Journal Information
Visits
78
Vol. 20. Issue 9.
Pages 490-510 (November 2024)
Consensus statement
Full text access
Clinical Practice Mexican Guidelines for the Treatment of Systemic Lupus Erythematosus: 2024 Update
Guías de Práctica Clínica para el tratamiento del lupus eritematoso sistémico del Colegio Mexicano de Reumatología. Actualización 2024
Visits
78
Lilia Andrade-Ortegaa,
Corresponding author
liliaandrade@prodigy.net.mx

Corresponding author.
, Daniel Xibillé-Friedmannb, Dionicio A. Galarza-Delgadoc, Miguel Ángel Saavedrad, José Alvarez-Nemegyeie, Mary-Carmen Amigo-Castañedaf, Hilda Fragoso-Loyog, María Vanessa Gordillo-Huertah, Fedra Irazoque-Palazuelosa, Luis Javier Jara-Quezada†i, Javier Merayo-Chalicog, Margarita Portela-Hernándezj, Sandra Sicsik-Ayalak, Carlos Abud-Mendozal, Deshire Alpizar-Rodriguezm, José Luis Amaya-Estradan, Yaneth R. Barragán-Navarroo, Sandra M. Carrillo-Vázquezp, Zully Castro-Colínq, Luis Javier Cruz-Álvarezr..., Sergio Durán-Barragáns, Jorge A. Esquivel-Valerioc, Jorge Iván Gamez-Navat, Conrado García-Garcíau, Laura Gonzalez-Lopezt, Jaime Hadid-Smekea, Alina Hernández-Bedollav, María Fernanda Hernández-Cabreraw, David A. Herrera-VanOostdamx, Gabriel Horta-Baasy, Alberto E. Iturbide-Escamillaz, Sandra Muñoz-Lopeza, Cesar Pacheco-Tenaaa, Mario Pérez-Cristóbalj, Rafael R. Pimentel-Leonz, Mercedes Pinto-Ortizab, María Azucena Ramos-Sánchezac, Diana V. Sandoval-Cabreraad, Karina Santana de Andag, Luis H. Silveiraae, Leonor A. Barile-FabrisafVer más
a Servicio de Reumatología, Centro Médico Nacional 20 de Noviembre, ISSSTE, CDMX, Mexico
b Departamento de Reumatología, Hospital General de Cuernavaca, SSM, Cuernavaca, Mexico
c Servicio de Reumatología, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Nuevo León, Mexico
d División de Investigación en Salud, Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza IMSS, CDMX, Mexico
e Escuela de Medicina, Universidad Marista de Mérida, Mérida, Mexico
f Departamento de Medicina Interna, Centro Médico ABC, CDMX, Mexico
g Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencia Médicas y Nutrición, Dr Salvador Zubirán, CDMX, Mexico
h Servicio de Medicina Interna, Hospital General Querétaro del ISSSTE, Querétaro, Mexico
i División de Reumatología, Instituto Nacional de Rehabilitación Dr. Luis Guillermo Ibarra Ibarra, CDMX, Mexico
j Departamento de Reumatología, Hospital de Especialidades del CMN SXXI, IMSS, CDMX, Mexico
k Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Mexico
l Departamento de Reumatología, Hospital Central "Dr. Ignacio Morones Prieto", Facultad de Medicina de la UASLP, San Luis Potosí, Mexico
m Centro Médico Althea, Cuernavaca, Mexico
n Servicio de Interna, Hospital Central Sur de Alta Especialidad, Petróleos Mexicanos, CDMX, Mexico
o Servicio de Medicina Interna, Hospital General Regional No. 20. IMSS, Tijuana, Mexico
p Coordinación de Medicina Interna, Hospital Regional 1ro de Octubre ISSSTE CDMX, Mexico
q Servicio de Reumatología, Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza IMSS, CDMX, Mexico
r Subdirección de Educación en Salud, Hospital General Reynosa, Reynosa, Mexico
s Departamento de Clínicas Médicas del Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
t Intituto de Terapéutica Experimental y Clínica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
u Servicio de Reumatología, Hospital General de México "Dr. Eduardo Liceaga", CDMX, Mexico
v Servicio de Reumatología, Centro Médico Dalinde, CDMX, Mexico
w Servicio de Medicina Interna y Reumatología, Hospital General Regional 220, IMSS, Toluca, Mexico
x Departamento de Reumatología, Hospital Central "Dr. Ignacio Morones Prieto", San Luis Potosí, Mexico
y Servicio de Reumatología, Hospital General Regional # 1, Instituto Mexicano del Seguro Social, Mérida, Mexico
z Servicio de Dermatología, Hospital General de Zona 1 IMSS, Tlaxcala, Mexico
aa Facultad de Medicina, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
ab Servicio de Reumatología, Hospital Ángeles Mocel, CDMX, Mexico
ac CMN Noreste, Hospital de Especialidades No. 25. IMSS, Monterrey, Mexico
ad Servicio de Medicina Interna, Hospital General Regional #2 “Dr. Guillermo Fajardo Ortiz”, IMSS, CDMX, Mexico
ae Departamento de Reumatología, Instituto Nacional de Cardiología Ignacio Chávez, CDMX, Mexico
af Hospital Ángeles Pedregal, CDMX, Mexico
Ver más
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (18)
Table 1. General principles.
Tables
Table 2. Definitions of remission and low activity.
Tables
Table 3. Recommendations for using glucocorticoids, antimalarials, and NSAIDs.
Tables
Table 4. Recommendations for using DMARDs and immunosuppressants.
Tables
Table 5. Recommendations for using biological therapy.
Tables
Table 6. Recommendations for the management of constitutional manifestations.
Tables
Table 7. Recommendations for the management of mucocutaneous manifestations.
Tables
Table 8. Recommendations for the management of musculoskeletal manifestations.
Tables
Table 9. Recommendations for the management of haematological manifestations.
Tables
Table 10. Recommendations for the management of renal manifestations.
Tables
Table 11. Recommendations for the management of neuropsychiatric manifestations.
Tables
Table 12. Recommendations for the management of cardiopulmonary manifestations.
Tables
Table 13. Recommendations for the management of gastrointestinal manifestations.
Tables
Table 14. Recommendations on vaccination.
Tables
Table 15. Recommendations on perioperative management.
Tables
Table 16. Recommendations in pregnancy and lactation.
Tables
Table 17. Use of drugs during pregnancy and lactation in SLE patients.
Tables
Table 18. Recommendations to reduce cardiovascular risk.
Tables
Show moreShow less
Abstract

Herein we present the update for the Mexican Guidelines for the Treatment of Systemic Lupus Erythematosus.

It involves the participation of several experts along the country, following the GRADE system.

We included aspects regarding vaccines, pregnancy and cardiovascular risk which were not presented in the previous guidelines in 2017.

Keywords:
Guidelines
Treatment
Systemic lupus erythematosus
Resumen

Presentamos la actualización de las Guías de Práctica Clínica para el tratamiento del lupus eritematoso sistémico del Colegio Mexicano de Reumatología elaboradas por un grupo de expertos en la enfermedad tanto a nivel privado como gubernamental, de acuerdo con la metodología GRADE.

Se incluyen por primera vez aspectos sobre vacunación, embarazo y riesgo cardiovascular.

Palabras clave:
Guías de práctica clínica
Tratamiento
Lupus eritematoso sistémico
Full Text
Introduction

In 2017, the Mexican College of Rheumatology published the first Clinical Practice Guidelines (CPG) for the treatment of Mexican patients with systemic lupus erythematosus (SLE) and committed to update them periodically.1

CPGs are an extremely useful tool to guide physicians on the treatment of a specific disease and require a strict methodology in their development and implementation. Therapeutic guidelines need to be updated when new drugs emerge with sufficient evidence to support their use, or when the indications change for drugs that are already in use.

SLE is a prototypical autoimmune disease, in terms of both its complexity and heterogeneity. Currently, the therapeutic strategy aims to achieve specific targets and, as much as is possible, limit accumulated damage, especially when secondary to drugs used in the treatment of the disease. Given the characteristics of SLE patients, recommendations should include general patient management, specific treatment of target organ manifestations, and monitoring of comorbidities arising from both treatment and disease. It is also important to consider genetic and ethnic factors (such as the higher severity observed in Mestizos), as well as access to health systems and appropriate therapies.

This paper includes topics that were not covered in previous guidelines, such as treat-to-target, cardiovascular risk, and treatment during pregnancy and lactation. In addition, all recommendations for drug groups and organ- and system-specific manifestations are updated.

Methodology

A working group of 40 rheumatologists, all certified and active members of the Mexican College of Rheumatology, with experience in the treatment of patients with SLE and professionally active in different institutions and regions of the country, in both private and governmental practice, was formed to undertake this update. Two medical experts in CPG methodology were also involved.

The expert panel met for the first time remotely in September 2023. At this meeting, the methodology was defined, and the content of the guidelines was divided into four sections: 1. General considerations, 2. Drugs, 3. Treatment by organ and system, and 4. Special situations. Specific working teams were formed for each of the subtopics and together they reviewed and updated the research questions using the PICO format (patient, intervention, comparison, outcomes), developing those that corresponded to topics not included in the previous recommendations.2

A systematic review of the literature was conducted covering information published since 2017. All articles from PubMed, Cochrane Central Register of Controlled Trials (CENTRAL), Google Scholar, and SCIELO published between January 2017 and December 2023 were taken into account for this update (considering that previous guidelines had included publications up to December 2016). Articles published from January 2010 onwards were considered for de novo questions.

All study designs were included, with priority given to meta-analyses, clinical trials, cohort, case-control, and cross-sectional studies. However, case reports, narrative reviews, or consensus studies were considered in the case of research questions with very limited information or where there were no papers of sufficient quality.

Each team received the PICO search mechanisms from the methodologists, which were adapted to the topic allocated. The articles were then reviewed, and the search was completed based on those that were related and the references consulted. The articles selected were those that provided clear answers to the research questions posed. The findings from the eligible studies were synthesised in evidence tables, using the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) approach.2 From the evidence tables, adjustments were made to previous recommendations or new recommendations were developed, grading the level of evidence and strength of each recommendation as per the GRADE system.

All initial recommendations were sent for review and suggestions to the team, who provided feedback. Disagreements were resolved by discussion among team members.

In January 2024, a hybrid meeting was held with the entire research team to review each of the recommendations issued, deliberate, and establish the degree of consensus, resulting in the final version. Recommendations were accepted with more than 80% agreement; in cases where this figure was not achieved, they were analysed and amended as necessary, repeating the vote to achieve consensus or to establish that consensus had not been reached.

Recommendations

This update of recommendations for the management of SLE is structured as follows:

  • 1

    General principles for management of the disease.

Recommendations:

  • 2

    On the indications for and monitoring of the main groups of drugs used in SLE.

  • 3

    For the specific treatment of the disease in the different organs and systems.

  • 4

    For special situations that are important because of their impact on SLE, such as the perioperative period, vaccination, pregnancy, and cardiovascular risk.

General principles

The general principles of treatment in SLE encompass the concept of comprehensive management of a very complex disease. They highlight the role of the rheumatologist as the leader of the multidisciplinary care team and the importance of regular patient assessment. They emphasise the need for an end goal of treatment (remission or low disease activity) and relapse prevention as the primary means to reduce progression and long-term damage. They consider the importance of non-pharmacological measures, as well as the selection of drugs based on their increased efficacy, safety, and availability. They also stress that the patient should always be involved in therapeutic decisions.3–6 (Tables 1 and 2)

Table 1.

General principles.

Optimal treatment of patients with SLE requires pharmacological and non-pharmacological interventions by a multidisciplinary team coordinated by the rheumatology specialist.3 
Treatment should be based on shared decision-making between patient and physician. This facilitates adherence, which in turn will improve disease control.3,4,7,8 
Early diagnosis and timely treatment improve the patient's prognosis. Disease activity should be determined at each clinical evaluation and the damage associated with the disease should be determined at least once a year, using validated instruments.9–11 
The goal of treatment is to control disease activity, limit damage, and improve patients' quality of life.10,12 The ideal therapeutic goal is to put the disease into remission and if this is not possible, at least reach a state of low disease activity (Table 2).13,14 
The treatment of SLE should be personalised and consider the specific organ involvement and its severity. It is also necessary to consider the efficacy, safety, and availability of treatment, the patient's comorbidities, as well as the societal, economic, and family impact of the disease.3,7,8 
Non-pharmacological treatment should include photoprotection, balanced diet, smoking cessation, regular exercise, psychological support, cardiovascular risk assessment, family planning counselling, and prenatal care.15–18 

SLE: systemic lupus erythematosus.

Table 2.

Definitions of remission and low activity.

Domain  Remission DORIS  LLDAS 
SLEDAI-2K  SLEDAI clinical=0(allows for serological activity)  SLEDAI ≤4*- no major organ activity- no haemolytic anaemia or gastrointestinal activity- no new features of activity compared with the previous assessment 
PGA (0−3)  <.,5  ≤1 
PDN dose (or equivalent)  ≤5 mg/day  ≤7.5 mg/day 
Other therapies  Stable doses of antimalarials, immunosuppressants, or biologicals

DORIS 2021: Definition of Remission In SLE; LLDAS: Lupus Low Disease Activity State; PDN: prednisone; PGA: Physician Global Assessment; SLEDAI-2K: Systemic Lupus Erythematosus Disease Activity Index 2000.

*

Includes serological activity.6,19

Drugs

Treatment of SLE includes a combination, often sequential, of antimalarials, glucocorticoids (GCs), non-steroidal anti-inflammatory drugs (NSAIDs), disease-modifying drugs, immunosuppressants, and biological therapy. The choice of drugs, as well as the dose and duration of administration, should be individualised according to the type and severity of clinical manifestations, disease activity, organs and systems involved, as well as associated comorbidities.

Antimalarials are considered mainstays in the treatment of SLE; they have photoprotective, lipid-lowering, anti-angiogenic, antithrombotic, and hypoglycaemic effects and, through their interaction with Toll-like receptors (TLRs), they also inhibit the function of B-cell activating factor (BAFF) and phospholipase A2 (PLA2). This pleiotropic effect makes them drugs of choice to treat cutaneous manifestations, mild to moderately active SLE, to concomitantly treat major organ involvement, and to prevent long-term relapse.20–22

GCs are the cornerstone of the immediate treatment of the disease. Their anti-inflammatory and immunomodulatory effect allows rapid control of many manifestations of lupus. The dose and time of administration of this group of drugs is determined by the severity of the condition and, by international consensus, the doses of prednisone (or its equivalent) have been defined as low dose (<7.5 mg), moderate dose (>7.5 mg and <30 mg), high dose (>30 mg and <100 mg), and very high dose (>100 mg).23 Despite their undoubted efficacy, because of their short- and long-term adverse effects, GCs should always be administered concomitantly with other disease-modifying or immunosuppressive drugs so as to reduce the dose and even discontinue them as quickly as possible.24

NSAIDs are often used for symptomatic treatment of pain and inflammation in SLE patients. Generally, NSAIDs are used at the full recommended dose for as short a time as possible in acute conditions, and at the minimum dose necessary to maintain clinical response in chronic conditions. The choice of individual NSAID will depend on the variability of response and the patient's risk factors for gastrointestinal, cardiovascular, renal, and hepatic toxicity, among others. It is necessary to periodically evaluate the indication for continued use and always assess the risk of interactions and adverse events when using other commonly used drugs in SLE patients (GCs, antihypertensives, hypoglycaemics, anticoagulants, etc.).25,26

Disease-modifying drugs such as methotrexate and leflunomide are useful when there are musculoskeletal manifestations.27 Immunosuppressants are used in various moderate and severe manifestations of the disease, but also in mild forms due to their GC-sparing effect. The most commonly used are azathioprine (AZA), mycophenolate mofetil (MMF), cyclophosphamide (CPM), and calcineurin inhibitors (cyclosporine, tacrolimus, and voclosporin). In some circumstances, a remission induction schedule followed by a lower dose as maintenance therapy is used to consolidate remission and prevent relapse. The choice of immunosuppressant will depend on the type of manifestation, its severity, and the individual toxicity profile. Multiple therapy is increasingly indicated as a way to achieve better short- and long-term outcomes with less toxicity.28–35

Biological therapy has been used for a decade in SLE, specifically monoclonal antibodies directed against B cells, T cells and, more recently, type I interferon. These mechanisms of action have been shown to be effective, and they work in conjunction with standard therapy to control various manifestations of the disease and to reduce the frequency and severity of relapses, as well as delay cumulative damage. Although results appear better in antinuclear antibody-positive patients, their use should not be limited to this subset of patients. Belimumab has recently been licensed for use as concomitant therapy in patients with lupus nephritis.36–41

Clinical trials of Janus tyrosine kinase inhibitors in SLE patients are currently underway with promising results for manifestations in various organs, including the kidney. CAR-T cells (chimeric molecules capable of redirecting T-cell specificity against target antigens) are a novel area of research in patients with severe and refractory SLE, with encouraging results.42–44

Tables 3–5 describe the treatment recommendations for SLE with the different drug groups.

Table 3.

Recommendations for using glucocorticoids, antimalarials, and NSAIDs.

Recommendation  Quality of evidence  Strength of recommendation 
It is recommended to use GCs with doses and route of administration according to the disease activity, type of manifestation, and severity. In mild forms doses ≤10 mg/day of prednisone or equivalent are recommended, in moderate forms .5−1 mg/kg/day, and in severe forms doses ≥1 mg/kg/day. Intravenous methylprednisolone pulses 250−1,000 mg/day, for 1−3 days should be indicated in patients with moderate and/or severe forms.23,24  High  Strong 
Gradual tapering of GCs is recommended in patients with stable disease, with close monitoring for early detection of relapse after discontinuation24,45High  Strong 
Hydroxychloroquine (HCQ) or chloroquine (CLQ) is recommended for all patients with SLE unless contraindicated.20–22  High  Strong 
It is recommended that the daily dose of HCQ not exceed 5 mg/kg day per actual body weight for all patients and CLQ 2.3 mg/kg.46  Low  Strong 
A baseline and then annual ophthalmological assessment are recommended for adverse effect monitoring.47 In patients with prolonged exposure (>5 years), OCT (optical coherence tomography) is recommended.47,48  Low  Strong 
The use of NSAIDs is recommended in mild SLE activity for short periods for the control of joint pain, arthritis, myalgias, chest pain, and/or fever, when the potential benefit outweighs the known risks of NSAIDs, and paracetamol has been insufficient or has not been tolerated.26  Low  Strong 
It is recommended that patients taking NSAIDs chronically should undergo cardiovascular and renal risk assessment, and monitoring of allergic reactions, skin reactions, and aseptic meningitis, due to the high risk in patients with SLE.25  Low  Strong 

GC: glucocorticoids; NSAIDs: nonsteroidal anti-inflammatory drugs; SLE: systemic lupus erythematosus.

Table 4.

Recommendations for using DMARDs and immunosuppressants.

Recommendation  Quality of evidence  Strength of recommendation 
It is recommended that methotrexate be used especially for musculoskeletal manifestations at doses of 7.5–25 mg orally or parenterally; adverse events to watch for are myelosuppression, hepatotoxicity, pneumonitis, alopecia, stomatitis, and teratogenicity.27  Moderate  Strong 
Using leflunomide is suggested in similar circumstances.49  Low  Weak 
It is recommended that the dose of azathioprine range from 1 to 3 mg/kg/day and closely monitor for adverse events (myelosuppression, hepatotoxicity, lymphoproliferative disorders, and teratogenicity).28  Moderate  Strong 
Mycophenolic acid/MMF is recommended at doses of 1–3 g/day according to the organ involved and the severity of the manifestation. Adverse events to be monitored are cytopenias, impaired liver function, diarrhoea, and teratogenicity.50  High  Strong 
It is recommended that calcineurin inhibitors (cyclosporine, tacrolimus, voclosporin) be used mainly in refractory nephropathy or as part of multiple therapy; their main adverse events are gingival hyperplasia, arterial hypertension, hirsutism, renal failure, and anaemia.31,32  High  Strong 
IV CPM at a dose of 500 to 1,000 mg/m2 based on body surface area is recommended. Adverse events to watch for are cytopenias, infertility, teratogenicity, myeloproliferative disorders, haemorrhagic cystitis, and bladder cancer. It is recommended to use the lowest dose of CPM possible on a case-by-case basis.29,30  Moderate  Strong 
The use of gonadotrophin-releasing hormone (GnRH) is recommended as ovarian protection in the case of women treated with CPM and who wish to have (more) children.51  High  Strong 
Combination therapy with immunosuppressants (multiple therapy) is recommended, and is considered a first treatment option in moderate to severe lupus after each patient has been appropriately assessed.31–35,41  High  Strong 
It is recommended that the prevention, follow-up, and treatment of infections be a priority in patients using immunosuppressants, particularly CPM.30,52,53  High  Strong 

CPM: cyclophosphamide.

Table 5.

Recommendations for using biological therapy.

Recommendation  Quality of evidence  Strength of recommendation 
Biological therapy (belimumab, anifrolumab) is recommended as adjuvant treatment to standard management in patients with active, antibody-positive SLE and failure of previous therapy.36,37  High  Strong 
Belimumab in combination with standard immunosuppressive therapies is recommended for the treatment of patients with active lupus nephritis.38  High  Strong 
Rituximab is suggested for moderate to severe manifestations of lupus with failure of previous treatments.39,40,43,54,55  Moderate  Weak 

SLE: systemic lupus erythematosus.

Constitutional manifestations

The constitutional manifestations of lupus, as is common in many chronic systemic conditions, are a multifactorial phenomenon that significantly impacts patients' quality of life. In the event of fever, infectious processes must be ruled out before attributing fever to disease activity alone.

Treatment of fatigue should be symptomatic and holistic, including regular assessments, exercise programmes, and psychoeducational interventions, always considering other related causes such as injury or comorbidities and taking the influence of psychological, societal, and lifestyle factors into account. The evidence found in the literature allows the following recommendations to be made for this group of manifestations of the disease.56,57 (Table 6)

Table 6.

Recommendations for the management of constitutional manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
It is recommended that assessment of the presence and intensity of fatigue and its impact on quality of life be included as part of the routine clinical care of the patient with SLE.56–58  Low  Strong 
It is recommended that a physical exercise programme, preferably aerobic, be included as an adjunct to treatment in SLE patients without clinical disease activity.59–63  Moderate  Strong 
It is recommended that an education and support programme related to the disease be provided and implemented.64,65  Moderate  Strong 
The addition of belimumab is suggested for the treatment of fatigue that is disabling or severely interferes with quality of life in SLE patients who have not responded to other treatment regimens.66,67  High  Weak 
Optimising vitamin D levels is suggested as adjuvant treatment.68–70  Low  Weak 
Using medium-dose GCs is recommended in the presence of fever attributed to SLE activity, expecting fever remission in 1−5 days.23,71,72  Very low  Strong 
Good clinical practice point. Infection or toxic effect of drugs must be ruled out before attributing fever to lupus activity.     

GC: glucocorticoids; SLE: systemic lupus erythematosus.

Mucocutaneous manifestations

Mucocutaneous involvement in SLE is very common and can occur in the context of patients with or without systemic involvement; it is usually classified into specific and non-specific manifestations.73 The choice of optimal treatment is difficult because most efficacy evaluations group the outcome as ‘cutaneous manifestations’ without any further distinction as to the type of manifestation.

Nevertheless, current information on this particular topic is useful for issuing a number of recommendations including management using increasingly fewer GCs, the addition of topical therapy, the use of antimalarials as a cornerstone and, if necessary, immunosuppressants and biological therapy.73–78 (Table 7)

Table 7.

Recommendations for the management of mucocutaneous manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
Localised cutaneous manifestations
Short-term medium potency topical GC preparations (e.g. fluocinolone acetonide, betamethasone valerate) as first line of treatment are recommended.73,74  Moderate  Strong 
Topical calcineurin inhibitor preparations are recommended as second-line therapy, or as a first choice for the treatment of localised lesions in skin areas at high risk of sequelae due to adverse effects of topical corticosteroid therapy (e.g. facial lesions).73,74  Moderate  Strong 
Good clinical practice point. Consider dermatology referral for patients with localised cutaneous lupus erythematosus (CLE) lesions that do not respond to the interventions outlined above. In this case using dermatological therapeutic interventions may be effective.     
Disseminated or severe cutaneous manifestations
Medium- or high-dose systemic GCs in combination with antimalarials are recommended as first-line treatment for acute, subacute, or chronic CLE lesions that are disseminated or significantly interfere with quality of life or are life-threatening.73,75,76  Moderate  Strong 
Additional use of methotrexate, mycophenolic acid (or MMF) is recommended in patients with SLE whose cutaneous manifestations have been refractory or who experience relapse despite GC and antimalarial therapy.73,77  Moderate  Strong 
The addition of belimumab or anifrolumab to treatment is suggested in SLE patients with severe skin manifestations refractory to GCs, antimalarials, and immunosuppressants.37,77–80  High  Weak 
Treatment with systemic retinoids, or alternatively dapsone, thalidomide, or lenalidomide, is suggested in patients with subacute or chronic CLE lesions who are refractory to or relapsing despite GC, antimalarial, and immunosuppressive therapy.73,75  Low  Weak 
Mucosal ulcers and alopecia
It is suggested that for the specific management of oral ulcers or ulcers in other mucosal sites, or transient diffuse alopecia due to lupus activity, the same treatment guidelines should be followed as those issued for the other cutaneous manifestations.73,75–77  Very low  Weak 
Good clinical practice point. For all manifestations, the importance of avoiding exposure to ultraviolet radiation, optimising vitamin D levels, and avoiding smoking should be emphasised.     

GC: glucocorticoids; SLE: systemic lupus erythematosus.

Musculoskeletal manifestations

Joint manifestations are common in SLE patients, occurring in up to 85% of cases in the course of the disease. Most patients present with arthralgias with inflammatory features, but a low percentage develop persistent and/or erosive arthritis. Myalgias and, in rare cases, myositis with weakness and elevated muscle enzymes may also occur, almost always with a self-limiting course. Treatment of this group of manifestations depends on their severity and includes the use of NSAIDs, GCs, disease-modifying drugs and, in some cases, biological therapy.26,81–84 (Table 8)

Table 8.

Recommendations for the management of musculoskeletal manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
The use of NSAIDs for short periods is recommended for the control of joint manifestations, assessing the risk/benefit in each patient according to comorbidities. The choice between traditional NSAID vs. selective COX-2 inhibitor will depend on each case and should be individualized.26  Moderate  Strong 
GCs are recommended for inflammatory joint manifestations refractory to NSAIDs or antimalarials. They can be used at the onset of the disease or in periods of relapse, in low doses and monitoring for dose response, and discontinued once control of inflammation and pain has been achieved. In cases of refractory monoarticular inflammation, we recommend intra-articular injection of GCs (ruling out an infectious process beforehand).81  Low  Strong 
HCQ or CLQ is recommended for the control of joint pain and inflammation.8,82  Moderate  Strong 
Methotrexate is recommended in patients with persistent joint inflammation despite NSAIDs, low-dose GCs, and antimalarials.8  Low  Strong 
Leflunomide is suggested in patients with persistent joint inflammation despite NSAIDs, low-dose GCs, and antimalarials.8,49  Low  Weak 
Belimumab is recommended in patients refractory to standard treatment, as an adjuvant treatment to achieve control of joint manifestations.84  Moderate  Strong 
Rituximab is recommended in cases refractory to previous treatment.8,26,85  Low  Strong 
Anifrolumab is suggested in cases refractory to previous treatment86–88Moderate  Weak 

CLQ: chloroquine; GCs: glucocorticoids; HCQ: hydroxychloroquine; NSAIDs: non-steroidal anti-inflammatory drugs.

Haematological manifestations

Haematological involvement in SLE is very common and occurs in more than 90% of patients.89 It affects all cell lines and is characterised by mild manifestations such as chronic leukolymphopenia, and severe and acute life-threatening conditions requiring prompt specific treatment, such as haemolytic anaemia, autoimmune thrombocytopenia, thrombotic thrombocytopenic purpura, and haemophagocytic syndrome. In addition, multifactorial disorders occur such as anaemia of chronic disease and cytopenias associated with chronic organ damage (liver or kidney), infectious events, and/or drugs.90

Under the heading of recommendations for the treatment of haematological manifestations, guidelines on the evaluation and treatment of antiphospholipid syndrome (APS) have been included in this update. Up to 30% of patients with SLE have APS or positive antibodies, and the impact of this association on patient morbidity and prognosis is well known.91–93 (Table 9)

Table 9.

Recommendations for the management of haematological manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
Thrombocytopenia
In cases of moderate thrombocytopenia without signs of bleeding, high-dose prednisone is recommended until platelet counts reach above 100,000 cells/µL and gradually tapering the GC dose until discontinuation, adding another immunosuppressant to reduce the risk of relapse.94,95  Moderate  Strong 
In cases of severe thrombocytopenia (<15,000 cells/µL) or with life-threatening signs of bleeding, the use of methylprednisolone pulses (1 g/day for 3−5 days, depending on the severity of the condition) is recommended for faster responses, making sure to continue high doses of prednisone or equivalent to avoid the risk of relapse. It is recommended that these doses be continued until the count is higher than 50,000 cells/µL and that some other immunosuppressant be added.94,96,97  Moderate  Strong 
Danazol (200−800 mg/day) and/or antimalarials are recommended as adjunct therapy to oral GC treatment.94,98,99  Moderate  Strong 
Rituximab is recommended in refractory cases.100,101  Moderate  Strong 
IV immunoglobulin is recommended only in patients with poor response to GCs.94,102–104  Moderate  Strong 
CPM is recommended for 3−6 months, depending on the severity of thrombocytopenia and clinical response, in patients who have not responded to previous therapy.105–107  Low  Strong 
MMF is suggested in patients who are refractory to previous treatment.108,109  Low  Weak 
AZA is suggested in patients who are refractory to other lines of treatment.95,110,111  Low  Weak 
Alternatively, human recombinant thrombopoietin plus dexamethasone or other GCs are recommended.94,112,113  High  Strong 
In patients with platelet counts below 10,000 cells/µL, regardless of whether or not there are signs of bleeding or a count below 50,000 cells/µL with active bleeding, the use of platelet concentrates is recommended.114  Moderate  Strong 
Splenectomy is suggested in cases refractory to the four previous lines of treatment.115,116  Low  Weak 
Autoimmune haemolytic anaemia
GCs are recommended in situations where anaemia is life-threatening; use methylprednisolone pulses, followed by high-dose oral GCs.117,118  High  Strong 
Concomitant use of AZA or danazol is recommended.119–121  Moderate  Strong 
MMF, CPM, or biological therapy (rituximab, belimumab, anifrolumab) is suggested in refractory cases.87,88,117,122–130  Low  Weak 
Packed red cell transfusion is not recommended, except in life-threatening cases, where washed red blood cells should be used.131  Moderate  Strong 
Neutropenia, leukolymphopenia
In neutropenia <1,000 cells/µL, associated with fever or infection, it is recommended to initiate granulocyte-colony-stimulating factor (300 μg/day) and continue with the minimum effective dose to achieve neutrophil counts greater than 1,000 cells /µL.89,132–136  Low  Strong 
High doses of prednisone or its equivalent are suggested.89,132  Low  Weak 
Concomitant use of AZA or MMF is suggested.132,137  Low  Weak 
Rituximab is suggested in refractory disease (375 mg/m2 body surface area for four weeks or 1 g on day zero and day 14).132,137  Very low  Weak 
Good clinical practice point. Rule out infection or drug toxicity.     
Thrombotic thrombocytopenic purpura
It is recommended to initiate plasma exchange as soon as the diagnosis is suspected (within 4−8 h) and administer until response is achieved, in combination with GCs. Exchange should be with fresh frozen plasma.138  Low  Strong 
Methylprednisolone pulses as first-line therapy (1 g/day for 5 days) are recommended, followed by oral GCs with progressive dose reduction.138,139  Low  Strong 
Second-line use of CPM, MMF, or biologicals such as rituximab, belimumab, or eculizumab is suggested in resistant cases.138,140,141  Low  Weak 
Haemophagocytic syndrome
Pulse methylprednisolone or prednisone is recommended.142,143  Low  Strong 
Concomitant addition of cyclosporine, MMF, CPM, or rituximab is suggested.144,145  Low  Weak 
Good clinical practice point. Supportive treatment (fluid resuscitation, antibiotics, covering transfusion requirements) should be provided, and search for and treat infectious foci.142     
Antiphospholipid antibody syndrome
Primary thromboprophylaxis
In any patient with antiphospholipid antibodies, treatment or correction of cardiovascular risk factors is highly recommended.146,147  Moderate  Strong 
In patients with a medium/high risk profile (presence of lupus anticoagulant, high titres of antiphospholipid antibodies, or double or triple positivity), treatment with antiplatelet doses of acetylsalicylic acid (ASA 75−100 mg/day) is recommended indefinitely, as long as not contraindicated.148  Low  Strong 
Good clinical practice point. In patients with antiphospholipid antibodies, in situations of high thrombotic risk (surgery, prolonged immobilisation, puerperium, among others), treatment with low-molecular-weight heparin at prophylactic doses should be used.     
Secondary thromboprophylaxis
For secondary prevention of venous thrombosis, indefinite anticoagulation therapy with vitamin K antagonists (VKA) to achieve an INR 2.0−3.0, is recommended.149  Moderate  Strong 
For the prevention of recurrence of arterial thrombosis, anticoagulation with VKA (INR 2.0−3.0 or 3.0−4.0 depending on individual thrombotic/haemorrhagic risk) or a combination of VKA (INR 2.0−3.0) plus ASA is recommended.150–153  Low  Strong 
In case of thrombotic recurrence (venous or arterial) despite the treatment established, it is recommended to increase the VKA dose to INR 3.0−4.0 or to add ASA if not already on this treatment. HCQ or statins may be useful.154,155  Low  Strong 
Direct oral anticoagulants (DOACs) are not recommended in patients with APS.91,149,156–163  Low  Strong 
Prudent use of DOACs is suggested only in situations of intolerance to or ineffectiveness of VKAs, and only in patients with venous thrombosis. DOACs should not be used in triple positive or arterial thrombosis.160,161,164,165  Low  Weak 

APS: antiphospholipid syndrome; AZA: azathioprine; CPM: cyclophosphamide; GCs: glucocorticoids; MMF: mycophenolate mofetil.

Renal manifestations

Renal involvement in lupus is one of the most frequent manifestations and has the greatest impact on patient prognosis. There are populations, such as the Mexican population, in which lupus nephritis presents at a younger age and is usually more severe.166,167

The optimal treatment of lupus nephritis is defined by both histological findings and clinical behaviour. In proliferative nephritis, a period of remission induction followed by prolonged maintenance therapy is still recommended, although multiple therapy may be initiated early on (e.g., MMF or CPM plus a calcineurin inhibitor), especially in patients with poor prognostic data. Recent evidence shows that this regimen may be superior to monotherapy in terms of long-term outcomes.168–170 The use of anti-Blys/BAFF biological therapy (specifically belimumab) is also suggested as an adjuvant to standard management, as this has been shown to result in longer renal survival and fewer relapses.50,171,172

Current recommendations emphasise the detection of associated poor prognostic data (e.g., antiphospholipid antibodies) and concomitant management of risk factors for progression of renal damage as an indispensable measure to improve long-term prognosis.173,174 (Table 10)

Table 10.

Recommendations for the management of renal manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
Nephritis in Mexican patients (Latino, Mesoamerican)
Remission induction therapy with MMF (2–3 g/day) or CPM (1 g/m2 body surface area) monthly is recommended in this population.166,175  Moderate  Strong 
Class I and II nephritis
Immunosuppressive therapy is suggested for class I and II nephritis with impaired renal function, active sediment, or proteinuria ≥1 g/day.167,176  Low  Weak 
Treatment with AZA, MMF (1–2 g/day), or CPM is suggested for a minimum period of six months, together with medium-dose GCs and gradual tapering.177,178  Low  Weak 
Remission induction therapy for class III/IV nephritis with proliferative component
Remission induction therapy with MMF (2–3 g/day) or CPM (monthly pulses of 1 g/m2 body surface area), combined with gradually tapered GCs, is recommended for a period of six months.168,177,178  High  Strong 
In the event of no response, multiple therapy is recommended: MMF plus tacrolimus (3–4 mg/day) and) and GCs.168.  High  Strong 
As an alternative, it is recommended to combine MMF plus voclosporin (23.7 mg/12 h).169,170  Moderate  Strong 
Adding belimumab to standard management is recommended in patients with poor prognostic factors or refractory cases.172,179  Moderate  Strong 
Adding rituximab or anifrolumab to standard management is suggested in patients with poor prognostic factors or refractory cases.50,80,171  Low  Weak 
Pulsed or high-dose oral GCs are recommended as initial treatment in addition to the immunosuppressive regimen, with gradual tapering.177,178  Moderate  Strong 
Class V nephritis without proliferative component     
MMF (2–3 g/day) or AZA (1–3 mg/kg) is recommended.177,178  High  Strong 
Tacrolimus, voclosporin, cyclosporin A, CPM, or rituximab are suggested for patients who are refractory to treatment.169,180  Moderate  Weak 
Maintenance therapy for class III/IV and V nephritis with proliferative component
MMF (1–2 g/day), AZA (1–3 mg/kg), or continuation of multiple therapy: MMF plus tacrolimus is recommended. Treatment should be long-term: at least 2−5 years.177,178,181  High  Strong 
Quarterly CPM, tacrolimus, cyclosporin A, or rituximab are suggested in patients with intolerance to MMF or AZA.177,178,182  Moderate  Weak 
Rapidly progressive nephritis/with cellular crescents
Induction therapy with monthly CPM (.750–1 g/m2 body surface area) or MMF (2–3 g/day) for six months is recommended. Both options combined with high-dose pulse methylprednisolone or prednisone, with gradual tapering depending on progression.177,178,183,184  High  Strong 
If there is no response, multiple therapy is suggested: MMF plus tacrolimus and GCs.184  Moderate  Weak 
The use of rituximab or belimumab is suggested in the treatment of refractory cases.172,185  Low  Weak 
Adjuvant management
Management to reduce renal risk factors is recommended in all patients: ACE inhibitors, ARA II, or SGLT2i, adequate control of hypertension, dyslipidaemia, hyperuricaemia, avoidance of smoking, and control of body weight.173  High  Strong 
HCQ is recommended on an ongoing basis to reduce the likelihood of renal relapse and for its benefits on dyslipidaemia.20–22  High  Strong 
It is recommended to assess for the presence of antibodies or APS and the use of antiplatelet agents or anticoagulants in patients at risk.174  High  Strong 
Treatment of relapse     
Repeat previously effective remission induction therapy is recommended.177,178  Moderate  Strong 
Management with renal replacement therapy and transplantation
In patients with end-stage chronic kidney disease due to lupus nephritis, renal transplantation is recommended as the best long-term treatment option.177,178,186  High  Strong 
Haemodialysis is recommended over peritoneal dialysis as replacement therapy in patients with end-stage chronic kidney disease due to lupus nephritis, because peritoneal dialysis has been associated with increased complications and mortality.177,178,187  Moderate  Strong 

ARA II: angiotensin II receptor antagonists; AZA: azathioprine; CPM: cyclophosphamide; GCs: glucocorticoids; ACEI: angiotensin-converting enzyme inhibitor; MMF: mycophenolate mofetil; SGLT2i: sodium-glucose cotransporter 2 inhibitors.

Neuropsychiatric manifestations

Neuropsychiatric involvement in SLE is highly variable, with frequencies reported to range from 14% to 75%.188 This wide variation in frequency is due to the difficulty in attributing neuropsychiatric manifestations with certainty to the disease, after secondary causes such as metabolic and infectious, have been ruled out.189 The American College of Rheumatology published the nomenclature for neuropsychiatric lupus syndromes,190 which includes 18 manifestations; however, virtually any manifestation can occur in SLE, whether or not accompanied by activity in another organ or system. Neuropsychiatric involvement is a major cause of mortality and cumulative damage. (Table 11)

Table 11.

Recommendations for the management of neuropsychiatric manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
General recommendations
Good clinical practice point. Other neuropsychiatric causes should be excluded before attributing manifestations to disease activity. Management of these manifestations should be assessed in conjunction with a neurologist or psychiatrist, as appropriate.     
GCs and immunosuppressive therapy are recommended for neuropsychiatric manifestations that are considered secondary to inflammatory process or activity (e.g., aseptic meningitis, myelitis, cranial and peripheral neuropathies, and psychosis).188  Moderate  Strong 
Anticoagulant therapy is recommended when manifestations are related to APS, especially in cerebral vascular events of thrombotic origin.188  Moderate  Strong 
Antiplatelet therapy is recommended when manifestations are related to APS.188  Moderate  Strong 
Cognitive dysfunction
Management of associated factors such as anxiety, depression, control of cardiovascular risk factors, as well as psychological support are suggested, as they may prevent further cognitive decline.191  Low  Weak 
Epileptic seizures
Antiepileptic drugs are recommended when there are recurrent seizures or when there are at least two episodes within the first 24 h or there is epileptogenic activity on EEG.188  Moderate  Strong 
IV methylprednisolone, followed by prednisone (1 mg/kg/day for no more than three months), tapered according to disease activity, is recommended for refractory seizures associated with SLE activity.192  Moderate  Strong 
Concomitant IV CPM (.75 g/m2 body surface area) monthly for 12 months is recommended.192  Moderate  Strong 
Peripheral neuropathy, myelopathy, and optic neuritis     
IV methylprednisolone is recommended, followed by prednisone (1 mg/kg/day), and tapering according to disease activity.192–195  Moderate  Strong 
Concomitant monthly IV CPM for 12 months is recommended.192–194  Moderate  Strong 
IV immunoglobulin (2 g/kg divided over five days) is suggested in refractory cases.193,194,196–198  Moderate  Weak 
Methylprednisolone, CPM, AZA, or rituximab is recommended in cases of extensive transverse myelitis.199,200  Moderate  Strong 
Chorea and other abnormal involuntary movements
ASA is suggested for use in antiphospholipid antibody-associated chorea and anticoagulation if there is coexisting APS.201–203  Low  Weak 
Symptomatic therapy with dopamine antagonists is recommended in case of abnormal involuntary movement.201  Moderate  Strong 
Psychosis
Prednisone 1 mg/kg/day for eight weeks with gradual tapering to 5 mg/day is recommended.204  Moderate  Strong 
Symptomatic therapy with dopamine antagonists is recommended.205  Moderate  Strong 
Concomitant monthly IV CPM for six months is recommended.204,206  Moderate  Strong 
Rituximab or plasmapheresis is suggested in refractory cases.188  Low  Weak 
Intravenous immunoglobulin is suggested if all of the above fails.188  Low  Weak 

ASA: acetylsalicylic acid; APS: antiphospholipid syndrome; AZA: azathioprine; CPM: cyclophosphamide; GCs: glucocorticoids.

Cardiopulmonary manifestations

The spectrum of cardiopulmonary manifestations in SLE patients is very diverse, ranging from mild, often subclinical conditions such as pleuritis or pericarditis, chronic manifestations with significant impact on quality of life such as interstitial pneumonitis or pulmonary arterial hypertension (PAH), to acute and life-threatening events such as diffuse alveolar haemorrhage.207,208 Timely detection of these manifestations requires a high degree of clinical suspicion and, of course, treatment will depend on the type and severity of each manifestation, ranging from symptomatic management to the use of high doses of GCs, immunosuppressants, or even biological therapies. Specific treatment recommendations are also included, for example for PAH, and the need for joint management with cardiology and pulmonology is highlighted.209 (Table 12)

Table 12.

Recommendations for the management of cardiopulmonary manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
Pericarditis
NSAIDs are suggested in cases of mild, acute, or chronic pericarditis, with or without effusion, until clinical symptoms improve.210,211  Low  Weak 
For acute or chronic pericarditis with pericardial effusion, prednisone at a dose of 1 mg/kg/day is recommended in patients with mild to moderate pericarditis as the initial manifestation. For severe or constrictive pericarditis, pulse methylprednisolone is recommended.211–215  Moderate  Strong 
In patients with new-onset or recurrent pericarditis, colchicine 1 mg in combination with GCs and immunosuppressants is suggested until remission is achieved. To avoid relapse, the addition of colchicine for at least one month is recommended.213,215,216  Moderate  Weak 
Good practice point. Surgery is indicated for treatment-resistant pericarditis or tamponade unresponsive to pharmacological treatment.     
Myocarditis
Pulse methylprednisolone is recommended for severe myocarditis with arrhythmias and/or ventricular ejection fraction of <55%, then treatment with moderate- to high-dose prednisone with progressive tapering.217  Low  Strong 
HCQ or CLQ is suggested for the maintenance stage (except in cases of prolonged QT interval).217  Low  Weak 
For severe manifestation with arrhythmias or ventricular ejection fraction <40%, CPM is suggested for 3−10 months as first-line therapy in conjunction with GCs. It is suggested response be evaluated at three months; if no improvement, discontinue to avoid risk of toxicity; if there is improvement, a minimum of six months of treatment is recommended.218  Moderate  Weak 
MMF is suggested as maintenance therapy after CPM, to reduce relapse.217  Moderate  Weak 
AZA is suggested as maintenance therapy after CPM in patients with intolerance to MMF.217  Low  Weak 
Immunoglobulin is suggested for patients with complicated myocarditis who have failed standard induction therapy with GCs and CPM.217  Low  Weak217 
Pulmonary Arterial Hypertension
High-dose GCs are recommended for four weeks with gradual tapering.219,220  Moderate  Strong 
Pulse methylprednisolone is recommended in severe cases when concurrent with other organ involvement and in relapse.221,222  Low  Strong 
CPM is recommended for 3−6 months as immunosuppressive therapy combined with GCs, vasodilators, diuretics, and other supportive measures.220,221,223  Moderate  Strong 
It is suggested to use MMF as maintenance after CPM or in case of intolerance or contraindication to CPM.222  Low  Weak 
Bosentan 62.5 mg twice daily for four weeks and follow up with 125 mg twice daily for 3–12 months in conjunction with GCs and immunosuppressants as first-line therapy in patients with NYHA functional classes II and III is recommended.224–226  High  Strong 
Additional use of sildenafil, at an initial dose of 20 mg three times daily, which can be gradually increased to 80 mg three times daily according to patient tolerability, is recommended as first-line therapy in patients with PAH and NYHA functional classes II and III.224,226  High  Strong 
It is suggested to consider its use in patients with functional class IV.224,226–228  Low  Weak 
Selexipag is suggested at doses of 200−600 mcg twice daily in patients with NYHA functional class I-II.209  High  Weak 
Good practice point. In patients with NYHA functional class III and IV, use of epoprostenol and teprostinil should be assessed individually in conjunction with the heart and lung services, as well as the use of combination therapy with vasodilators, phosphodiesterase inhibitors, and endothelin-1 antagonists.     
Pleuritis with or without pleural effusion     
NSAIDs are recommended.229,230  Moderate  Strong 
GCs are recommended when there is no therapeutic response with NSAIDs for 1−2 weeks, prednisone in medium doses and tapering within 2−3 weeks.229,230  Moderate  Strong 
Good practice point. There is insufficient evidence on the use of immunosuppressants in this context.     
Acute lupus pneumonitis
Prednisone (1 mg/kg/day) for three days and assessing clinical response is recommended; if no response, consider IV methylprednisolone for three days.231  Moderate  Strong 
It is suggested that CPM (.5–1 g/m2 body surface area) monthly for 3–6 months be considered in cases refractory to GCs.231  Low  Weak 
It is suggested that immunoglobulin be considered for cases refractory to or contraindicated for immunosuppressive therapy.230–232  Low  Weak 
Interstitial lung disease
Prednisone (.5−1 mg/kg/day) is recommended for 2−4 weeks with gradual tapering.233  Moderate  Strong 
Monthly CPM is recommended for 6−12 months in severe cases.232,233  Moderate  Strong 
Rituximab is suggested in refractory cases.232,233  Low  Weak 
Good practice point. Smoking cessation, consideration of supplemental oxygen and vaccination schedule for influenza, pneumococcal, respiratory syncytial virus, and SARS-CoV-2232 are indicated.232     
Diffuse alveolar haemorrhage
Methylprednisolone pulses are recommended.234,235  Moderate  Strong 
Monthly CPM is recommended for 6−12 months.235,236  Low  Strong 
MMF or AZA is recommended as maintenance therapy.234,236  Low  Strong 
Rituximab with CPM is recommended for refractory cases.237  Low  Strong 
Immunoglobulin is recommended for cases resistant to previous treatment.236  Low  Strong 
Plasma exchange (5–7 sessions) is suggested in patients resistant to previous treatment.235,238  Low  Weak 
It is suggested to consider recombinant factor VIIa in refractory cases.237  Low  Weak 
Good practice point. Take bronchial secretion cultures if there is suspected association with pulmonary infectious processes.236     

AZA: azathioprine; CLQ: chloroquine; CPM: cyclophosphamide; GCs: glucocorticoids; HCQ: hydroxychloroquine; MMF: mycophenolate mofetil; NSAIDs: nonsteroidal anti-inflammatory drugs; NYHA: New York Heart Association; PAH: pulmonary arterial hypertension.

Gastrointestinal manifestations, hepatitis, and pancreatitis

Gastrointestinal and hepatic manifestations are common in SLE patients. They are complex to assess and treat because they are often multifactorial. Gastrointestinal involvement varies from drug-associated dyspepsia to episodes of intestinal pseudo-obstruction and severe vasculitis.239 Hepatitis and pancreatitis especially, can be explained by disease activity, but may also be due to primary autoimmune conditions, metabolic events, infectious conditions, or drug toxicity.239–241 (Table 13)

Table 13.

Recommendations for the management of gastrointestinal manifestations.

Recommendation  Quality of evidence  Strength of recommendation 
Intestinal vasculitis
Administration of pulse methylprednisolone followed by high-dose prednisone is recommended.239,242–244  Low  Strong 
Concomitant use of GCs and CPM is recommended.239,242–244  Low  Strong 
MMF and AZA are suggested as maintenance therapy.239  Low  Weak 
Immunoglobulin IV is suggested in patients who are refractory to previous treatment (lack of response in 24−48 h) or if an associated infectious process is suspected.239  Low  Weak 
Good clinical practice point. Exploratory laparotomy is indicated in refractory cases or if there are complications.     
Intestinal pseudo-obstruction
Methylprednisolone pulses are recommended, followed by high-dose prednisone or equivalent.245–248  Low  Strong 
Addition of CPM is recommended.240,245,247  Low  Strong 
MMF or AZA is suggested as maintenance treatment.240,245  Low  Weak 
Immunoglobulin is recommended in refractory cases (lack of response within 24−48 h after GC administration).240,247  Low  Strong 
Good practice point. General measures such as prokinetics, decompression catheters, hydration, and electrolyte control should be administered in all cases, and exploratory laparotomy should be considered in refractory cases or in the event of complications.240     
Protein-losing enteropathy
High-dose GCs are recommended. Pulse methylprednisolone should be considered if the patient has severe disease complications (severe hypoalbuminemia with capillary leak and pleural or pericardial effusion, or severe hepatic involvement).1,239,242,244,249  Moderate  Strong 
Concomitant GCs and immunosuppressants are suggested.1,239,242,244,249  Low  Weak 
Rituximab is suggested in patients resistant to previous treatment.239  Low  Weak 
Good practice point. Diets high in protein and medium-chain triglycerides are useful, as well as treatment with octreotide.     
Lupus hepatitis
GCs at medium and high doses are recommended until normalization or reduction of liver enzymes <2 times their normal values.240,241,250–252  Low  Strong 
Immunosuppressants (AZA or MMF) are recommended in combination with GCs.173,240,250–252  Low  Strong 
Belimumab is suggested in patients who have failed previous treatment.253,254  Low  Weak 
Good practice point. Causes of secondary hepatitis associated with hepatic autoimmune diseases (autoimmune hepatitis, primary biliary cholangitis, primary sclerosing cholangitis, overlap syndromes); alternative liver diseases, such as liver damage from drugs, alcohol, fatty liver, vascular causes, hypercoagulable states, and viral infectious hepatitis should always be considered.244,255,256     
Lupus pancreatitis
High-dose GCs are recommended in patients with acute pancreatitis. Pulse methylprednisolone for three doses can be used in patients who do not respond to the initial dose.1,239,257  Low  Strong 
Plasma exchange is recommended for patients who are refractory to GC therapy.1,239  Moderate  Strong 
IV immunoglobulin is suggested in patients who are refractory to GCs or when an associated infectious process is suspected.239  Low  Weak 
Concomitant use of GCs and immunosuppressants is suggested.1,239  Low  Weak 
Rituximab is suggested in patients who are refractory to previous treatment.239  Low  Weak 
Good practice point. The diagnosis of lupus pancreatitis is made after excluding other causes, such as obstructive, toxic-metabolic, drug-induced, or viral. Treatment should be escalated if there is no response to GCs in 24−48 h and should include the usual supportive management of pancreatitis.     

AZA: azathioprine; CFM: cyclophosphamide; GCs: glucocorticoids; MMF: mycophenolate mofetil.

Vaccination

Current evidence is clear that most vaccines are safe and effective in lupus patients. Despite initial controversies regarding the possibility of polyclonal activation in patients upon vaccination, or doubts regarding the level of seroprotection and seroconversion, the benefit of vaccination has been shown to outweigh the risk and it is now recommended that all lupus patients have a full vaccination schedule. Ideally, vaccinations should be given before the start of immunosuppressive treatment, but when not possible they should be scheduled for the most appropriate time. A special situation is the live attenuated virus vaccines, which can be administered in patients with inactive SLE who are not receiving immunosuppressive drugs.258–261 (Table 14)

Table 14.

Recommendations on vaccination.

Recommendation  Quality of evidence  Strength of recommendation 
The following vaccines are recommended in patients with SLE: influenza (H1N1, H3N2, influenza type B),259,261–264 pneumococcal (heptavalent pneumococcal conjugate vaccine, pneumococcal conjugate vaccine PCV13 or pneumococcal polysaccharide vaccine PPSV23),258,261,265,266 SARS-CoV-2 (mRNA),267,268 herpes zoster,269–271 human papillomavirus (in people under 45 years of age).272,273  Moderate  Strong 
Vaccination against hepatitis B is suggested in patients with inactive SLE.260  Moderate  Weak 

SLE: systemic lupus erythematosus.

Perioperative period

Undertaking surgical procedures in patients with lupus requires special attention because these patients are often medically complex due to disease activity, potential organ damage, comorbidities, and immunosuppressive therapy. It has been shown that patients with lupus suffer more perioperative complications than the general population and therefore, whenever possible, surgery in these patients requires adequate planning, adjustment of treatment, and careful monitoring during and after the procedure, with a multidisciplinary approach. The following recommendations highlight the most important aspects regarding the adjustment of treatment with GCs, immunosuppressants, and biological therapies during the perioperative period.274,275 (Table 15)

Table 15.

Recommendations on perioperative management.

Recommendation  Quality of evidence  Strength of recommendation 
It is suggested to continue the same dose of methotrexate, antimalarials, and sulfasalazine during the perioperative period.274,276–278  Low  Weak 
In patients with moderate lupus undergoing elective surgery, it is recommended that MMF, AZA and/or tacrolimus be discontinued one week before surgery and restarted in the absence of infection or healing complications.274,278–280  Low  Strong 
In patients with severe lupus undergoing elective surgery, it is recommended not to discontinue MMF, AZA, and/or tacrolimus.274,278–280  Low  Strong 
In patients on biological therapy with rituximab or belimumab, it is recommended to withhold the next dose before surgery and to restart in the absence of infection or healing complications.281–284  Low  Strong 
In SLE patients receiving moderate or high doses of GCs, perioperative stress doses (50 mg hydrocortisone every 8 h) are recommended.280  Low  Strong 
In SLE patients receiving low doses of GCs, the use of stress doses is not suggested, as long as they continue their usual dose.280  Low  Weak 

AZA: azathioprine; GCs: glucocorticoids; MMF: mycophenolate mofetil; SLE: systemic lupus erythematosus.

Pregnancy

The period of pregnancy and lactation requires special attention in patients with SLE associated or not with antiphospholipid antibody syndrome. Because disease relapses are more frequent during pregnancy and there is a higher risk of complications for both mother and infant, basic recommendations for pregnancy planning, appropriate contraception, surveillance, and treatment of women during this period are included in this document.285,286 (Table 16)

Table 16.

Recommendations in pregnancy and lactation.

Recommendation  Quality of evidence  Strength of recommendation 
It is recommended that women with SLE in remission or with low disease activity, without antiphospholipid antibodies, use a highly effective contraceptive method (hormonal contraceptives or IUD) instead of less effective methods or no contraception.288–290  High  Strong 
It is recommended that women with SLE with moderate or severe disease activity or with positive antiphospholipid antibodies use progestins or IUDs as a contraceptive method.291,292  Moderate  Strong 
Preconception counselling is recommended for all women with SLE of childbearing age, especially if they are receiving potentially teratogenic drugs.285,286,293,294  Low  Strong 
It is recommended that pregnancy in a woman with SLE be planned to decrease the risk of obstetric complications.295–299  Low  Strong 
It is recommended that pregnancy be contraindicated in SLE patients with severe organ damage, such as class III-IV heart failure, moderate to severe renal disease, severe PAH, cerebrovascular event, or recent thrombosis, due to the high risk of complications for the mother and foetus.300  High  Strong 
It is recommended that a woman with SLE planning pregnancy be in remission or at least low disease activity for at least six months.301–304  Low  Strong 
It is recommended that anti-Ro and anti-La antibodies be determined before or immediately after conception to better stratify the patient's obstetric risk.305  Moderate  Strong 
Determination of antiphospholipid antibodies (lupus anticoagulant, anticardiolipin, anti-Beta-2 glycoprotein) is recommended before or immediately after conception to better stratify the patient's obstetric risk.306–311  High  Strong 
In women with pregnancy plans, switching to non-contraindicated drugs is recommended rather than discontinuing treatment.312–314  High  Strong 
Good practice point. The pregnant patient with SLE should be followed up at least once in each trimester of gestation and postpartum, including clinical and analytical data to assess disease activity.287,315–319     
Good practice point. The pregnant patient should be followed up and assessed in conjunction with specialists in maternal-foetal medicine and any others necessary for the monitoring and timely detection of pregnancy-related complications.287,317–319     

GCs: glucocorticoids.

Table 17 summarises the current evidence on the safety of the main drugs used in this disease during pregnancy and lactation. For a more extensive description of the topic refer to the CPG on the management of pregnancy in women with rheumatological autoimmune diseases.287

Table 17.

Use of drugs during pregnancy and lactation in SLE patients.

Drugs  Use during pregnancy  Use during lactation  Comments 
Non-selective non-steroidal anti-inflammatory drugs  YES  YES  Discontinue if the patient cannot conceive.Discontinue by gestational week 30 due to the risk of premature closure of the ductus arteriosus.Preferably use those with a short half-life.320 
COX-2 inhibitors320  NO  NO   
Glucocorticoids  YES  YES  Prednisone equivalent <20 mg/day is relatively safe, but lower doses and a GC-sparing drug should be used whenever possible.If the dose is >20 mg/day, lactation should be delayed four hours.321 
Chloroquine/hydroxychloroquine314  YES  YES   
Azathioprine/6-mercaptopurine287,319,322  YES  YES   
Methotrexate  NO  NO  Discontinue three months before attempting pregnancy.287,319,321 
Leflunomide  NO  NO  Discontinue 24 months before attempting conception or attempt cholestyramine washout and determine serum levels.287,319,321 
Danazol  NO  NO  Discontinue 1−3 months before attempting pregnancy.319 
Thalidomide  NO  NO  Discontinue 1−3 months before attempting pregnancy.319 
Mycophenolate mofetil  NO  NO  Discontinue three months before attempting pregnancy.287,319,323 
Cyclophosphamide  NO  NO  Discontinue three months before attempting pregnancy.Consider use in the second and third trimester of pregnancy if the patient has a life-threatening condition.287,319,321 
Cyclosporine A287,319  YES  YES   
Tacrolimus319,321  YES  YES   
Vitamin K antagonists  NO  YES  Substitute for heparin during pregnancy.287,319 
Unfractionated heparin/low molecular weight heparin319,323  YES  YES   
Rituximab  NO  NO  Consider if patient has no other treatment option.Drug levels in breast milk are low, and therefore exposure in the infant is similarly low. 319 
Belimumab  NO  NO  Consider if the patient has no other treatment option.Drug levels in breast milk are low, and therefore exposure in the infant is similarly low.324 
Anifrolumab  NO  NO  No information available. 

IUD: intrauterine device; PAH: pulmonary arterial hypertension; SLE: systemic lupus erythematosus.

Cardiovascular risk

Patients with SLE are known to have an increased cardiovascular and cerebrovascular risk, which may be more than double that of the general population and is one of the main determinants of long-term morbidity and mortality. This increased risk is explained by a multifactorial process of accelerated atherosclerosis involving endothelial dysfunction, autoantibody-induced damage, chronic inflammation, changes in lipid and lipoprotein profile, increases in traditional risk factors and even adverse effects of GC and NSAID therapy.325–327 Therefore, in this update of lupus treatment recommendations, it was considered essential to include guidelines on the best methods of assessment, prevention, and treatment of cerebrovascular and cardiovascular events. (Table 18)

Table 18.

Recommendations to reduce cardiovascular risk.

Recommendation  Quality of evidence  Strength of recommendation 
It is recommended that cardiovascular risk be assessed based on the risk calculators used for the general population in all patients with SLE.325–330  Moderate  Strong 
In patients with SLE and moderate cardiovascular risk according to the risk calculators, it is recommended that the approach to identifying subclinical atherosclerosis be complemented by coronary artery calcium scoring (CAC) or carotid US.331,332  Weak  Strong 
Acetylsalicylic acid is recommended for primary prevention in patients with SLE and positive for antiphospholipid antibodies.333–337  Low  Strong 
Statins are suggested to reduce progression in patients with SLE and evidence of subclinical atherosclerosis (identified by CAC or carotid US).331,332,338–340  High  Weak 

SLE: systemic lupus erythematosus; US: ultrasound.

Discussion

The treatment of a disease as complex as SLE undoubtedly benefits from treatment guidelines that guide rheumatologists and other treating physicians on the best management options for patients with a personalised approach.

Given the advance of knowledge and the emergence of new therapeutic options for the treatment of SLE, this new version of the Mexican College of Rheumatology guidelines is the result of the collaborative work of a large group of rheumatologists with expertise in the field who, after systematically reviewing the most recent evidence in the literature, issue recommendations that update the therapeutic guidelines for the manifestations in the different organs and systems affected by the disease based on the most recent knowledge.

By including in these recommendations general principles on the management of the disease, the role of the rheumatologist as a leader in the treatment of these patients is highlighted, the general concepts that should govern treatment, always having a treat-to-target (T2T) strategy, and the importance of the treatment being chosen in agreement with the patient considering the individual benefit/risk profile.

The importance of the use of antimalarials is highlighted as long as there is no contraindication as a strategy to control the disease, avoid relapses, spare GCs, and reduce cardiovascular risk. There is a clear need to use GCs more rationally, with appropriate doses at baseline according to the manifestation, and tapering as quickly as possible, due to evidence of an association between steroid use and long-term damage.

The timely use of disease course modifiers or immunosuppressants indicated according to the different manifestations of the condition, or even multiple therapy in the case of lupus nephritis, is increasingly evident in the literature. This strategy allows for better and faster control of disease activity, and spares GCs. Recommendations on the use of new drugs such as voclosporin and others, which have been shown to be effective in the treatment of lupus, have been included.

The role of biological therapy, in addition to standard treatment and even in the future as a first line of treatment, for various manifestations of lupus is now clearer, and recommendations for belimumab, anifrolumab, and rituximab have been updated.

The update addresses special aspects of lupus treatment such as comprehensive treatment during pregnancy and lactation, as well as safe medications during this period, therapeutic adjustment during the perioperative period and current indications for vaccination.

Finally, the increased cardiovascular risk of lupus patients is well known, explained by an accelerated atherosclerosis process caused by the inflammatory phenomenon of the disease itself, traditional risk factors, and as a side effect of GC use. Therefore, general recommendations to address cardiovascular risk have been included.

These updated SLE treatment guidelines are intended to be a valuable and useful tool not only for specialist physicians in making treatment decisions for individual patients, but also as guidance for first-contact physicians or physicians in other specialties who sometimes have to initiate or modify treatments, as well as refer patients to rheumatology specialists. It is certainly desirable that they also form the basis for implementing healthcare policies for SLE patients at institutional and governmental level.

Of course, there are still areas of opportunity and rheumatologists must be prepared to address them. New evidence regarding disease diagnosis, markers of therapeutic response, and the design of pharmacological strategies to achieve remission, the latter a dynamic and rapidly changing definition, combined with the growing role of artificial intelligence in data analysis and drug design, portend an interesting and complex future for patients and specialists, a challenge that we must take up together.

Funding

Unrestricted funding for these guidelines was received from Astra Zeneca and GlaxoSmithKline.

Acknowledgements

We would like to thank Dr. Marcela Pérez Rodríguez and Dr. Ivonne Roy García for their coordination of the methodological aspect of these guidelines.

References
[1]
D. Xibillé-Friedmann, M. Pérez-Rodríguez, S. Carrillo-Vázquez, et al.
Guía de práctica clínica para el manejo del lupus eritematoso sistémico propuesta por el Colegio Mexicano de Reumatología.
Reumatol Clin., 15 (2019), pp. 3-20
[2]
G. Guyatt, A.D. Oxman, E.A. Akl, et al.
GRADE guidelines: 1. Introduction—GRADE evidence profiles and summary of findings tables.
J Clin Epidemiol., 64 (2011), pp. 383-394
[3]
A. Fanouriakis, M. Kostopoulou, J. Andersen, et al.
EULAR recommendations for the management of systemic lupus erythematosus: 2023 update.
Ann Rheum Dis., 83 (2024), pp. 15-29
[4]
T. Morrison, E. Foster, J. Dougherty, J. Barton.
Shared decision making in rheumatology: a scoping review.
Semin Arthritis Rheum., 56 (2022),
[5]
M.F. Ugarte-Gil, C. Mendoza-Pinto, C. Reátegui-Sokolova, et al.
Achieving remission or low disease activity is associated with better outcomes in patients with systemic lupus erythematosus: a systematic literature review.
[6]
R.F. van Vollenhoven, G. Bertsias, A. Doria, et al.
2021 DORIS definition of remission in SLE: final recommendations from an international task force.
[7]
L. Durcan, T. O’Dwyer, M. Petri.
Management strategies and future directions for systemic lupus erythematosus in adults.
Lancet., 393 (2019), pp. 2332-2343
[8]
B.A. Pons-Estel, E. Bonfa, E.R. Soriano, et al.
First Latin American clinical practice guidelines for the treatment of systemic lupus erythematosus: Latin American Group for the Study of Lupus (GLADEL, Grupo Latino Americano de Estudio del Lupus)-Pan-American League of Associations of Rheumatology (PANLAR).
Ann Rheum Dis., 77 (2018), pp. 1549-1557
[9]
Y.M. Chen, W.T. Hung, Y.W. Liao, et al.
Combination immunosuppressant therapy and lupus nephritis outcome: a hospital-based study.
Lupus., 28 (2019), pp. 658-666
[10]
M. Gatto, M. Zen, L. Iaccarino, A. Doria.
New therapeutic strategies in systemic lupus erythematosus management.
Nat Rev Rheumatol., 15 (2019), pp. 30-48
[11]
A. Kugasia, N. Sehgal, M. Dollear, W. Sequeira, J.A. Block, M. Jolly.
Practice patterns in longitudinal lupus care provision: patient and physician perspectives.
Lupus, 26 (2017), pp. 1556-1561
[12]
R. Ríos-Garcés, G. Espinosa, R. van Vollenhoven, R. Cervera.
Treat to target in systemic lupus erythematosus.
Rev Colomb Reumatol., 28 (2021), pp. 101-106
[13]
E.F. Morand, T. Trasieva, A. Berglind, G.G. Illei, R. Tummala.
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., 77 (2018), pp. 706-713
[14]
K. Tselios, D.D. Gladman, Z. Touma, J. Su, N. Anderson, M.B. Urowitz.
Clinical remission and low disease activity outcomes over 10 years in systemic lupus erythematosus.
Arthritis Care Res (Hoboken)., 71 (2019), pp. 822-828
[15]
W.D. Abdul Kadir, A. Jamil, S.S. Shaharir, N. Md Nor, A.H. Abdul Gafor.
Photoprotection awareness and practices among patients with systemic lupus erythematosus and its association with disease activity and severity.
Lupus., 27 (2018), pp. 1287-1295
[16]
K. Chanprapaph, M. Ploydaeng, K. Pakornphadungsit, T. Mekwilaiphan, V. Vachiramon, S. Kanokrungsee.
The behavior, attitude, and knowledge towards photoprotection in patients with cutaneous/systemic lupus erythematosus: a comparative study with 526 patients and healthy controls.
Photochem Photobiol Sci., 19 (2020), pp. 1201-1210
[17]
G.C. Drosos, D. Vedder, E. Houben, et al.
EULAR recommendations for cardiovascular risk management in rheumatic and musculoskeletal diseases, including systemic lupus erythematosus and antiphospholipid syndrome.
Ann Rheum Dis., 81 (2022), pp. 768-779
[18]
A. Kuhn, K. Gensch, M. Haust, 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., 64 (2011), pp. 37-48
[19]
K. Franklyn, C.S. Lau, S.V. Navarra, et al.
Definition and initial validation of a Lupus Low Disease Activity State (LLDAS).
Ann Rheum Dis., 75 (2016), pp. 1615-1621
[20]
Y. Molad, A. Gorshtein, A.J. Wysenbeek, et al.
Protective effect of hydroxychloroquine in systemic lupus erythematosus.
Lupus., 11 (2002), pp. 356-361
[21]
B.J. Fessler, G.S. Alarcón, G. McGwin, et al.
Systemic lupus erythematosus in three ethnic groups: XVI. Association of hydroxychloroquine use with reduced risk of damage accrual.
Arthritis Rheum., 52 (2005), pp. 1473-1480
[22]
G.J. Pons-Estel, G.S. Alarcón, L. Hachuel, et al.
Anti-malarials exert a protective effect while Mestizo patients are at increased risk of developing SLE renal disease: data from a Latin-American cohort.
Rheumatology., 51 (2012), pp. 1293-1298
[23]
F. Buttgereit.
Standardised nomenclature for glucocorticoid dosages and glucocorticoid treatment regimens: current questions and tentative answers in rheumatology.
Ann Rheum Dis., 61 (2002), pp. 718-722
[24]
M. Mosca, C. Tani, L. Carli, S. Bombardieri.
Glucocorticoids in systemic lupus erythematosus.
Clin Exp Rheumatol., 29 (2011), pp. S126-9
[25]
N. Bello, J.A. Birt, J. Workman, X. Zhou, J.A. Ross-Terres, M. Petri.
Treatment patterns and clinical characteristics of patients with systemic lupus erythematosus and musculoskeletal symptoms: a retrospective, observational study.
Adv Ther., 39 (2022), pp. 3131-3145
[26]
D.D. Hill, A.M. Eudy, P.J. Egger, Q. Fu, M.A. Petri.
Impact of systemic lupus erythematosus disease activity, hydroxychloroquine and NSAID on the risk of subsequent organ system damage and death: analysis in a single US medical centre.
[27]
B.B. Yang, H. Xiao, X.J. Li, M. Zheng.
Safety and efficacy of etanercept-methotrexate combination therapy in patients with rhupus: an observational study of non-glucocorticoid treatment for rheumatic diseases.
Discov Med., 25 (2018), pp. 14-20
[28]
S.C. Palmer, D.J. Tunnicliffe, D. Singh-Grewal, et al.
Induction and maintenance immunosuppression treatment of proliferative lupus nephritis: a network meta-analysis of randomized trials.
Am J Kidney Dis., 70 (2017), pp. 324-336
[29]
M. Tian, X. Song, L. Dong, X. Xin, J. Dong.
Systematic evaluation of different doses of cyclophosphamide induction therapy for lupus nephritis.
[30]
Q. Yuan, X. Xing, Z. Lu, X. Li.
Clinical characteristics and risk factors of infection in patients with systemic lupus erythematosus: a systematic review and meta-analysis of observational studies.
Semin Arthritis Rheum., 50 (2020), pp. 1022-1039
[31]
Y.H. Lee, G.G. Song.
A network meta-analysis of randomized controlled trials comparing the effectiveness and safety of voclosporin or tacrolimus plus mycophenolate mofetil as induction treatment for lupus nephritis.
Z Rheumatol., 82 (2023), pp. 580-586
[32]
B.H. Rovin, N. Solomons, W.F. Pendergraft, et al.
A randomized, controlled double-blind study comparing the efficacy and safety of dose-ranging voclosporin with placebo in achieving remission in patients with active lupus nephritis.
Kidney Int., 95 (2019), pp. 219-231
[33]
G.G. Song, Y.H. Lee.
Comparison of treatment response and serious infection using tacrolimus, tacrolimus with mycophenolate mofetil, in comparison to cyclophosphamide as induction treatment for lupus nephritis.
Int J Clin Pharmacol Ther., 58 (2020), pp. 550-556
[34]
J. Deng, L. Luo, L. Zhu, H. Xie.
Multitarget therapy versus intravenous cyclophosphamide in the induction treatment of lupus nephritis: a metaanalysis of randomized controlled trials.
Turk J Med Sci., 48 (2018), pp. 901-910
[35]
A. Jorge, X. Fu, C. Cook, et al.
Kidney transplantation and cardiovascular events among patients with end-stage renal disease due to lupus nephritis: a nationwide cohort study.
Arthritis Care Res (Hoboken)., 74 (2022), pp. 1829-1834
[36]
T. Maslen, I.N. Bruce, D. D’Cruz, et al.
Efficacy of belimumab in two serologically distinct high disease activity subgroups of patients with systemic lupus erythematosus: post-hoc analysis of data from the phase III programme.
[37]
E.F. Morand, G. Abreu, R.A. Furie, V. Golder, R. Tummala.
Lupus low disease activity state attainment in the phase 3 TULIP trials of anifrolumab in active systemic lupus erythematosus.
Ann Rheum Dis., 82 (2023), pp. 639-645
[38]
H. Zhang, J. Chen, Y. Zhang, N. Zhao, D. Xu.
Efficacy and safety of belimumab therapy in lupus nephritis: a systematic review and meta-analysis.
[39]
Y. Tanaka, S. Nakayamada, K. Yamaoka, K. Ohmura, S. Yasuda.
Rituximab in the real-world treatment of lupus nephritis: a retrospective cohort study in Japan.
Mod Rheumatol., 33 (2023), pp. 145-153
[40]
E. Krustev, A.E. Clarke, M.R.W. Barber.
B cell depletion and inhibition in systemic lupus erythematosus.
Expert Rev Clin Immunol., 19 (2023), pp. 55-70
[41]
Y. Atisha-Fregoso, S. Malkiel, K.M. Harris, et al.
Phase II randomized trial of rituximab plus cyclophosphamide followed by belimumab for the treatment of lupus nephritis.
Arthritis Rheumatol., 73 (2021), pp. 121-131
[42]
P. Richter, A. Cardoneanu, A.M. Burlui, et al.
Why do we need JAK inhibitors in systemic lupus erythematosus?.
Int J Mol Sci., 23 (2022),
[43]
A. González-García, I. Cusácovich, G. Ruiz-Irastorza.
Treatment of systemic lupus erythematosus: new therapeutic options.
Rev Clin Esp., 223 (2023), pp. 629-639
[44]
C. Lin, Y. Lee, P. Shih, J.C. Wei.
From concept to reality: CAR‐T therapy as a glimmer of hope for systemic lupus erythematosus patients.
Int J Rheum Dis., 26 (2023), pp. 2137-2140
[45]
J.N. Hoes, J.W.G. Jacobs, M. Boers, et al.
EULAR evidence-based recommendations on the management of systemic glucocorticoid therapy in rheumatic diseases.
Ann Rheum Dis., 66 (2007), pp. 1560-1567
[46]
M.F. Marmor, U. Kellner, T.Y.Y. Lai, R.B. Melles, W.F. Mieler, American Academy of Ophthalmology.
Recommendations on screening for chloroquine and hydroxychloroquine retinopathy (2016 Revision).
Ophthalmology, 123 (2016), pp. 1386-1394
[47]
Canadian Hydroxychloroquine Study Group.
A randomized study of the effect of withdrawing hydroxychloroquine sulfate in systemic lupus erythematosus.
New England Journal of Medicine., 324 (1991), pp. 150-154
[48]
Y.S. Goker, C. Ucgul Atılgan, K. Tekin, et al.
The validity of optical coherence tomography angiography as a screening test for the early detection of retinal changes in patients with hydroxychloroquine therapy.
Curr Eye Res., 44 (2019), pp. 311-315
[49]
J.M. Pego‐Reigosa, T. Cobo‐Ibáñez, J. Calvo‐Alén, et al.
Efficacy and safety of nonbiologic immunosuppressants in the treatment of nonrenal systemic lupus erythematosus: a systematic review.
Arthritis Care Res (Hoboken)., 65 (2013), pp. 1775-1785
[50]
K. Li, Y. Yu, Y. Gao, F. Zhao, Z. Liang, J. Gao.
Comparative effectiveness of rituximab and common induction therapies for lupus nephritis: a systematic review and network meta-analysis.
[51]
K. Ejaz, D. Abid, P. Juneau, J. Chu, S. Hasni.
Use of gonadotropin-releasing hormone agonists for ovarian preservation in patients receiving cyclophosphamide for systemic lupus erythematosus: a meta-analysis.
Lupus., 31 (2022), pp. 1706-1713
[52]
V.R. Pimentel-Quiroz, M.F. Ugarte-Gil, G. Harvey, et al.
Factors predictive of serious infections over time in systemic lupus erythematosus patients: data from a multi-ethnic, multi-national, Latin American lupus cohort.
Lupus., 28 (2019), pp. 1101-1110
[53]
M. García-Carrasco, C. Mendoza-Pinto, A. Rojas-Villarraga, et al.
Prevalence of cervical HPV infection in women with systemic lupus erythematosus: A systematic review and meta-analysis.
Autoimmun Rev., 18 (2019), pp. 184-191
[54]
Y. Jiang, Y. Cheng, S. Ma, et al.
Systemic lupus erythematosus-complicating immune thrombocytopenia: from pathogenesis to treatment.
[55]
Shin J. Il, K.H. Lee, S. Park, et al.
Systemic lupus erythematosus and lung involvement: a comprehensive review.
J Clin Med., 11 (2022), pp. 6714
[56]
L. Kawka, A. Schlencker, P. Mertz, T. Martin, L. Arnaud.
Fatigue in systemic lupus erythematosus: an update on its impact, determinants and therapeutic management.
[57]
E.J.F. Santos, B. Farisogullari, E. Dures, R. Geenen, P.M. Machado.
EULAR taskforce on recommendations for the management of fatigue in people with inflammatory rheumatic diseases. Efficacy of non-pharmacological interventions: a systematic review informing the 2023 EULAR recommendations for the management of fatigue in people with inflammatory rheumatic and musculoskeletal diseases.
[58]
E. Dures, B. Farisoğulları, E.J.F. Santos, et al.
2023 EULAR recommendations for the management of fatigue in people with inflammatory rheumatic and musculoskeletal diseases.
[59]
M.K. Youssef.
Effect of training on health outcome including fatigue, depression and quality of life in patients with systemic lupus erythromatosus.
Beni Suef Univ J Basic Appl Sci., 10 (2021), pp. 90
[60]
M. Youseff.
Efffect of excercises training on fatige, depression and physical activity in patients with systemic lupus erythematosus.
J Health Medical Res., 1 (2019), pp. 4-11
[61]
B. Gavilán-Carrera, J.A. Vargas-Hitos, P. Morillas-de-Laguno, et al.
Effects of 12-week aerobic exercise on patient-reported outcomes in women with systemic lupus erythematosus.
Disabil Rehabil., 44 (2022), pp. 1863-1871
[62]
S. Frade, S. O’Neill, D. Greene, E. Nutter, M. Cameron.
Exercise as adjunctive therapy for systemic lupus erythematosus.
Cochrane Database Syst Rev., 4 (2023),
[63]
L. Vandenbulcke, M. Erard, D. Van Assche, E. De Langhe.
The effect of physical exercise on fatigue in systemic lupus erythematosus: a systematic review.
Acta Clin Belg., 78 (2023), pp. 342-357
[64]
H. Kankaya, A. Karadakovan.
Effects of web-based education and counselling for patients with systemic lupus erythematosus: self-efficacy, fatigue and assessment of care.
Lupus., 29 (2020), pp. 884-891
[65]
F. Khan, N. Granville, R. Malkani, Y. Chathampally.
Health-related quality of life improvements in systemic lupus erythematosus derived from a digital therapeutic plus tele-health coaching intervention: randomized controlled pilot trial.
J Med Internet Res., 22 (2020),
[66]
A. Doria, W. Stohl, A. Schwarting, et al.
Efficacy and safety of subcutaneous belimumab in anti-double-stranded DNA-positive, hypocomplementemic patients with systemic lupus erythematosus.
Arthritis Rheumatol., 70 (2018), pp. 1256-1264
[67]
V. Strand, P. Berry, X. Lin, Y. Asukai, R. Punwaney, S. Ramachandran.
Long-term impact of belimumab on health-related quality of life and fatigue in patients with systemic lupus erythematosus: six years of treatment.
Arthritis Care Res (Hoboken)., 71 (2019), pp. 829-838
[68]
G.L. Lima, J. Paupitz, N.E. Aikawa, L. Takayama, E. Bonfa, R.M.R. Pereira.
Vitamin D supplementation in adolescents and young adults with juvenile systemic lupus erythematosus for improvement in disease activity and fatigue scores: a randomized, double-blind, placebo-controlled trial.
Arthritis Care Res (Hoboken)., 68 (2016), pp. 91-98
[69]
G. Ruiz-Irastorza, S. Gordo, N. Olivares, M.V. Egurbide, C. Aguirre.
Changes in vitamin D levels in patients with systemic lupus erythematosus: Effects on fatigue, disease activity, and damage.
Arthritis Care Res (Hoboken)., 62 (2010), pp. 1160-1165
[70]
R. Magro, C. Saliba, L. Camilleri, C. Scerri, A.A. Borg.
Vitamin D supplementation in systemic lupus erythematosus: relationship to disease activity, fatigue and the interferon signature gene expression.
BMC Rheumatol., 5 (2021), pp. 53
[71]
W. Zhou, C. Yang.
The causes and clinical significance of fever in systemic lupus erythematosus: a retrospective study of 487 hospitalised patients.
Lupus., 18 (2009), pp. 807-812
[72]
B.H. Rovin, Y. Tang, J. Sun, et al.
Clinical significance of fever in the systemic lupus erythematosus patient receiving steroid therapy.
Kidney Int., 68 (2005), pp. 747-759
[73]
Q. Lu, H. Long, S. Chow, et al.
Guideline for the diagnosis, treatment and long-term management of cutaneous lupus erythematosus.
[74]
S. Jessop, D.A. Whitelaw, M.J. Grainge, P. Jayasekera.
Drugs for discoid lupus erythematosus.
Cochrane Database Syst Rev., 2017 (2017),
[75]
F. Chasset, J.D. Bouaziz, N. Costedoat-Chalumeau, C. Francès, L. Arnaud.
Efficacy and comparison of antimalarials in cutaneous lupus erythematosus subtypes: a systematic review and meta-analysis.
Br J Dermatol., 177 (2017), pp. 188-196
[76]
N. Yokogawa, H. Eto, A. Tanikawa, et al.
Effects of hydroxychloroquine in patients with cutaneous lupus erythematosus: a multicenter, double‐blind, randomized, parallel‐group trial.
Arthritis Rheumatol., 69 (2017), pp. 791-799
[77]
J.L. Fairley, S. Oon, A.M. Saracino, M. Nikpour.
Management of cutaneous manifestations of lupus erythematosus: a systematic review.
Semin Arthritis Rheum., 50 (2020), pp. 95-127
[78]
L. Iaccarino, S. Bettio, R. Reggia, et al.
Effects of belimumab on flare rate and expected damage progression in patients with active systemic lupus erythematosus.
Arthritis Care Res (Hoboken)., 69 (2017), pp. 115-123
[79]
R. Kneeland, D. Montes, J. Endo, B. Shields, C.M. Bartels, S. Garg.
Improvement in cutaneous lupus erythematosus after twenty weeks of belimumab use: a systematic review and meta‐analysis.
Arthritis Care Res (Hoboken)., 75 (2023), pp. 1838-1848
[80]
E.F. Morand, R.A. Furie, I.N. Bruce, et al.
Efficacy of anifrolumab across organ domains in patients with moderate-to-severe systemic lupus erythematosus: a post-hoc analysis of pooled data from the TULIP-1 and TULIP-2 trials.
Lancet Rheumatol., 4 (2022), pp. e282-e292
[81]
M. Yates.
Commonly used medication for Lupus.
[82]
J.T. Rosenbaum, K.H. Costenbader, J. Desmarais, et al.
American College of Rheumatology, American Academy of Dermatology, Rheumatologic Dermatology Society, and American Academy of Ophthalmology 2020 Joint Statement on Hydroxychloroquine Use With Respect to Retinal Toxicity.
Arthritis Rheumatol, 73 (2021), pp. 908-911
[83]
B.A. Pons-Estel, E. Bonfa, E.R. Soriano, et al.
First Latin American clinical practice guidelines for the treatment of systemic lupus erythematosus: Latin American Group for the Study of Lupus (GLADEL, Grupo Latino Americano de Estudio del Lupus )–Pan-American League of Associations of Rheumatology (PANLAR).
Ann Rheum Dis., 77 (2018), pp. 1549-1557
[84]
F. Ceccarelli, E. Cipriano, F. Natalucci, et al.
Belimumab is able to induce a significant improvement of joint activity status in patients diagnosed with systemic lupus erythematosus: results from a 12-month longitudinal study.
Isr Med Assoc J., 22 (2020), pp. 415-419
[85]
A. Fanouriakis, N. Tziolos, G. Bertsias, D.T. Boumpas.
Update on the diagnosis and management of systemic lupus erythematosus.
Ann Rheum Dis., 80 (2021), pp. 14-25
[86]
J.T. Merrill, R. Furie, V.P. Werth, et al.
Anifrolumab effects on rash and arthritis: impact of the type I interferon gene signature in the phase IIb MUSE study in patients with systemic lupus erythematosus.
[87]
R.A. Furie, E.F. Morand, I.N. Bruce, et al.
Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial.
Lancet Rheumatol., 1 (2019), pp. e208-e219
[88]
E.F. Morand, R. Furie, Y. Tanaka, et al.
Trial of anifrolumab in active systemic lupus erythematosus.
N Engl Jf Med., 382 (2020), pp. 211-221
[89]
K. Newman, M.B. Owlia, I. El-Hemaidi, M. Akhtari.
Management of immune cytopenias in patients with systemic lupus erythematosus — Old and new.
Autoimmun Rev., 12 (2013), pp. 784-791
[90]
A. Fayyaz, A. Igoe, B.T. Kurien, et al.
Haematological manifestations of lupus.
[91]
S.V. Porta, D.C.O. de Andrade, D. Erkan, et al.
Controversies in the management of antiphospholipid syndrome.
J Clin Rheumatol., 29 (2023), pp. e107-e112
[92]
O. Unlu, S. Zuily, D. Erkan.
The clinical significance of antiphospholipid antibodies in systemic lupus erythematosus.
Eur J Rheumatol., 3 (2016), pp. 75-84
[93]
L. Riancho-Zarrabeitia, V. Martínez-Taboada, I. Rúa-Figueroa, et al.
Antiphospholipid syndrome (APS) in patients with systemic lupus erythematosus (SLE) implies a more severe disease with more damage accrual and higher mortality.
Lupus., 29 (2020), pp. 1556-1565
[94]
M.P. Lambert, T.B. Gernsheimer.
Clinical updates in adult immune thrombocytopenia.
Blood., 129 (2017), pp. 2829-2835
[95]
C. Arnal, J.C. Piette, J. Léone, et al.
Treatment of severe immune thrombocytopenia associated with systemic lupus erythematosus: 59 cases.
J Rheumatol., 29 (2002), pp. 75-83
[96]
S. Eyanson, M.H. Passo, M.A. Aldo-Benson, M.D. Benson.
Methylprednisolone pulse therapy for nonrenal lupus erythematosus.
Ann Rheum Dis., 39 (1980), pp. 377-380
[97]
D.P. Lurie, M.B. Kahaleh.
Pulse corticosteroid therapy for refractory thrombocytopenia in systemic lupus erythematosus.
J Rheumatol., 9 (1982), pp. 311-314
[98]
P. Letchumanan, J. Thumboo.
Danazol in the treatment of systemic lupus erythematosus: a qualitative systematic review.
Semin Arthritis Rheum., 40 (2011), pp. 298-306
[99]
S.G. West.
Danazol for the treatment of refractory autoimmune thrombocytopenia in systemic lupus erythematosus.
Ann Intern Med., 108 (1988), pp. 703
[100]
C. Lindholm, K. Börjesson-Asp, K. Zendjanchi, A.C. Sundqvist, A. Tarkowski, M. Bokarewa.
Longterm clinical and immunological effects of anti-CD20 treatment in patients with refractory systemic lupus erythematosus.
J Rheumatol., 35 (2008), pp. 826-833
[101]
H. Chen, W. Zheng, J. Su, et al.
Low-dose rituximab therapy for refractory thrombocytopenia in patients with systemic lupus erythematosus--a prospective pilot study.
Rheumatology., 50 (2011), pp. 1640-1644
[102]
E.J. ter Borg, C.G. Kallenberg.
Treatment of severe thrombocytopenia in systemic lupus erythematosus with intravenous gammaglobulin.
Ann Rheum Dis., 51 (1992), pp. 1149-1151
[103]
W.P. Maier, D.S. Gordon, R.F. Howard, et al.
Intravenous immunoglobulin therapy in systemic lupus erythematosus-associated thrombocytopenia.
Arthritis Rheum., 33 (1990), pp. 1233-1239
[104]
G. Zandman-Goddard, Y. Levy, Y. Shoenfeld.
Intravenous immunoglobulin therapy and systemic lupus erythematosus.
Clin Rev Allergy Immunol., 29 (2005), pp. 219-228
[105]
B.A. Roach, G.J. Hutchinson.
Treatment of refractory, systemic lupus erythematosus-associated thrombocytopenia with intermittent low-dose intravenous cyclophosphamide.
Arthritis Rheum., 36 (1993), pp. 682-684
[106]
D.T. Boumpas, S. Barez, J.H. Klippel, J.E. Balow.
Intermittent cyclophosphamide for the treatment of autoimmune thrombocytopenia in systemic lupus erythematosus.
Ann Intern Med., 112 (1990), pp. 674-677
[107]
K. Takada, G.G. Illei, D.T. Boumpas.
Cyclophosphamide for the treatment of systemic lupus erythematosus.
Lupus., 10 (2001), pp. 154-161
[108]
M. Dall’Era.
Mycophenolate mofetil in the treatment of systemic lupus erythematosus.
Curr Opin Rheumatol., 23 (2011), pp. 454-458
[109]
S. Vasoo, J. Thumboo, K.Y. Fong.
Refractory immune thrombocytopenia in systemic lupus erythematosus: response to mycophenolate mofetil.
Lupus., 12 (2003), pp. 630-632
[110]
K.M. Goebel, W.D. Gassel, F.D. Goebel.
Evaluation of azathioprine in autoimmune thrombocytopenia and lupus erythematosus.
Scand J Haematol., 10 (1973), pp. 28-34
[111]
M. Abu-Shakra, Y. Shoenfeld.
Azathioprine therapy for patients with systemic lupus erythematosus.
Lupus., 10 (2001), pp. 152-153
[112]
M.C. Maroun, R. Ososki, J.C. Andersen, J.P. Dhar.
Eltrombopag as steroid sparing therapy for immune thrombocytopenic purpura in systemic lupus erythematosus.
Lupus., 24 (2015), pp. 746-750
[113]
P. Scheinberg, C.C. Singulane, L.S.G. Barbosa, M. Scheinberg.
Successful platelet count recovery in lupus-associated thrombocytopenia with the thrombopoietin agonist eltrombopag.
Clin Rheumatol., 33 (2014), pp. 1347-1349
[114]
L.J. Estcourt, J. Birchall, S. Allard, et al.
Guidelines for the use of platelet transfusions.
Br J Haematol., 176 (2017), pp. 365-394
[115]
W.W. Coon.
Splenectomy for cytopenias associated with systemic lupus erythematosus.
Am J Surg., 155 (1988), pp. 391-394
[116]
S.J. Rivero, M. Alger, D. Alarcón-Segovia.
Splenectomy for hemocytopenia in systemic lupus erythematosus. A controlled appraisal.
Arch Intern Med., 139 (1979), pp. 773-776
[117]
E. Gomard-Mennesson, M. Ruivard, M. Koenig, et al.
Treatment of isolated severe immune hemolytic anaemia associated with systemic lupus erythematosus: 26 cases.
Lupus., 15 (2006), pp. 223-231
[118]
S.I.G. Kokori, J.P.A. Ioannidis, M. Voulgarelis, A.G. Tzioufas, H.M. Moutsopoulos.
Autoimmune hemolytic anemia in patients with systemic lupus erythematosus.
Am J Med., 108 (2000), pp. 198-204
[119]
C.C. Corley, H.E. Lessner, W.E. Larsen.
Azathioprine therapy of? autoimmune? diseases.
Am J Med., 41 (1966), pp. 404-412
[120]
Y.S. AHN.
Danazol therapy for autoimmune hemolytic anemia.
Ann Intern Med., 102 (1985), pp. 298
[121]
R. Blanco, V.M. Martinez-Taboada, V. Rodriguez-Valverde, A. Sanchez-Andrade, M.A. Gonzalez-Gay.
Successful therapy with danazol in refractory autoimmune thrombocytopenia associated with rheumatic diseases.
Rheumatology., 36 (1997), pp. 1095-1099
[122]
A. Mak, C.C. Mok.
Mycophenolate mofetil for refractory haemolytic anemia in systemic lupus erythematosus.
Lupus., 14 (2005), pp. 856-858
[123]
K. Tselios, D.D. Gladman, J. Su, M.B. Urowitz.
Mycophenolate mofetil in nonrenal manifestations of systemic lupus erythematosus: an observational cohort study.
J Rheumatol., 43 (2016), pp. 552-558
[124]
J. Howard, A.V. Hoffbrand, H.G. Prentice, A. Mehta.
Mycophenolate mofetil for the treatment of refractory auto‐immune haemolytic anaemia and auto‐immune thrombocytopenia purpura.
Br J Haematol., 117 (2002), pp. 712-715
[125]
V.M. Moyo, D. Smith, I. Brodsky, P. Crilley, R.J. Jones, R.A. Brodsky.
High-dose cyclophosphamide for refractory autoimmune hemolytic anemia.
Blood., 100 (2002), pp. 704-706
[126]
M. Tokunaga, K. Fujii, K. Saito, et al.
Down-regulation of CD40 and CD80 on B cells in patients with life-threatening systemic lupus erythematosus after successful treatment with rituximab.
Rheumatology., 44 (2005), pp. 176-182
[127]
W. Barcellini, F. Zaja, A. Zaninoni, et al.
Low-dose rituximab in adult patients with idiopathic autoimmune hemolytic anemia: clinical efficacy and biologic studies.
Blood., 119 (2012), pp. 3691-3697
[128]
S. Perrotta, F. Locatelli, A. La Manna, L. Cennamo, P. De Stefano, B. Nobili.
Anti-CD20 monoclonal antibody (Rituximab) for life-threatening autoimmune haemolytic anaemia in a patient with systemic lupus erythematosus.
Br J Haematol, 116 (2002), pp. 465-467
[129]
D. Dierickx, A. Kentos, A. Delannoy.
The role of rituximab in adults with warm antibody autoimmune hemolytic anemia.
Blood., 125 (2015), pp. 3223-3229
[130]
R. Abdwani, R. Mani.
Anti-CD20 monoclonal antibody in acute life threatening haemolytic anaemia complicating childhood onset SLE.
Lupus., 18 (2009), pp. 460-464
[131]
O.W. Buetens, P.M. Ness.
Red blood cell transfusion in autoimmune hemolytic anemia.
Curr Opin Hematol., 10 (2003), pp. 429-433
[132]
A.B. Levine, D. Erkan.
Clinical assessment and management of cytopenias in lupus patients.
Curr Rheumatol Rep., 13 (2011), pp. 291-299
[133]
H.H. Euler, P. Harten, R.A. Zeuner, U.M. Schwab.
Recombinant human granulocyte colony stimulating factor in patients with systemic lupus erythematosus associated neutropenia and refractory infections.
J Rheumatol., 24 (1997), pp. 2153-2157
[134]
I.M. Vasiliu, M.A. Petri, A.N. Baer.
Therapy with granulocyte colony-stimulating factor in systemic lupus erythematosus may be associated with severe flares.
J Rheumatol., 33 (2006), pp. 1878-1880
[135]
H.H. Euler, U.M. Schwab, J.O. Schroeder.
Filgrastim for lupus neutropenia.
Lancet., 344 (1994), pp. 1513-1514
[136]
K.A. Newman, M. Akhtari.
Management of autoimmune neutropenia in Felty’s syndrome and systemic lupus erythematosus.
Autoimmun Rev., 10 (2011), pp. 432-437
[137]
M. Abu-Hishmeh, A. Sattar, F. Zarlasht, et al.
Systemic lupus erythematosus presenting as refractory thrombotic thrombocytopenic purpura: a diagnostic and management challenge. A case report and concise review of the literature.
Am J Case Rep., 17 (2016), pp. 782-787
[138]
W. Ma, W. Bai, X. Wu, J. Zhao, M. Li, X. Zeng.
Successful treatment of refractory thrombotic thrombocytopenic purpura associated with systemic lupus erythematosus with combination of plasma exchange and low-dose rituximab.
Lupus., 29 (2020), pp. 1961-1967
[139]
E.S. Kotzen, S. Roy, K. Jain.
Antiphospholipid syndrome nephropathy and other thrombotic microangiopathies among patients with systemic lupus erythematosus.
Adv Chronic Kidney Dis., 26 (2019), pp. 376-386
[140]
S.J. Liang, Q.Y. Zheng, M.S. Li, M.Y. Lv, W.T. Chen, Y. Yang.
Response to belimumab in thrombotic thrombocytopenic purpura associated with systemic lupus erythematosus: a case-based review.
Clin Rheumatol., 41 (2022), pp. 2561-2569
[141]
N. Kello, L.E. Khoury, G. Marder, R. Furie, E. Zapantis, D.L. Horowitz.
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., 49 (2019), pp. 74-83
[142]
P.E. Gavand, I. Serio, L. Arnaud, et al.
Clinical spectrum and therapeutic management of systemic lupus erythematosus-associated macrophage activation syndrome: a study of 103 episodes in 89 adult patients.
Autoimmun Rev., 16 (2017), pp. 743-749
[143]
A. Wafa, H. Hicham, R. Naoufal, et al.
Clinical spectrum and therapeutic management of systemic lupus erythematosus-associated macrophage activation syndrome: a study of 20 Moroccan adult patients.
Clin Rheumatol., 41 (2022), pp. 2021-2033
[144]
K. Shah, A. Porter, G. Takhar, V. Reddy.
Case of using mycophenolate in combination with steroids for concurrent macrophage activation syndrome and lupus flare.
[145]
Y. Tomofuji, Y. Ishikawa, K. Hattori, M. Fujiwara, Y. Kita.
Successful treatment of refractory acute lupus haemophagocytic syndrome using rituximab: a case report.
Mod Rheumatol Case Rep., 4 (2020), pp. 222-228
[146]
F. Posch, J. Gebhart, J.H. Rand, et al.
Cardiovascular risk factors are major determinants of thrombotic risk in patients with the lupus anticoagulant.
[147]
M.J. Cuadrado, M.L. Bertolaccini, P.T. Seed, et al.
Low-dose aspirin vs low-dose aspirin plus low-intensity warfarin in thromboprophylaxis: a prospective, multicentre, randomized, open, controlled trial in patients positive for antiphospholipid antibodies (ALIWAPAS).
Rheumatology (Oxford)., 53 (2014), pp. 275-284
[148]
M. Anderson, H.M. Belmont.
Severe thrombotic events associated with pre-procedural interruption of anticoagulation in systemic lupus erythematosus with secondary antiphospholipid syndrome: cases and literature review.
Lupus., 31 (2022), pp. 261-267
[149]
R. Cáliz Cáliz, P. Díaz Del Campo Fontecha, M. Galindo Izquierdo, et al.
Recommendations of the Spanish rheumatology society for primary antiphospholipid syndrome. Part I: diagnosis, evaluation and treatment.
Reumatol Clin., 16 (2020), pp. 71-86
[150]
H. Okuma, Y. Kitagawa, T. Yasuda, K. Tokuoka, S. Takagi.
Comparison between single antiplatelet therapy and combination of antiplatelet and anticoagulation therapy for secondary prevention in ischemic stroke patients with antiphospholipid syndrome.
Int J Med Sci., 7 (2009), pp. 15-18
[151]
J. Aibar, S. Schulman.
Arterial thrombosis in patients with antiphospholipid syndrome: a review and meta-analysis.
Semin Thromb Hemost., 47 (2021), pp. 709-723
[152]
T. Attachaipanich, A. Aungsusiripong, P. Piriyakhuntorn, et al.
Antithrombotic therapy in antiphospholipid syndrome with arterial thrombosis: a systematic review and network meta-analysis.
Front Med (Lausanne)., 10 (2023),
[153]
M.M. Bala, M. Celinska-Lowenhoff, W. Szot, et al.
Antiplatelet and anticoagulant agents for secondary prevention of stroke and other thromboembolic events in people with antiphospholipid syndrome.
Cochrane Database Syst Rev., 10 (2017),
[154]
O.C. Kwon, Y.B. Park, M.C. Park.
Effect of statins on the prevention of recurrent thrombosis in thrombotic antiphospholipid syndrome.
Rheumatology (Oxford)., 61 (2022), pp. 1548-1555
[155]
A. Broder, C. Putterman.
Hydroxychloroquine use is associated with lower odds of persistently positive antiphospholipid antibodies and/or lupus anticoagulant in systemic lupus erythematosus.
J Rheumatol., 40 (2013), pp. 30-33
[156]
S.C. Woller, S.M. Stevens, D. Kaplan, et al.
Apixaban compared with warfarin to prevent thrombosis in thrombotic antiphospholipid syndrome: a randomized trial.
Blood Adv., 6 (2022), pp. 1661-1670
[157]
K. Malec, T. Góralczyk, A. Undas.
The use of direct oral anticoagulants in 56 patients with antiphospholipid syndrome.
Thromb Res., 152 (2017), pp. 93-97
[158]
J. Ordi-Ros, L. Sáez-Comet, M. Pérez-Conesa, et al.
Rivaroxaban versus vitamin K antagonist in antiphospholipid syndrome: a randomized noninferiority trial.
Ann Intern Med., 171 (2019), pp. 685-694
[159]
V. Pengo, A. Banzato, E. Bison, G. Zoppellaro, S. Padayattil Jose, G. Denas.
Efficacy and safety of rivaroxaban vs warfarin in high-risk patients with antiphospholipid syndrome: Rationale and design of the Trial on Rivaroxaban in AntiPhospholipid Syndrome (TRAPS) trial.
Lupus., 25 (2016), pp. 301-306
[160]
Y.H. Lee, G.G. Song.
Direct oral anticoagulants versus warfarin in patients with antiphospholipid syndrome: a meta-analysis of randomized controlled trials.
Lupus., 31 (2022), pp. 1335-1343
[161]
P. Cerdà, C. Becattini, A. Iriarte, J.C. Hernández, X. Corbella, A. Riera-Mestre.
Direct oral anticoagulants versus vitamin K antagonists in antiphospholipid syndrome: a meta-analysis.
Eur J Intern Med., 79 (2020), pp. 43-50
[162]
X. Wu, S. Cao, B. Yu, T. He.
Comparing the efficacy and safety of direct oral anticoagulants versus Vitamin K antagonists in patients with antiphospholipid syndrome: a systematic review and meta-analysis.
Blood Coagul Fibrinolysis., 33 (2022), pp. 389-401
[163]
V. Dufrost, J. Risse, T. Reshetnyak, et al.
Increased risk of thrombosis in antiphospholipid syndrome patients treated with direct oral anticoagulants. Results from an international patient-level data meta-analysis.
Autoimmun Rev., 17 (2018), pp. 1011-1021
[164]
G.G.M. Balbi, S. Pacheco M de, O.A. Monticielo, et al.
Antiphospholipid Syndrome Committee of the Brazilian Society of Rheumatology position statement on the use of direct oral anticoagulants (DOACs) in antiphospholipid syndrome (APS).
Adv Rheumatol., 60 (2020), pp. 29
[165]
K. Al Sulaiman, A. Hafiz, H.A. Badreldin, et al.
Evaluation of apixaban in patients with antiphospholipid syndrome: a case series and review of literature.
J Investig Med High Impact Case Rep., 10 (2022),
[166]
D. Isenberg, G.B. Appel, G. Contreras, et al.
Influence of race/ethnicity on response to lupus nephritis treatment: the ALMS study.
Rheumatology., 49 (2010), pp. 128-140
[167]
F.G. Arévalo-Martínez, L. Andrade-Ortega, F. Irazoque-Palazuelos, J.J. Badía-Flores.
Presentación atípica y evolución clínina de la nefropatía lúpica mesangial. Estudio de 20 pacientes.
Reumatol Clin., 2 (2006), pp. 4-9
[168]
D.J. Tunnicliffe, S.C. Palmer, L. Henderson, et al.
Immunosuppressive treatment for proliferative lupus nephritis.
Cochrane Database Syst Rev., 2018 (2018),
[169]
B.H. Rovin, Y.K.O. Teng, E.M. Ginzler, et al.
Efficacy and safety of voclosporin versus placebo for lupus nephritis (AURORA 1): a double-blind, randomised, multicentre, placebo-controlled, phase 3 trial.
Lancet., 397 (2021), pp. 2070-2080
[170]
A. Saxena, E.M. Ginzler, K. Gibson, et al.
Safety and efficacy of long-term voclosporin treatment for lupus nephritis in the phase 3 AURORA 2 clinical trial.
Arthritis Rheumatol., 76 (2024), pp. 59-67
[171]
A.R. Parra Sánchez, A.E. Voskuyl, R.F. van Vollenhoven.
Treat-to-target in systemic lupus erythematosus: advancing towards its implementation.
Nat Rev Rheumatol., 18 (2022), pp. 146-157
[172]
R.A. Levy, T. Gonzalez-Rivera, M. Khamashta, et al.
10 years of belimumab experience: what have we learnt?.
Lupus., 30 (2021), pp. 1705-1721
[173]
H. Wang, T. Li, F. Sun, et al.
Safety and efficacy of the SGLT2 inhibitor dapagliflozin in patients with systemic lupus erythematosus: a phase I/II trial.
[174]
S. Sciascia, J. Yazdany, M. Dall’Era, et al.
Anticoagulation in patients with concomitant lupus nephritis and thrombotic microangiopathy: a multicentre cohort study.
Ann Rheum Dis., 78 (2019), pp. 1004-1006
[175]
G.B. Appel, G. Contreras, M.A. Dooley, et al.
Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis.
J Am Soc Nephrol., 20 (2009), pp. 1103-1112
[176]
M.V. Collado, E. Dorado, S. Rausch, et al.
Long-term outcome of lupus nephritis class II in Argentine patients.
J Clin Rheumatol., 22 (2016), pp. 299-306
[177]
A. Fanouriakis, M. Kostopoulou, K. Cheema, et al.
2019 Update of the Joint European League Against Rheumatism and European Renal Association–European Dialysis and Transplant Association (EULAR/ERA–EDTA) recommendations for the management of lupus nephritis.
Ann Rheum Dis., 79 (2020), pp. 713-723
[178]
B.H. Rovin, I.M. Ayoub, T.M. Chan, Z.H. Liu, J.M. Mejía-Vilet, J. Floege.
KDIGO 2024 Clinical Practice Guideline for the management of LUPUS NEPHRITIS.
Kidney Int., 105 (2024), pp. S1-S69
[179]
Kidney Disease: Improving Global Outcomes (KDIGO) Lupus Nephritis Work Group.
KDIGO 2024 Clinical Practice Guideline for the management of LUPUS NEPHRITIS.
Kidney Int., 105 (2024), pp. S1-S69
[180]
Z. Zheng, H. Zhang, X. Peng, et al.
Effect of tacrolimus vs intravenous cyclophosphamide on complete or partial response in patients with lupus nephritis: a randomized clinical trial.
[181]
G.M. Sahin, S. Sahin, S. Kiziltas, S. Masatlioglu, F. Oguz, H. Ergin.
Mycophenolate mofetil versus azathioprine in the maintenance therapy of lupus nephritis.
Ren Fail., 30 (2008), pp. 865-869
[182]
H. Ikeuchi, K. Hiromura, S. Takahashi, et al.
Efficacy and safety of multi-target therapy using a combination of tacrolimus, mycophenolate mofetil and a steroid in patients with active lupus nephritis.
Mod Rheumatol., 24 (2014), pp. 618-625
[183]
Z. Tang, Z. Wang, H.T. Zhang, et al.
Clinical features and renal outcome in lupus patients with diffuse crescentic glomerulonephritis.
Rheumatol Int., 30 (2009), pp. 45-49
[184]
S. Chen, H. Chen, Z. Liu, et al.
Pathological spectrums and renal prognosis of severe lupus patients with rapidly progressive glomerulonephritis.
Rheumatol Int., 35 (2015), pp. 709-717
[185]
I. Cancarevic, B.H. Malik.
Use of rituximab in management of rapidly progressive glomerulonephritis.
[186]
S.H. Kang, B.H. Chung, S.R. Choi, et al.
Comparison of clinical outcomes by different renal replacement therapy in patients with end-stage renal disease secondary to lupus nephritis.
Korean J Intern Med., 26 (2011), pp. 60
[187]
C.H. Weng, C.W. Hsu, C.C. Yu, T.H. Yen, C.W. Yang, C.C. Hung.
Peritoneal dialysis and hemodialysis in systemic lupus erythematosus patients: comparison of clinical outcomes.
Kidney Blood Press Res., 32 (2009), pp. 451-456
[188]
G.K. Bertsias, J.P.A. Ioannidis, M. Aringer, et al.
EULAR recommendations for the management of systemic lupus erythematosus with neuropsychiatric manifestations: report of a task force of the EULAR standing committee for clinical affairs.
Ann Rheum Dis., 69 (2010), pp. 2074-2082
[189]
L.A. Barile-Fabris, H. Fragoso-Loyo, D. Wojdyla, et al.
Factors associated with neuropsychiatric involvement in Latin American patients with systemic lupus erythematosus.
Lupus., 30 (2021), pp. 1481-1491
[190]
The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes.
[191]
E.Y. McLaurin, S.L. Holliday, P. Williams, R.L. Brey.
Predictors of cognitive dysfunction in patients with systemic lupus erythematosus.
[192]
L. Barile-Fabris.
Controlled clinical trial of IV cyclophosphamide versus IV methylprednisolone in severe neurological manifestations in systemic lupus erythematosus.
Ann Rheum Dis., 64 (2005), pp. 620-625
[193]
J. Saison, N. Costedoat-Chalumeau, D. Maucort-Boulch, et al.
Systemic lupus erythematosus-associated acute transverse myelitis: manifestations, treatments, outcomes, and prognostic factors in 20 patients.
[194]
G. Espinosa, A. Mendizábal, S. Mínguez, et al.
Transverse myelitis affecting more than 4 spinal segments associated with systemic lupus erythematosus: clinical, immunological, and radiological characteristics of 22 patients.
Semin Arthritis Rheum., 39 (2010), pp. 246-256
[195]
Y. Lin, A. Wang, M. Yen.
Systemic lupus erythematosus‐associated optic neuritis: clinical experience and literature review.
Acta Ophthalmol., 87 (2009), pp. 204-210
[196]
Y. Levy.
Intravenous immunoglobulins in peripheral neuropathy associated with vasculitis.
Ann Rheum Dis., 62 (2003), pp. 1221-1223
[197]
E.R. Vina, A.J. Fang, D.J. Wallace, M.H. Weisman.
Chronic inflammatory demyelinating polyneuropathy in patients with systemic lupus erythematosus: prognosis and outcome.
Semin Arthritis Rheum., 35 (2005), pp. 175-184
[198]
B.L. Man, C.C. Mok, Y.P. Fu.
Neuro‐ophthalmologic manifestations of systemic lupus erythematosus: a systematic review.
Int J Rheum Dis., 17 (2014), pp. 494-501
[199]
R.C. Dale, K. Yin, A. Ding, et al.
Antibody binding to neuronal surface in movement disorders associated with lupus and antiphospholipid antibodies.
Dev Med Child Neurol., 53 (2011), pp. 522-528
[200]
B.A. Binstadt, A.M.C. Caldas, S.E. Turvey, et al.
Rituximab therapy for multisystem autoimmune diseases in pediatric patients.
J Pediatr., 143 (2003), pp. 598-604
[201]
E. Menozzi, E. Mulroy, L. Akbarian-Tefaghi, K.P. Bhatia, B. Balint.
Movement disorders in systemic autoimmune diseases: clinical spectrum, ancillary investigations, pathophysiological considerations.
Parkinsonism Relat Disord., 88 (2021), pp. 116-128
[202]
C. Magro-Checa, E.J. Zirkzee, T.W. Huizinga, G.M. Steup-Beekman.
Management of neuropsychiatric systemic lupus erythematosus: current approaches and future perspectives.
Drugs., 76 (2016), pp. 459-483
[203]
S. Peluso, A. Antenora, A. De Rosa, et al.
Antiphospholipid-related chorea.
Front Neurol., (2012), pp. 3
[204]
C.C. Mok, C.S. Lau, R.W.S. Wong.
Treatment of lupus psychosis with oral cyclophosphamide followed by azathioprine maintenance: an open-label study.
Am J Med., 115 (2003), pp. 59-62
[205]
E. Abrol, E. Coutinho, M. Chou, et al.
Psychosis in systemic lupus erythematosus (SLE): 40-year experience of a specialist centre.
Rheumatology., 60 (2021), pp. 5620-5629
[206]
L.S.H. Lim, A. Lefebvre, S. Benseler, E.D. Silverman.
Longterm outcomes and damage accrual in patients with childhood systemic lupus erythematosus with psychosis and severe cognitive dysfunction.
J Rheumatol, 40 (2013), pp. 513-519
[207]
J.J. Miner, A.H.J. Kim.
Cardiac manifestations of systemic lupus erythematosus.
Rheum Dis Clin North Am., 40 (2014), pp. 51-60
[208]
K. Tselios, M.B. Urowitz.
Cardiovascular and pulmonary manifestations of systemic lupus erythematosus.
[209]
S. Gaine, K. Chin, G. Coghlan, et al.
Selexipag for the treatment of connective tissue disease-associated pulmonary arterial hypertension.
[210]
G. Bertsias, J.P.A. Ioannidis, J. Boletis, et al.
EULAR recommendations for the management of systemic lupus erythematosus. Report of a Task Force of the EULAR Standing Committee for International Clinical Studies Including Therapeutics.
Ann Rheum Dis., 67 (2008), pp. 195-205
[211]
M. Imazio.
Pericardial involvement in systemic inflammatory diseases.
Heart., 97 (2011), pp. 1882-1892
[212]
B.L. Man, C.C. Mok.
Serositis related to systemic lupus erythematosus: prevalence and outcome.
Lupus., 14 (2005), pp. 822-826
[213]
G. Artom, N. Koren-Morag, D.H. Spodick, et al.
Pretreatment with corticosteroids attenuates the efficacy of colchicine in preventing recurrent pericarditis: a multi-centre all-case analysis.
Eur Heart J., 26 (2005), pp. 723-727
[214]
P. Kruzliak, M. Novak, P. Piler, G. Kovacova.
Pericardial involvement in systemic lupus erythematosus: current diagnosis and therapy.
Acta Cardiol., 68 (2013), pp. 629-633
[215]
N. Morel, M. Bonjour, V. Le Guern, et al.
Colchicine: a simple and effective treatment for pericarditis in systemic lupus erythematosus? A report of 10 cases.
Lupus., 24 (2015), pp. 1479-1485
[216]
M. Imazio, R. Belli, A. Brucato, et al.
Efficacy and safety of colchicine for treatment of multiple recurrences of pericarditis (CORP-2): a multicentre, double-blind, placebo-controlled, randomised trial.
Lancet., 383 (2014), pp. 2232-2237
[217]
G. Thomas, F. Cohen Aubart, L. Chiche, et al.
Lupus myocarditis: initial presentation and longterm outcomes in a multicentric series of 29 patients.
J Rheumatol., 44 (2017), pp. 24-32
[218]
M.A. Garcia, G.S. Alarcon, G. Boggio, et al.
Primary cardiac disease in systemic lupus erythematosus patients: protective and risk factors--data from a multi-ethnic Latin American cohort.
Rheumatology., 53 (2014), pp. 1431-1438
[219]
L. Gonzalez-Lopez, E.G. Cardona-Muñoz, A. Celis, et al.
Therapy with intermittent pulse cyclophosphamide for pulmonary hypertension associated with systemic lupus erythematosus.
Lupus., 13 (2004), pp. 105-112
[220]
S. Kommireddy, S. Bhyravavajhala, K. Kurimeti, et al.
Pulmonary arterial hypertension in systemic lupus erythematosus may benefit by addition of immunosuppression to vasodilator therapy: an observational study.
Rheumatology., 54 (2015), pp. 1673-1679
[221]
E. Tanaka, M. Harigai, M. Tanaka, Y. Kawaguchi, M. Hara, N. Kamatani.
Pulmonary hypertension in systemic lupus erythematosus: evaluation of clinical characteristics and response to immunosuppressive treatment.
J Rheumatol., 29 (2002), pp. 282-287
[222]
C. Muangchan, R.F. van Vollenhoven, S.R. Bernatsky, et al.
Treatment algorithms in systemic lupus erythematosus.
Arthritis Care Res (Hoboken)., 67 (2015), pp. 1237-1245
[223]
S. Miyamichi-Yamamoto, Y. Fukumoto, K. Sugimura, et al.
Intensive immunosuppressive therapy improves pulmonary hemodynamics and long-term prognosis in patients with pulmonary arterial hypertension associated with connective tissue disease.
Circulation J., 75 (2011), pp. 2668-2674
[224]
N. Galiè, B.H. Brundage, H.A. Ghofrani, et al.
Tadalafil therapy for pulmonary arterial hypertension.
Circulation., 119 (2009), pp. 2894-2903
[225]
N. Galiè, M. Humbert, J.L. Vachiery, et al.
2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension.
Eur Heart J., 37 (2016), pp. 67-119
[226]
M.E. Beltrán-Gámez, J. Sandoval-Zárate, T. Pulido.
Inhibidores de fosfodiesterasa-5 para el tratamiento de la hipertensión arterial pulmonar.
Arch Cardiol Mex., 85 (2015), pp. 215-224
[227]
D.B. Badesch, N.S. Hill, G. Burgess, et al.
Sildenafil for pulmonary arterial hypertension associated with connective tissue disease.
J Rheumatol., 34 (2007), pp. 2417-2422
[228]
L.J. Rubin, D.B. Badesch, R.J. Barst, et al.
Bosentan therapy for pulmonary arterial hypertension.
N Engl J Med., 346 (2002), pp. 896-903
[229]
M.P. Keane.
Rare diseases bullet 7: pleuropulmonary manifestations of systemic lupus erythematosus.
Thorax, 55 (2000), pp. 159-166
[230]
J.M. Pego-Reigosa, D.A. Medeiros, D.A. Isenberg.
Respiratory manifestations of systemic lupus erythematosus: old and new concepts.
Best Pract Res Clin Rheumatol., 23 (2009), pp. 469-480
[231]
R.A. Matthay, M.I. Schwarz, T.L. Petty, et al.
Pulmonary manifestations of systemic lupus erythematosus: review of twelve cases of acute lupus pneumonitis.
[232]
D. Allen, A. Fischer, Z. Bshouty, et al.
Evaluating systemic lupus erythematosus patients for lung involvement.
Lupus., 21 (2012), pp. 1316-1325
[233]
L. Weinrib, O.P. Sharma, F.P. Quismorio.
A long-term study of interstitial lung disease in systemic lupus erythematosus.
Semin Arthritis Rheum., 20 (1990), pp. 48-56
[234]
H. Badsha, C.L. Teh, K.O. Kong, T.Y. Lian, H.H. Chng.
Pulmonary hemorrhage in systemic lupus erythematosus.
Semin Arthritis Rheum., 33 (2004), pp. 414-421
[235]
C. Andrade, T. Mendonça, F. Farinha, et al.
Alveolar hemorrhage in systemic lupus erythematosus: a cohort review.
[236]
E.P. Schwab, H.R. Schumacher, B. Freundlich, P.E. Callegari.
Pulmonary alveolar hemorrhage in systemic lupus erythematosus.
Semin Arthritis Rheum., 23 (1993), pp. 8-15
[237]
J.R. Tse, K.E. Schwab, M. McMahon, W. Simon.
Rituximab: an emerging treatment for recurrent diffuse alveolar hemorrhage in systemic lupus erythematosus.
Lupus., 24 (2015), pp. 756-759
[238]
A.H. Law, T.Y. Chuah, W. Lee, et al.
Diffuse alveolar haemorrhage in systemic lupus erythematosus: a multicentre retrospective study in Singapore.
Lupus., 32 (2023), pp. 952-963
[239]
R.B. Frittoli, J.F. Vivaldo, L.T.L. Costallat, S. Appenzeller.
Gastrointestinal involvement in systemic lupus erythematosus: a systematic review.
J Transl Autoimmun., 4 (2021),
[240]
Z. Li, D. Xu, Z. Wang, et al.
Gastrointestinal system involvement in systemic lupus erythematosus.
Lupus., 26 (2017), pp. 1127-1138
[241]
C.R. Wang, H.W. Tsai.
Autoimmune liver diseases in systemic rheumatic diseases.
World J Gastroenterol., 28 (2022), pp. 2527-2545
[242]
J.A. Ronen, A. Mekala, C. Wiechmann, S. Mungara.
A flare-up of systemic lupus erythematosus with unusual enteric predominance.
[243]
C. Quintana Lazopulos, M. Orozco, D. Balderramo.
Small-vessel intestinal vasculitis as an initial manifestation of systemic lupus erythematosus.
Dig Liver Dis., 50 (2018), pp. 1096
[244]
B.N. Brewer, D.L. Kamen.
Gastrointestinal and hepatic disease in systemic lupus erythematosus.
Rheum Dis Clin North Am., 44 (2018), pp. 165-175
[245]
B.L. Adler, H. Timlin, J. Birnbaum.
Lupus intestinal pseudo-obstruction and hydronephrosis.
[246]
M. Ayari, A. Nakhli, Z. Teyeb, I. Abdelaali, S. Bellakhal, T. Jomni.
Intestinal pseudo‐obstruction: unusual presentation of systemic lupus erythematous.
Clin Case Rep., 9 (2021), pp. 1759-1762
[247]
F.J. Zhang, J. Zhang, L.P. Zhou, A.M. Wang, X.M. Li.
Intestinal pseudo-obstruction as the initial manifestation of systemic lupus erythematosus.
Am J Emerg Med., 37 (2019), pp. 176.e1-176.e2
[248]
R. Wang, B. Zheng, B. Wang, P. Ma, F. Chen, L. Tang.
A report of chronic intestinal pseudo-obstruction related to systemic lupus erythematosus.
Open Med., 13 (2018), pp. 562-564
[249]
S. Alharbi.
Gastrointestinal manifestations in patients with systemic lupus erythematosus.
Open Access Rheumatol., 14 (2022), pp. 243-253
[250]
M. Mustafa, Y.M. Bawazir.
Acute liver failure as the first feature of systemic lupus erythematosus.
Rheumatol Int., 41 (2021), pp. 469-474
[251]
L. Zhang, L. Yin, W. Lv, et al.
Clinical analysis of patients with systemic lupus erythematosus complicated with liver failure.
Clin Rheumatol., 42 (2023), pp. 1545-1553
[252]
F.S. Chaudhary, A. Sureen, M.N. Yousaf, D.S. Rao, C. Siva.
Co-presentation of lupus nephritis with autoimmune hepatitis.
Am J Case Reports., (2023), pp. 24
[253]
P. Arvaniti, G. Giannoulis, S. Gabeta, K. Zachou, G.K. Koukoulis, G.N. Dalekos.
Belimumab is a promising third-line treatment option for refractory autoimmune hepatitis.
[254]
M. Kolev, A.C. Sarbu, B. Möller, B. Maurer, F. Kollert, N. Semmo.
Belimumab treatment in autoimmune hepatitis and primary biliary cholangitis – a case series.
J Transl Autoimmun., 6 (2023),
[255]
A. Gebreselassie, F. Aduli, C.D. Howell.
Rheumatologic diseases and the liver.
Clin Liver Dis., 23 (2019), pp. 247-261
[256]
J.A. González-Regueiro, M. Cruz-Contreras, J. Merayo-Chalico, et al.
Hepatic manifestations in systemic lupus erythematosus.
Lupus., 29 (2020), pp. 813-824
[257]
A. Dima, D.V. Balaban, C. Jurcut, M. Jinga.
Systemic lupus erythematosus-related acute pancreatitis.
[258]
S. Grabar, M. Groh, M. Bahuaud, et al.
Pneumococcal vaccination in patients with systemic lupus erythematosus: a multicenter placebo-controlled randomized double-blind study.
Vaccine., 35 (2017), pp. 4877-4885
[259]
Y. Huang, H. Wang, L. Wan, X. Lu, W.W.S. Tam.
Is systemic lupus erythematosus associated with a declined immunogenicity and poor safety of influenza vaccination?.
[260]
K.A.M. Kuruma, E.F. Borba, M.H. Lopes, J.F. de Carvalho, E. Bonfá.
Safety and efficacy of hepatitis B vaccine in systemic lupus erythematosus.
Lupus., 16 (2007), pp. 350-354
[261]
M. Pugès, P. Biscay, T. Barnetche, et al.
Immunogenicity and impact on disease activity of influenza and pneumococcal vaccines in systemic lupus erythematosus: a systematic literature review and meta-analysis.
Rheumatology., 55 (2016), pp. 1664-1672
[262]
M. Milanovic, L. Stojanovich, A. Djokovic, M. Kontic, E. Gvozdenovic.
Influenza vaccination in autoimmune rheumatic disease patients.
Tohoku J Exp Med., 229 (2013), pp. 29-34
[263]
O. Elkayam, S. Amir, E. Mendelson, et al.
Efficacy and safety of vaccination against pandemic 2009 influenza A (H1N1) virus among patients with rheumatic diseases.
Arthritis Care Res (Hoboken)., 63 (2011), pp. 1062-1067
[264]
Z. Liao, H. Tang, X. Xu, Y. Liang, Y. Xiong, J. Ni.
Immunogenicity and safety of influenza vaccination in systemic lupus erythematosus patients compared with healthy controls: a meta-analysis.
[265]
M. Adawi, N.L. Bragazzi, D. McGonagle, et al.
Immunogenicity, safety and tolerability of anti-pneumococcal vaccination in systemic lupus erythematosus patients: An evidence-informed and PRISMA compliant systematic review and meta-analysis.
Autoimmun Rev., 18 (2019), pp. 73-92
[266]
R.P.V. Rezende, M. Oliveira-Santos, L.E.C. Andrade, E.M. Klumb.
Combined 13-valent pneumococcal conjugate and 23-valent pneumococcal polysaccharide vaccine regimens for adults with systemic lupus erythematosus: does the sequence of pneumococcal vaccination affect immunogenicity responses? A single-center cohort study in Brazil.
Lupus., 32 (2023), pp. 694-703
[267]
A. Jena, S. Mishra, P. Deepak, et al.
Response to SARS-CoV-2 vaccination in immune mediated inflammatory diseases: Systematic review and meta-analysis.
[268]
C. Ferri, L. Gragnani, V. Raimondo, et al.
Absent or suboptimal response to booster dose of COVID-19 vaccine in patients with autoimmune systemic diseases.
[269]
J. Law, C. Sorrento, A. Saxena.
Vaccination updates and special considerations for systemic lupus erythematosus patients.
Curr Opin Rheumatol., 36 (2024), pp. 148-153
[270]
M. Krasselt, C. Baerwald, U.G. Liebert, O. Seifert.
Humoral immunity to varicella zoster virus in patients with systemic lupus erythematosus and rheumatoid arthritis compared to healthy controls.
Vaccines (Basel)., 9 (2021), pp. 325
[271]
C.C. Mok, K.H. Chan, L.Y. Ho, Y.F. Fung, W.F. Fung, P.C.Y. Woo.
Safety and immune response of a live-attenuated herpes zoster vaccine in patients with systemic lupus erythematosus: a randomised placebo-controlled trial.
Ann Rheum Dis., 78 (2019), pp. 1663-1668
[272]
V. Infante, K.T. Miyaji, P.C. Soarez, A.M.C. Sartori.
Systematic review and meta-analysis of HPV vaccination in women with systemic lupus erythematosus (SLE).
Expert Rev Vaccines., 20 (2021), pp. 309-318
[273]
C.C. Mok, L.Y. Ho, L.S. Fong, C.H. To.
Immunogenicity and safety of a quadrivalent human papillomavirus vaccine in patients with systemic lupus erythematosus: a case–control study.
Ann Rheum Dis., 72 (2013), pp. 659-664
[274]
K. Albrecht, D. Poddubnyy, J. Leipe, et al.
Perioperative management of patients with inflammatory rheumatic diseases.
Z Rheumatol., 82 (2023), pp. 1-11
[275]
D. Zisa, S.M. Goodman.
Perioperative management of rheumatic disease and therapies.
Med Clin North Am., 105 (2021), pp. 273-284
[276]
D.M. Grennan.
Methotrexate and early postoperative complications in patients with rheumatoid arthritis undergoing elective orthopaedic surgery.
Ann Rheum Dis., 60 (2001), pp. 214-217
[277]
S. Goodman, R. Friedlander, C. Figgie, et al.
Flares occur frequently in RA patients undergoing arthroplasty [abstract].
Arthritis Rheumatol., 67 (2015),
[278]
S.M. Goodman, B.D. Springer, A.F. Chen, et al.
2022 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the Perioperative Management of Antirheumatic Medication in Patients With Rheumatic Diseases Undergoing Elective Total Hip or Total Knee Arthroplasty.
Arthritis Care Res (Hoboken)., 74 (2022), pp. 1399-1408
[279]
M.R. Klement, C.T. Penrose, A. Bala, S.S. Wellman, M.P. Bolognesi, T.M. Seyler.
How do previous solid organ transplant recipients fare after primary total knee arthroplasty?.
J Arthroplasty, 31 (2016), pp. 609-615.e1
[280]
P.E. Marik.
Requirement of perioperative stress doses of corticosteroids.
Arch Surg., 143 (2008), pp. 1222
[281]
S. Tarp, D. Eric Furst, M. Boers, et al.
Risk of serious adverse effects of biological and targeted drugs in patients with rheumatoid arthritis: a systematic review meta-analysis.
[282]
M. Ramos-Casals, M. Soto, M. Cuadrado, M. Khamashta.
Rituximab in systemic lupus erythematosus: a systematic review of off-label use in 188 cases.
Lupus., 18 (2009), pp. 767-776
[283]
E. Murray, M. Perry.
Off-label use of rituximab in systemic lupus erythematosus: a systematic review.
Clin Rheumatol., 29 (2010), pp. 707-716
[284]
R. Furie, M. Petri, O. Zamani, 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., 63 (2011), pp. 3918-3930
[285]
P.K. Bundhun, M.Z.S. Soogund, F. Huang.
Impact of systemic lupus erythematosus on maternal and fetal outcomes following pregnancy: a meta-analysis of studies published between years 2001–2016.
J Autoimmun., 79 (2017), pp. 17-27
[286]
S. Zhang, X. Han, W. Liu, Q. Wen, J. Wang.
Pregnancy in patients with systemic lupus erythematosus: a systematic review.
Arch Gynecol Obstet., 308 (2023), pp. 63-71
[287]
M.Á Saavedra Salinas, A. Barrera Cruz, A.R. Cabral Castañeda, et al.
Guías de práctica clínica para la atención del embarazo en mujeres con enfermedades reumáticas autoinmunes del Colegio Mexicano de Reumatología. Parte II.
Reumatol Clin., 11 (2015), pp. 305-315
[288]
M. Petri, M.Y. Kim, K.C. Kalunian, et al.
Combined oral contraceptives in women with systemic lupus erythematosus.
N Engl J Med., 353 (2005), pp. 2550-2558
[289]
J. Sánchez-Guerrero, A.G. Uribe, L. Jiménez-Santana, et al.
A trial of contraceptive methods in women with systemic lupus erythematosus.
N Engl J Med., 353 (2005), pp. 2539-2549
[290]
A. Rojas-Villarraga, J.V. Torres-Gonzalez, ÁM. Ruiz-Sternberg.
Safety of hormonal replacement therapy and oral contraceptives in systemic lupus erythematosus: a systematic review and meta-analysis.
[291]
N. Chabbert-Buffet, Z. Amoura, P.Y. Scarabin, et al.
Pregnane progestin contraception in systemic lupus erythematosus: a longitudinal study of 187 patients.
Contraception., 83 (2011), pp. 229-237
[292]
R.C. Rebelo, E. Pignaton, M. Valeria Bahamondes, et al.
Disease activity and thromboembolic events in women with systemic lupus erythematosus with and without anti-phospholipid syndrome: users of the 52-mg levonorgestrel-releasing intrauterine system.
Arch Gynecol Obstet., 299 (2019), pp. 1597-1605
[293]
R.G. Silverstein, V. Fitz, M. Thornton, A. Carter, J.E. Morse.
Contraceptive use and counseling in patients with systemic lupus erythematosus.
Contraception., 105 (2022), pp. 46-50
[294]
S. Chandramouli, C. Alvarez, T.R. Englund, R.G. Silverstein, S.Z. Sheikh.
Teratogenic medication use associated with favourable odds of contraception counselling in a cohort of women with systemic lupus erythematosus at a large tertiary academic medical centre.
[295]
Q. Wei, Y. Ouyang, W. Zeng, L. Duan, J. Ge, H. Liao.
Pregnancy complicating systemic lupus erythematosus: a series of 86 cases.
Arch Gynecol Obstet., 284 (2011), pp. 1067-1071
[296]
A. Rajendran, A.M. Eudy, S.J. Balevic, M.E.B. Clowse.
The importance of pregnancy planning in lupus pregnancies.
Lupus., 30 (2021), pp. 741-751
[297]
L. Hamijoyo, J.W. Martha, S. Hidayat, et al.
Risk factors for poor pregnancy outcome in systemic lupus erythematosus patients.
Acta Med Indones., 51 (2019), pp. 102-109
[298]
J. Wu, W.H. Zhang, J. Ma, C. Bao, J. Liu, W. Di.
Prediction of fetal loss in Chinese pregnant patients with systemic lupus erythematosus: a retrospective cohort study.
[299]
A. Mokbel, D.H. Attia, H.S. Zayed, et al.
Pregnancy outcomes among Egyptian women with systemic lupus erythematosus: a prospective cohort study.
Lupus., 32 (2023), pp. 521-530
[300]
R. Ma, H. Gao, J. Cui, et al.
Pregnancy feasibility in women with mild pulmonary arterial hypertension: a systematic review and meta-analysis.
BMC Pregnancy Childbirth., 23 (2023), pp. 427
[301]
C.G. Skorpen, S. Lydersen, I.M. Gilboe, et al.
Influence of disease activity and medications on offspring birth weight, pre-eclampsia and preterm birth in systemic lupus erythematosus: a population-based study.
Ann Rheum Dis., 77 (2018), pp. 264-269
[302]
C. Tani, D. Zucchi, I. Haase, et al.
Are remission and low disease activity state ideal targets for pregnancy planning in systemic lupus erythematosus? A multicentre study.
Rheumatology., 60 (2021), pp. 5610-5619
[303]
J.W. Kim, J.Y. Jung, H.A. Kim, J.I. Yang, D.W. Kwak, C.H. Suh.
Lupus low disease activity state achievement is important for reducing adverse outcomes in pregnant patients with systemic lupus erythematosus.
J Rheumatol., 48 (2021), pp. 707-716
[304]
S. Ntali, D. Nikolopoulos, L. Pantazi, et al.
Remission or low disease activity at pregnancy onset are linked to improved foetal outcomes in women with systemic lupus erythematosus: results from a prospective observational study.
Clin Exp Rheumatol., 40 (2022), pp. 1769-1778
[305]
X. Sheng, X. Song, Y. Xiong, et al.
Maternal and infant outcomes of pregnancy associated with anti-SSA/RO antibodies: a systematic review and meta-analysis.
Pediatr Rheumatol., 21 (2023), pp. 22
[306]
E. Borella, A. Lojacono, M. Gatto, et al.
Predictors of maternal and fetal complications in SLE patients: a prospective study.
Immunol Res., 60 (2014), pp. 170-176
[307]
I. Jakobsen, R. Helmig, K. Stengaard-Pedersen.
Maternal and foetal outcomes in pregnant systemic lupus erythematosus patients: an incident cohort from a stable referral population followed during 1990–2010.
Scand J Rheumatol., 44 (2015), pp. 377-384
[308]
J.P. Buyon, M.Y. Kim, M.M. Guerra, et al.
Predictors of pregnancy outcomes in patients with lupus.
Ann Intern Med., 163 (2015), pp. 153-163
[309]
C.M. Yelnik, T.F. Porter, D.W. Branch, et al.
Brief report: changes in antiphospholipid antibody titers during pregnancy: effects on pregnancy outcomes.
Arthritis Rheumatol., 68 (2016), pp. 1964-1969
[310]
M. Deguchi, Y. Maesawa, S. Kubota, et al.
Factors associated with adverse pregnancy outcomes in women with systematic lupus erythematosus.
J Reprod Immunol., 125 (2018), pp. 39-44
[311]
C.R. Palma dos Reis, G. Cardoso, C. Carvalho, I. Nogueira, A. Borges, F. Serrano.
Prediction of adverse pregnancy outcomes in women with systemic lupus erythematosus.
Clin Rev Allergy Immunol., 59 (2020), pp. 287-294
[312]
R. Fischer-Betz, C. Specker, R. Brinks, M. Aringer, M. Schneider.
Low risk of renal flares and negative outcomes in women with lupus nephritis conceiving after switching from mycophenolate mofetil to azathioprine.
Rheumatology., 52 (2013), pp. 1070-1076
[313]
M.E.B. Clowse, K.G. Saag.
Unintended consequences of SCOTUS abortion decision for patients with rheumatic diseases.
Ann Intern Med., 175 (2022), pp. 1328-1329
[314]
Z. Hu, R. Gao, W. Huang, H. Wang, L. Qin.
Effect of hydroxychloroquine on lupus activity, preeclampsia and intrauterine growth restriction in pregnant women with systemic lupus erythematosus and/or antiphospholipid syndrome: a systematic review and meta-analysis.
J Clin Med., 12 (2023), pp. 485
[315]
E.G. McDonald, L. Bissonette, S. Ensworth, et al.
Monitoring of systemic lupus erythematosus pregnancies: a systematic literature review.
J Rheumatol., 45 (2018), pp. 1477-1490
[316]
J.A. Martínez López, M.L. García Vivar, R. Cáliz, et al.
Recomendaciones sobre actuaciones a seguir durante la edad fértil, el embarazo, posparto y lactancia en pacientes con enfermedades reumáticas inflamatorias y autoinmunes.
Reumatol Clin., 13 (2017), pp. 264-281
[317]
L. Andreoli, G.K. Bertsias, N. Agmon-Levin, et al.
EULAR recommendations for women’s health and the management of family planning, assisted reproduction, pregnancy and menopause in patients with systemic lupus erythematosus and/or antiphospholipid syndrome.
Ann Rheum Dis., 76 (2017), pp. 476-485
[318]
S.O. Keeling, Z. Alabdurubalnabi, A. Avina-Zubieta, et al.
Canadian rheumatology association recommendations for the assessment and monitoring of systemic lupus erythematosus.
J Rheumatol., 45 (2018), pp. 1426-1439
[319]
L.R. Sammaritano, B.L. Bermas, E.E. Chakravarty, et al.
2020 American College of Rheumatology Guideline for the Management of Reproductive Health in Rheumatic and Musculoskeletal Diseases.
Arthritis Rheumatol., 72 (2020), pp. 529-556
[320]
K. Schreiber, M. Frishman, M.D. Russell, et al.
British Society for Rheumatology guideline on prescribing drugs in pregnancy and breastfeeding: comorbidity medications used in rheumatology practice.
Rheumatology., 62 (2023), pp. e89-e104
[321]
M.D. Russell, M. Dey, J. Flint, et al.
British Society for Rheumatology guideline on prescribing drugs in pregnancy and breastfeeding: immunomodulatory anti-rheumatic drugs and corticosteroids.
Rheumatology., 62 (2023), pp. e48-e88
[322]
Y. Zhang, D. Li, H. Guo, W. Wang, X. Li, S. Shen.
Association between thiopurines use and pregnancy outcomes in female patients with inflammatory bowel disease: a meta-analysis.
Curr Pharm Des., 27 (2021), pp. 2317-2324
[323]
H.L. Le, M.I. Francke, L.M. Andrews, B.C.M. de Winter, T. van Gelder, D.A. Hesselink.
Usage of tacrolimus and mycophenolic acid during conception, pregnancy, and lactation, and its implications for therapeutic drug monitoring: a systematic critical review.
Ther Drug Monit., 42 (2020), pp. 518-531
[324]
M. Petri, H. Landy, M.E.B. Clowse, et al.
Belimumab use during pregnancy: a summary of birth defects and pregnancy loss from belimumab clinical trials, a pregnancy registry and postmarketing reports.
Ann Rheum Dis., 82 (2023), pp. 217-225
[325]
L.S. Magder, M. Petri.
Incidence of and risk factors for adverse cardiovascular events among patients with systemic lupus erythematosus.
Am J Epidemiol., 176 (2012), pp. 708-719
[326]
M.H. Chuang, T.L. Chuang, K.Y. Huang, Y.F. Wang.
Age-adjusted Charlson Comorbidity Index scores predict major adverse cardiovascular events and all-cause mortality among systemic lupus erythematosus patients.
Tzu Chi Med J., 29 (2017), pp. 154
[327]
M. Fernández-Garcés, G. Haro, M.L. Micó.
Predisposing factors to nonfatal cardiovascular events in women with systemic lupus erythematosus. An observational, cross-sectional, multicenter study in Spain from the risk/systemic lupus erythematosus thematic network.
[328]
J.H. Haugaard, L. Dreyer, M.B. Ottosen, G. Gislason, K. Kofoed, A. Egeberg.
Use of hydroxychloroquine and risk of major adverse cardiovascular events in patients with lupus erythematosus: a Danish nationwide cohort study.
J Am Acad Dermatol., 84 (2021), pp. 930-937
[329]
D.H. Yang, P.Y. Leong, S.K. Sia, Y.H. Wang, J.C.C. Wei.
Long-term hydroxychloroquine therapy and risk of coronary artery disease in patients with systemic lupus erythematosus.
J Clin Med., 8 (2019), pp. 796
[330]
S. Demir, B. Artim-Esen, Y. Şahinkaya, et al.
Metabolic syndrome is not only a risk factor for cardiovascular diseases in systemic lupus erythematosus but is also associated with cumulative organ damage: a cross-sectional analysis of 311 patients.
Lupus., 25 (2016), pp. 177-184
[331]
M.A. Petri, A.N. Kiani, W. Post, L. Christopher-Stine, L.S. Magder.
Lupus Atherosclerosis Prevention Study (LAPS).
Ann Rheum Dis., 70 (2011), pp. 760-765
[332]
W. Plazak, K. Gryga, H. Dziedzic, et al.
Influence of atorvastatin on coronary calcifications and myocardial perfusion defects in systemic lupus erythematosus patients: a prospective, randomized, double-masked, placebo-controlled study.
Arthritis Res Ther., 13 (2011),
[333]
L. Arnaud, A. Mathian, H. Devilliers, et al.
Patient-level analysis of five international cohorts further confirms the efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies.
Autoimmun Rev., 14 (2015), pp. 192-200
[334]
L. Arnaud, A. Mathian, A. Ruffatti, et al.
Efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies: An international and collaborative meta-analysis.
Autoimmun Rev., 13 (2014), pp. 281-291
[335]
S. Fasano, L. Pierro, I. Pantano, M. Iudici, G. Valentini.
Longterm hydroxychloroquine therapy and low-dose aspirin may have an additive effectiveness in the primary prevention of cardiovascular events in patients with systemic lupus erythematosus.
J Rheumatol., 44 (2017), pp. 1032-1038
[336]
M. Iudici, S. Fasano, L. Gabriele Falcone, et al.
Low-dose aspirin as primary prophylaxis for cardiovascular events in systemic lupus erythematosus: a long-term retrospective cohort study.
Rheumatology., 55 (2016), pp. 1623-1630
[337]
S. Fasano, D.P. Margiotta, L. Navarini, et al.
Primary prevention of cardiovascular disease in patients with systemic lupus erythematosus: case series and literature review.
Lupus., 26 (2017), pp. 1463-1472
[338]
G.A. Ferreira, T.P. Navarro, R.W. Telles, L.E.C. Andrade, E.I. Sato.
Atorvastatin therapy improves endothelial-dependent vasodilation in patients with systemic lupus erythematosus: an 8 weeks controlled trial.
Rheumatology., 46 (2007), pp. 1560-1565
[339]
G.E. Norby, I. Holme, B. Fellström, et al.
Effect of fluvastatin on cardiac outcomes in kidney transplant patients with systemic lupus erythematosus: a randomized placebo‐controlled study.
Arthritis Rheum., 60 (2009), pp. 1060-1064
[340]
A. Sahebkar, J. Rathouska, G. Derosa, P. Maffioli, P. Nachtigal.
Statin impact on disease activity and C-reactive protein concentrations in systemic lupus erythematosus patients: a systematic review and meta-analysis of controlled trials.
Autoimmun Rev., 15 (2016), pp. 344-353
Copyright © 2024. Elsevier España, S.L.U. and Sociedad Española de Reumatología y Colegio Mexicano de Reumatología
Download PDF
Idiomas
Reumatología Clínica (English Edition)
Article options
Tools
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?