bullous systemic lupus erythematosus. Rheumatology (Oxford). 2022;61:e341–e343.10.1093/rheumatology/keac309Suche in Google Scholar PubMed

[228] Chen SL, Zheng HJ, Zhang LY, et al. Case report: Joint deformity associated with systemic lupus erythematosus. Immun Inflamm Dis. 2022;10:e717.10.1002/iid3.717Suche in Google Scholar PubMed PubMed Central

[229] You H, Zhang G, Wang Q, et al. Successful treatment of arthritis and rash with tofacitinib in systemic lupus erythematosus: the experience from a single centre. Ann Rheum Dis. 2019;78:1441–1443.10.1136/annrheumdis-2019-215455Suche in Google Scholar PubMed

[230] Chen D, Yin H, Tang G, et al. Efficacy of low-dose of baricitinib in the treatment of patchy alopecia and sicca syndrome in an SLE patient. Scand J Rheumatol. 2022;51:428–430.10.1080/03009742.2022.2087901Suche in Google Scholar PubMed

[231] Maeshima K, Shibata H. Efficacy of JAK 1/2 inhibition in the treatment of diffuse non-scarring alopecia due to systemic lupus erythematosus. Ann Rheum Dis. 2020;79:674–675.10.1136/annrheumdis-2019-216571Suche in Google Scholar PubMed

[232] Fornaro M, Coladonato L, Venerito V, et al. Efficacy of baricitinib on refractory skin papulosquamous rash in a patient with systemic lupus erythematosus. Rheumatology (Oxford). 2020;59:1792.10.1093/rheumatology/kez561Suche in Google Scholar PubMed

[233] Morand E, Pike M, Merrill JT, et al. Deucravacitinib, a Tyrosine Kinase 2 Inhibitor, in Systemic Lupus Erythematosus: A Phase II, Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheumatol. 2023;75:242–252.10.1002/art.42391Suche in Google Scholar PubMed PubMed Central

[234] Baker M, Chaichian Y, Genovese M, et al. Phase II, randomised, double-blind, multicentre study evaluating the safety and efficacy of filgotinib and lanraplenib in patients with lupus membranous nephropathy. RMD Open. 2020;6:e001490.10.1136/rmdopen-2020-001490Suche in Google Scholar PubMed PubMed Central

[235] Chen Y, Wang L, Li N, et al. Tripterygium glycosides for safely controlling disease activity in systemic lupus erythematosus: a systematic review with meta-analysis and trial sequential analysis. Front Pharmacol. 2023;14:1207385.10.3389/fphar.2023.1207385Suche in Google Scholar PubMed PubMed Central

[236] Sun F, Yang X, Ma D, et al. [Reproductive Toxicity of Tripterygium Wilfordii Hook .f: A Systematic Review and Meta-analysis.] Chin J Pharmacovigil. 2014; 11: 94-99.Suche in Google Scholar

[237] Mougiakakos D, Krönke G, Völkl S, et al. CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus. N Engl J Med. 2021;385:567–569.10.1056/NEJMc2107725Suche in Google Scholar PubMed

[238] Mackensen A, Müller F, Mougiakakos D, et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat Med. 2022;28:2124–2132.10.1038/s41591-022-02017-5Suche in Google Scholar PubMed

[239] Müller F, Taubmann J, Bucci L, et al. CD19 CAR T-Cell Therapy in Autoimmune Disease - A Case Series with Follow-up. N Engl J Med. 2024;390:687–700.10.1056/NEJMoa2308917Suche in Google Scholar PubMed

[240] Li M, Zhang Y, Jiang N, et al. Anti-CD19 CAR T Cells in Refractory Immune Thrombocytopenia of SLE. N Engl J Med. 2024;391:376–378.10.1056/NEJMc2403743Suche in Google Scholar PubMed

[241] Wang W, He S, Zhang W, et al. BCMA-CD19 compound CAR T cells for systemic lupus erythematosus: a phase 1 open-label clinical trial. Ann Rheum Dis. 2024;83:1304–1314.10.1136/ard-2024-225785Suche in Google Scholar PubMed

[242] O’Dwyer T, Durcan L, Wilson F. Exercise and physical activity in systemic lupus erythematosus: A systematic review with meta-analyses. Semin Arthritis Rheum. 2017;47:204–215.10.1016/j.semarthrit.2017.04.003Suche in Google Scholar PubMed

[243] Wu ML, Yu KH, Tsai JC. The Effectiveness of Exercise in Adults With Systemic Lupus Erythematosus: A Systematic Review and Meta-Analysis to Guide Evidence-Based Practice. Worldviews Evid Based Nurs. 2017;14:306–315.10.1111/wvn.12221Suche in Google Scholar PubMed

[244] Zhang J, Wei W, Wang CM. Effects of psychological interventions for patients with systemic lupus erythematosus: a systematic review and meta-analysis. Lupus. 2012;21:1077–1087.10.1177/0961203312447667Suche in Google Scholar PubMed

[245] Zandman-Goddard G, Solomon M, Rosman Z, et al. Environment and lupus-related diseases. Lupus. 2012;21:241–250.10.1177/0961203311426568Suche in Google Scholar PubMed

[246] Yang S, Chris TL. Eyebrow threading–induced tumid lupus erythematosus. J Am Acad Dermatol. 2019;81:Supplement 1, AB210.10.1016/j.jaad.2019.06.773Suche in Google Scholar

[247] Smyk D, Rigopoulou EI, Bizzaro N, et al. Hair dyes as a risk for autoimmunity: from systemic lupus erythematosus to primary biliary cirrhosis. Auto Immun Highlights. 2012;4:1–9.10.1007/s13317-011-0027-7Suche in Google Scholar PubMed PubMed Central

[248] Zahn S, Graef M, Patsinakidis N, et al. Ultraviolet light protection by a sunscreen prevents interferon-driven skin inflammation in cutaneous lupus erythematosus. Exp Dermatol. 2014;23:516–518.10.1111/exd.12428Suche in Google Scholar PubMed

[249] Kuhn A, Gensch K, Haust M, et al. Photoprotective effects of a broad-spectrum sunscreen in ultraviolet-induced cutaneous lupus erythematosus: a randomized, vehicle-controlled, double-blind study. J Am Acad Dermatol. 2011;64:37–48.10.1016/j.jaad.2009.12.053Suche in Google Scholar PubMed

[250] Abdul Kadir WD, Jamil A, Shaharir SS, et al. Photoprotection awareness and practices among patients with systemic lupus erythematosus and its association with disease activity and severity. Lupus. 2018;27:1287–1295.10.1177/0961203318770016Suche in Google Scholar PubMed

[251] Parisis D, Bernier C, Chasset F, et al. Impact of tobacco smoking upon disease risk, activity and therapeutic response in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev. 2019;18:102393.10.1016/j.autrev.2019.102393Suche in Google Scholar PubMed

[252] Najeeb O Ghaussy, Wilmer Sibbitt Jr, Arthur D Bankhurst, et al. Cigarette smoking and disease activity in systemic lupus erythematosus. J Rheumatol. 2003;30:1215–1221.Suche in Google Scholar

[253] Sousa JR, Rosa ÉPC, Nunes IFOC, et al. Effect of vitamin D supplementation on patients with systemic lupus erythematosus: a systematic review. Rev Bras Reumatol Engl Ed. 2017;57:466–471.10.1016/j.rbre.2017.08.001Suche in Google Scholar PubMed

[254] de Medeiros MCS, Medeiros JCA, de Medeiros HJ, et al. Dietary intervention and health in patients with systemic lupus erythematosus: A systematic review of the evidence. Crit Rev Food Sci Nutr. 2019;59:2666–2673.10.1080/10408398.2018.1463966Suche in Google Scholar PubMed

[255] Fei Y, Shi X, Gan F, et al. Death causes and pathogens analysis of systemic lupus erythematosus during the past 26 years. Clin Rheumatol. 2014;33:57–63.10.1007/s10067-013-2383-3Suche in Google Scholar PubMed

[256] Sun X, Xu L, Deng Y, et al. [The risk factors of infections in patients with systemic lupus erythematosus.] Chin J Nurs. 2015;50:828–835.Suche in Google Scholar

[257] Firooz N, Albert DA, Wallace DJ, et al. High-sensitivity C-reactive protein and erythrocyte sedimentation rate in systemic lupus erythematosus. Lupus. 2011;20:588–597.10.1177/0961203310393378Suche in Google Scholar PubMed

[258] Serio I, Arnaud L, Mathian A, et al. Can procalcitonin be used to distinguish between disease flare and infection in patients with systemic lupus erythematosus: a systematic literature review. Clin Rheumatol. 2014;33:1209–1215.10.1007/s10067-014-2738-4Suche in Google Scholar PubMed

[259] Yang MJ, Chen CY, Chang WH, et al. Pregnancy outcome of systemic lupus erythematosus in relation to lupus activity before and during pregnancy. J Chin Med Assoc. 2015;78:235–240.10.1016/j.jcma.2014.11.008Suche in Google Scholar PubMed

[260] Knight CL, Nelson-Piercy C. Management of systemic lupus erythematosus during pregnancy: challenges and solutions. Open Access Rheumatol. 2017;9:37–53.10.2147/OARRR.S87828Suche in Google Scholar PubMed PubMed Central

[261] Ren S, Liu E, Yang Y. A Meta-Analysis of Pregnancy Outcomes in Patients with Lupus Nephritis. Journal of North China University of Science and Technology (Health Sciences Edition). 2018;20:7.Suche in Google Scholar

[262] Chen S, Sun X, Wu B, et al. Pregnancy in Women with Systemic Lupus Erythematosus: A Retrospective Study of 83 Pregnancies at a Single Centre. Int J Environ Res Public Health. 2015;12:9876–9888.10.3390/ijerph120809876Suche in Google Scholar PubMed PubMed Central

[263] Wei Q, Ouyang Y, Zeng W, et al. Pregnancy complicating systemic lupus erythematosus: a series of 86 cases. Arch Gynecol Obstet. 2011;284:1067–1071.10.1007/s00404-010-1786-5Suche in Google Scholar PubMed

[264] Lazzaroni MG, Dall’Ara F, Fredi M, et al. A comprehensive review of the clinical approach to pregnancy and systemic lupus erythematosus. J Autoimmun. 2016;74:106–117.10.1016/j.jaut.2016.06.016Suche in Google Scholar PubMed

[265] Wu J, Ma J, Bao C, et al. Pregnancy outcomes among Chinese women with and without systemic lupus erythematosus: a retrospective cohort study. BMJ Open. 2018;8:e020909.10.1136/bmjopen-2017-020909Suche in Google Scholar PubMed PubMed Central

[266] Borella E, Lojacono A, Gatto M, et al. Predictors of maternal and fetal complications in SLE patients: a prospective study. Immunol Res. 2014;60:170–176.10.1007/s12026-014-8572-6Suche in Google Scholar PubMed

[267] Hussein Aly EA, Mohamed Riyad R, Nabil Mokbel A. Pregnancy outcome in patients with systemic lupus erythematosus: A single center study in the High Risk Pregnancy unit. Mid East Fertil Soc J. 2016;21:168–174.10.1016/j.mefs.2015.12.003Suche in Google Scholar

[268] National Clinical Research Center for Dermatologic and Immunologic Diseases; National Clinical Research Center for Obstetrics and Gynecology; Chinese Research Committee of Pregnancy and Reproduction in Autoimmune Rheumatic Diseases; Chinese Rheumatism Data Center; Chinese Systemic Lupus Erythematosus Treatment and Research Group. [2022 Chinese guideline for the management of pregnancy and reproduction in patients with systemic lupus erythematous.] Zhong Hua Nei Ke Za Zhi. 2022; 61:1184–1205.Suche in Google Scholar

[269] McDonald EG, Bissonette L, Ensworth S, et al. Monitoring of Systemic Lupus Erythematosus Pregnancies: A Systematic Literature Review. J Rheumatol. 2018;45:1477–1490.10.3899/jrheum.171023Suche in Google Scholar PubMed

[270] Bundhun PK, Soogund MZ, Huang F. Impact of systemic lupus erythematosus on maternal and fetal outcomes following pregnancy: A meta-analysis of studies published between years 2001–2016. J Autoimmun. 2017;79:17–27.10.1016/j.jaut.2017.02.009Suche in Google Scholar PubMed

[271] Yousef Yengej FA, van Royen-Kerkhof A, Derksen RHWM, et al. The development of offspring from mothers with systemic lupus erythematosus. A systematic review. Autoimmun Rev. 2017;16:701–711.10.1016/j.autrev.2017.05.005Suche in Google Scholar PubMed

[272] Guillotin V, Bouhet A, Barnetche T, et al. Hydroxychloroquine for the prevention of fetal growth restriction and prematurity in lupus pregnancy: A systematic review and meta-analysis. Joint Bone Spine. 2018;85:663–668.10.1016/j.jbspin.2018.03.006Suche in Google Scholar PubMed

[273] Chinese SLE Treatment and Research Group, Chinese Rheumatism Data Center. [Perinatal Management Recommendations for Chinese Systemic Lupus Erythematosus Patients.] Chin Med J. 2015;95:1056–1060.Suche in Google Scholar

[274] Furer V, Rondaan C, Heijstek MW, et al. 2019 update of EU-LAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann Rheum Dis. 2020;79:39–52.10.1136/annrheumdis-2019-215882Suche in Google Scholar PubMed

[275] Bass AR, Chakravarty E, Akl EA, et al. 2022 American College of Rheumatology Guideline for Vaccinations in Patients With Rheumatic and Musculoskeletal Diseases. Arthritis Rheumatol. 2023;75:333–348.10.1002/art.42386Suche in Google Scholar PubMed PubMed Central

Received: 2025-08-03
Accepted: 2025-08-31
Published Online: 2025-10-04

© 2025 Jiuliang Zhao, Shangzhu Zhang, Qian Wang, Xinping Tian, Yaolong Chen, Mengtao Li, Xiaofeng Zeng, published by De Gruyter on behalf of NCRC-DID

This work is licensed under the Creative Commons Attribution 4.0 International License.

Heruntergeladen am 7.10.2025 von https://www.degruyterbrill.com/document/doi/10.1515/rir-2025-0017/html
Button zum nach oben scrollen