Abstract
Patients with systemic lupus erythematosus experience high rates of infections. The use of immunosuppressive drugs to treat the disease, along with the fact that both the innate and adaptive branches of the immune system are compromised, account for the development of infections. In this communication, we briefly discuss the aberrant function of the immune system in patients with systemic lupus erythematosus and review the occurrence of infections that have been reported in clinical trials conducted to develop new therapeutics. Understanding the immune dysfunction in patients with systemic lupus erythematosus and the appearance of infections while trying to control the disease using immunosuppressive or immunomodulatory drugs should help limit infections and mitigate the associated morbidity and mortality.
Keywords: Systemic lupus erythematosus, infections, immunosuppression
Introduction
Patients with systemic lupus erythematosus (SLE) display a compromised immune system due to abnormalities affecting both the innate and adaptive components of the immune response.1 In addition, treatment invariantly involves using immunosuppressive drugs. Both the immunocompromised status and drugs used to treat patients with SLE contribute to increased susceptibility to acute and chronic infections, leading to increased morbidity and mortality.2 While new therapeutic approaches have been introduced or are under investigation, it should be noted that they also possess immunosuppressive properties and thereby increase the vulnerability of patients to infections.3
Apart from the heightened susceptibility to infections in individuals with SLE, it is important to keep in mind that infections can trigger disease flares by stimulating the innate immune system or by cross-reacting with receptors of the adaptive immune cells. This can complicate the diagnostic process and the selection of appropriate treatment. Diagnosis may present challenges because symptoms, such as fever, lymphadenopathy, pulmonary infiltrates, skin and mucosal rashes, and coagulopathy, may be shared by a disease flare and an ongoing infection.2 Accordingly, it is important to maintain a high level of clinical vigilance when it comes to diagnosing and treating infections in individuals with SLE. The enhanced recognition and treatment of infections in individuals with SLE during the past 4 decades have significantly improved the survival rates.4 This review will present a discussion of the increased susceptibility of patients with SLE to infections as a result of their immunocompromised status as well as the infections that result from the use of immunosuppressive drugs.
Immune Defects Leading to Increased Risk to Infections
Patients with SLE are vulnerable to infectious agents because of deficiencies in both the adaptive and innate immune systems.1 (Table 1)
Table 1.
Cells, Proteins, and Cytokines | Relation to Infections |
Monocytes/macrophages |
|
Neutrophils |
|
Lymphocytes |
|
NK cells |
|
Cytokine dysregulation |
|
Complement system |
|
CD, cluster of differentiation; CR1, complement receptor type 1; IFN-γ, interferon gamma; Ig, immunoglobulins; IL, interleukin; G-CSF, granulocyte colony-stimulating factor; NET, neutrophil extracellular traps; NK, natural killer; SLE, systemic lupus erythematosus; TNFα, tumor necrosis factor alpha.
Cells of the Innate Immune Response
Several abnormalities in neutrophils, macrophages, and monocytes have been reported in individuals with SLE. Monocytes exhibit impaired engulfment of apoptotic cells and reduced phagocytic activity.5 The antigen-presenting function of the macrophage/monocyte system is also defective.2 Production of superoxide following Fcγ receptor-mediated phagocytosis, which is important in the defense against infectious agents, is impaired.6 This can be further compromised by autoantibodies against the Fcγ receptors.7 The phagocytic function of macrophages and neutrophils may be influenced by autoantibodies targeting the 3 subclasses of Fcγ receptors (FcγRI, FcγRII, FcγRIII). These autoantibodies can further disrupt the immune system because Fcγ receptors are expressed on the surface membrane of B cells, natural killer cells, and specific γδT cells.8
Neutrophil counts are frequently decreased in patients with SLE, and their function can also be defective.1,9 For more than 50 years, there has been evidence suggesting impaired phagocytosis by polymorphonuclear leukocytes.10 Sera from patients with SLE and active disease suppress the opsonization of protein A-containing Staphylococcus aureus.11 Neutrophil numbers and function may be diminished by complement-activating anti-neutrophil antibodies and, in some cases, antibodies targeting myeloid precursors.12 Moreover, some SLE patients possess circulating autoantibodies targeting the neutrophil adhesion glycoproteins αMβ2, antibodies that may inhibit the receptor function of Mac-1 proteins.13
The presence of excessive amounts of immune complexes in the circulation is likely the primary cause of persistent neutrophil activation during active phases of the disease.14 As a consequence of prior neutrophil activation, patients with SLE might display impaired neutrophil function in response to infections. Increased levels of neutrophil apoptosis contribute to the probability of functional neutropenia and serve as an additional source of autoantigens. Abnormal levels of neutrophil extracellular traps (NETs), a host defense mechanism designed to trap pathogens, have been extensively documented in patients with SLE. Production of NET becomes even more pronounced during infections, resulting in an excessive release of autoantigens and cytokines, which contribute to collateral damage from vascular and endothelial injury.15 Furthermore, SLE neutrophils exhibit decreased responsiveness to IL-8, potentially leading to inadequate mobilization and a limited granulocyte response.16
The function and numbers of Natural Killer (NK)cells are notably diminished, especially during phases of disease exacerbation. Circulating NK antibodies can also induce cytotoxic effects, resulting in decreased NK numbers.17–22
In summary, cells of the innate immune system display compromised function through multiple mechanisms.
Lymphocytes
Lymphocytes in patients with SLE present a number of abnormalities.17,22–25 Lymphopenia, particularly cluster of differentiation (CD) 4 cytopenia, is frequent. There is a reduction in the production of key cytokines, such as interleukin (IL)-2 and interferon-γ, while the production of cytokines with proinflammatory properties, including IL-17, is increased.26
Despite the evident polyclonal B-cell activation and hyperglobulinemia in lupus patients, B cells seem to maintain proper functionality. An early study revealed that CD8 T cells from lupus patients were unable to control and eliminate autologous B cells infected with the Epstein–Barr virus (EBV). As a result, there is a continuous expansion of antibody and autoantibody production. The number of CD8 T cells able to bind EBV antigen is diminished in the circulating blood of lupus patients.27 The failure of CD8+ T cells to control other viruses can explain the increased susceptibility of SLE patients to viral infections.
To demonstrate that systemic autoimmunity accounts for an inherently immunocompromised status, Lieberman and Tsokos28 infected lupus-prone mice with the intracellular parasite T. gondii. Lupus-prone mice succumbed early because their T cells failed to produce interferon gamma (IFN-γ), a cytokine that is needed for the proper defense against T. gondii.
Among CD8+ T cells, a unique subset (CD8+CD38high T cells) is expanded in patients with SLE.29–31 CD38 is an ectoenzyme that degrades nicotinamide dinucleotide (NAD) and, through distinct molecular mechanisms, suppresses the expression of molecules that are responsible for the expression of cytotoxic activity, such as CD107, perforin, and granzymes.32–34 In a cross-sectional study, infections occurred at high rates almost exclusively with a CD8+CD38high T cell subset. Indeed, CD8+CD38high T cells display limited, if not absent, cytotoxic activity.35 Further, this CD8+CD38high T cell subset displayed limited mitomicrophagy and lysosomal activity. Of great translational value is the finding that CD8-targeted delivery of a CD38 small drug inhibitor suppressed hepatitis in lupus-prone mice infected with lymphocytic choriomengitis virus.36
In a prospective evaluation of 80 patients with SLE, we found that the patients with an expanded CD8+CD38high T cell subpopulation experienced more infectious events than the remaining patients (unpublished data).
In individuals with SLE, T-cell abnormalities encompass diminished cytotoxic capacity as assessed by allogeneic cell-mediated lympholysis, along with reduced NK cell activity. Early studies had shown that IL-2 can restore allogeneic and NK cell cytotoxicity.37 More recently, a study involving 665 lupus patients revealed that treatment with low-dose IL-2 therapy reduces by threefold the incidence of infections in SLE patients.38
Complement System
Genetic deficiency in various components of the complement pathways not only increases the risk of developing SLE but also predisposes individuals to infections.39 Immune complexes formed in patients with SLE can consume complement proteins, leading to a reduction in the available complement for formal defense mechanisms. Complement 1q is involved in the clearance of apoptotic material, and patients with C1q deficiency have heightened vulnerability to bacterial and viral infections.40,41 Also, certain mannan-binding lectin deficiencies have been linked to impaired opsonization. Increased consumption of complement components in SLE patients further impairs their ability to combat encapsulated microorganisms.42
A significant number of patients exhibit low levels of CR1, the complement receptor for C3b, on the plasma membrane of erythrocytes.40,43 It is possible that during disease flares, these levels are further decreased because they are occupied by immune complexes.44 Additionally, there is a decreased expression of CR1 on polymorphonuclear cells, and the presence of autoantibodies against CR1 has also been identified. Reduced expression of CR1 on these cells can lead to weakened phagocytosis.45 CR1 on erythrocytes transports immune complexes to the spleen, the main harbor of reticuloendothelial cells. Dysfunction of the spleen has been reported in patients with SLE. Up to 5% of SLE patients exhibit functional hyposplenism, which leads to an elevated risk of infections caused by encapsulated microorganisms and bacteria like Haemophilus sp., Salmonella, and Pneumococci.46-48
Compromised Anatomic Barriers to Infectious Agents
Anatomic lesions in patients with SLE caused by the disease itself present additional risk factors for infection. Skin lesions can compromise the integrity of the protective epidermal and other skin layers, increasing the vulnerability to secondary infections. The presence of skin lesions in SLE patients creates an entry point for pathogens, raising the risk of infection. Small vessel injury and glomerular scarring in the kidneys can lead to urinary tract infections (UTIs).2 End-stage renal disease and pulmonary fibrosis in patients with SLE can compromise the defensive capacity of the host against pathogens.1 In the gastrointestinal mucosa, capillary vasculitis may enable the leakage of pathogens into the circulation, thus increasing the risk of sepsis.49 These anatomic lesions in SLE patients significantly contribute to the heightened vulnerability to different types of infections. Managing these risk factors and promptly addressing any infections that arise are crucial in the comprehensive care of individuals with SLE.
Epidemiology and Types of Infections
Epidemiology
Infections play a substantial role in the morbidity and mortality of lupus patients. They are responsible for a substantial proportion of hospitalizations (13%-37%) and approximately one-third of overall deaths.50 (Table 2) Infection rates, types of infectious agents, and gravity of infections differ between developed and developing countries. Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, Escherichia coli, and Candida infections are more prevalent in developing nations. The occurrence of tuberculosis is significantly lower in developed countries. In general, infectious diseases are known to be more prevalent and pose twice the risk in developing countries.51 The majority of epidemiologic studies on the incidence and mortality of infections in patients with SLE have relied on inpatient data. In a study involving nearly 175 000 SLE-related hospitalizations in the USA in 2016, infections were the primary cause of in-hospital mortality (38.18%), with cardiac disease ranking second (12.04%).52
Table 2.
Lead Author (Reference) | Duration (Years) | Number of Patients | Number of Deaths | Death Caused by Infections (%) | Published Year |
---|---|---|---|---|---|
Wallace et al53 | 30 | 609 | 128 | 21 | 1981 |
Rosner et al54 | 13 | 1103 | 222 | 33 | 1982 |
Huicochea Grob et al55 | 23 | 65 | 14 | 29 | 1996 |
Kim et al56 | 4 | 544 | 43 | 33 | 1999 |
Jacobsen et al57 | 20 | 513 | 122 | 21 | 1999 |
Mok et al58 | 24 | 186 | 9 | 67 | 2000 |
Rodriquez et al59 | 34 | 662 | 161 | 27 | 2000 |
Bernatsky et al60 | 30 | 9547 | 1255 | 5 | 2006 |
Wadee et al61 | 15 | 226 | 55 | 44 | 2007 |
Nossent et al62 | 5 | 2500 | 91 | 57 | 2007 |
Al Arfaj et al63 | 30 | 624 | 25 | 48 | 2009 |
Goldblatt et al64 | 29 | 104 | 67 | 25 | 2009 |
Feng et al65 | 11 | 1956 | 116 | 25.9 | 2011 |
K McElhoneet al66 | 3 | 382 | 37 | 37.8 | 2014 |
Dubula et al67 | 6.5 | 167 | 24 | 62.5 | 2015 |
Tektonidou et al50 |
63 | 20 318 | 2179 | 33 | 2015 |
Chen et al68 | 10 | 3815 | 84 | 57 | 2016 |
Budhoo et al69 | 9.5 | 408 | 53 | 49.1 | 2016 |
Padjen et al70 | 10 | 967 | 90 | 33 | 2018 |
Wu et al71 | 10 | 29 510 | 360 | 65.8 | 2019 |
Dhital et al52 | 1 | 174 105 | 3405 | 38.1 | 2019 |
Ingvarsson et al72 | 25 | 175 | 60 | 15 | 2019 |
Gonzalez Lucero et al73 | 8 | 353 | 32 | 43.17 | 2020 |
Bultink et al74 | 25 | 4356 | 442 | 32 | 2021 |
Moghaddam et al75 | 18 | 6092 | 451 | 25 | 2022 |
Bacterial Infections
Bacterial infections are prominent in SLE patients and account for two-thirds of infections. Most bacterial infections are caused by opportunistic bacteria. Retrospective studies worldwide (North America, India, Africa, Europe, and Asia) have reported pathogens such as Escherichia coli, Streptococcus pneumoniae, Staphylococcus aureus, Salmonella enterica, and mycobacteria species. Although these bacteria can be found in the general population, they tend to induce more severe infections in patients with SLE. Colonization by these bacteria has the potential to exacerbate the ongoing autoimmune response and/or trigger new disease flares. In SLE, the respiratory and urinary tracts, along with the skin, are the common infection sites, representing over 60% of reported cases.51,76–78
Viral Infections
SLE patients have an increased susceptibility for viral diseases. Viruses like EBV, cytomegalovirus (CMV), and parvovirus B19 may trigger the development of SLE. Herpes zoster (HZ) is the most prevalent viral infection in SLE patients, with an incidence of 6-32/1000 person-years.51 HZ occurs when the varicella-zoster virus reactivates from dorsal root ganglia following primary infection. Cellular immune mechanisms primarily regulate this latent virus, and its reactivation can occur when CD8 cytotoxic cell function is compromised. SLE patients have a 2-3 times higher incidence of HZ compared to the general population.79
Fungal Infections
Invasive fungal infections are being increasingly reported in patients with SLE. In a study of 24 541 SLE patients, 445 fungal infections were recorded with 26.7% lethality. Among the fungi, Candida (52.8%), Cryptococcus (18.2%), and Aspergillus (18.2%) were the common pathogens.80
Coronavirus Disease 2019
Most SLE patients are not at increased risk of contracting coronavirus disease 2019 (COVID-19). Nevertheless, SLE patients may have taken more restrictive measures, including isolation, for fear of having a worse outcome if they contracted COVID-19.81 The COVID-19 Global Rheumatology Alliance (C-19-GRA) registry, consisting of over 20 000 patients with rheumatic diseases and COVID-19, provided data suggesting that SLE patients with moderate or high disease activity, as well as those taking specific medications (such as moderate or high doses of prednisone, rituximab, and immunosuppressive drugs like Azathioprine (AZA), mycophenolate mofetil (MMF)/mycophenolic acid (MPA),Cyclophosphamide (CYC), and tacrolimus (TAC)), experience more unfavorable outcomes compared to a reference group of individuals receiving methotrexate.82,83
A recent extensive analysis conducted on the medical records from primary care of over 17 million adults found that individuals diagnosed with autoimmune diseases like SLE, rheumatoid arthritis (RA), or psoriasis had heightened susceptibility to COVID-19-related mortality. It is important to note that this increased risk persists even after adjusting for demographic characteristics and comorbidities. Nonetheless, the study did not take into account medication use, nor did it assess SLE as a distinct disease.83
In an extensive analysis by Ugarte-Gil et al,83 the C19-GRA and European Alliance of Associations for Rheumatology (EULAR) COVID-19 registries were examined to assess the outcomes of patients with rheumatic diseases, including SLE, from March 2020 to June 2021. The study included a total of 1606 individuals with SLE. Using a multivariable model, the study reported that older age, male sex, high prednisone doses, absence of current treatment, comorbidities, and more severe COVID-19 outcomes were linked to moderate to high disease activity in SLE patients. Furthermore, after adjusting for sex and age, it was reported that MMF, rituximab, and CYC were linked to worse outcomes in comparison to hydroxychloroquine. On the other hand, methotrexate and belimumab were associated with favorable outcomes.
Type-I interferons (IFNs-I) are excessively produced in SLE patients, and they play an essential role in early viral infection control. Autoantibodies against interferon-α (IFN-α) have been observed in SLE patients, but their exact significance remains unclear, whether they are pathogenic, protective, or merely indicative of a general autoreactivity tendency.84–87 A cohort of over 600 SLE patients was examined for the presence of serum antibodies against IFN-α. Neutralizing and non-neutralizing IFN-α antibodies were found in 3.3% and 8.4% of individuals with SLE, respectively. Neutralizing antibodies were associated with decreased levels of IFN-α in the serum and a lower risk of developing active disease. However, they did increase the susceptibility to severe COVID-19 pneumonia and HZ. Severe COVID-19 pneumonia in SLE patients was primarily associated with the combined neutralization of various IFNs-I, but IFN-α autoantibodies did not affect the humoral immunogenicity of COVID-19 vaccines.88
Infections Caused by Treatment
Treatment choices for SLE are limited to corticosteroids and various immunosuppressive drugs. While these drugs demonstrate efficacy in managing SLE, they also raise the susceptibility to infections. Lupus patients treated with immunosuppressive agents appear to have a higher vulnerability to infections in comparison to individuals with other rheumatic diseases receiving similar treatment.89 This suggests that while immunosuppressive agents contribute to a higher infection risk, they are not the sole determining factor.89 Immunosuppressive treatment in patients with SLE has a double impact on immunity. While it suppresses the activity of abnormal cells, it can also normalize other features of the immune system. For instance, in untreated patients, the migration of the neutrophil is significantly decreased. However, treatment can normalize neutrophil migration,90 and high doses of corticosteroids can enhance Fc receptor-mediated mononuclear phagocyte function. Consequently, patients receiving therapy may demonstrate enhanced neutrophil migration and phagocytic function in contrast to untreated SLE patients.91 The risk of infection in SLE can vary according to the type of immunosuppression used. We will review studies investigating drugs used to treat patients with SLE and studies attempting to introduce new biologics and small drugs.
Hydroxychloroquine
Chloroquine and hydroxychloroquine were initially developed as antimalarial drugs, but they have also been proven to be effective in treating autoimmune diseases, particularly SLE. In addition to their antimalarial properties, they exhibit antibacterial, antifungal, and antiviral effects, achieved through a pH-dependent iron deficiency and the ability to increase lysosomal pH, which hampers the proliferation of intracellular organisms. Chloroquine and hydroxychloroquine demonstrate antibacterial activity against Staphylococcus aureus, Mycobacterium tuberculosis, Salmonella typhi, and Escherichia coli, while also exhibiting antifungal properties against Cryptococcus, Histoplasma, and Aspergillus.92
There is no evidence suggesting an enhanced risk of infection associated with the use of hydroxychloroquine. Yet, certain studies93-95 have reported a significant protective effect against infections in patients with SLE, including those with lupus nephritis (LN). Furthermore, a comprehensive analysis encompassing 44 trials involving more than 9000 patients revealed that treatment with hydroxy chloroquine resulted in a reduced susceptibility to infections when compared to other medications such as methotrexate, placebo, rituximab, low-dose belimumab, medium-dose belimumab, and high-dose belimumab. These results emphasize the potential benefits of hydroxy chloroquine in mitigating the risk of infection in SLE patients.3
Methotrexate
Methotrexate inhibits purine and thymidylic acid synthesis and interferes with DNA synthesis, repair, and cell replication.96 Despite the immunomodulatory properties of methotrexate, the doses used to treat rheumatologic diseases are not significantly immunosuppressive, and typically they do not lead to opportunistic infections unless the patients simultaneously receive high doses of steroids or other immunosuppressive agents.96 A meta-analysis of 12 randomized controlled trials with more than 1100 subjects reported that methotrexate was associated with an elevated risk of infections among patients with RA but not in people suffering from other inflammatory rheumatic diseases.97 In patients taking methotrexate who develop an infection, it is recommended to continue the medication for mild infections that do not require antibiotics; the same holds for patients going through low-risk surgery. For moderate infections necessitating antibiotics, it is advisable to withhold methotrexate until finishing the use of antibiotic and symptoms are resolved. In cases of severe infections requiring hospitalization or parenteral antibiotics, methotrexate should be suspended until antibiotic treatment is finished, inflammatory markers return to normal, and symptoms are solved. In patients with severe infections, particularly those with renal disease, early administration of intravenous folinic acid rescue may also prove advantageous.98
Azathioprine
Azathioprine is a prodrug that undergoes rapid metabolism in the intestinal tract, liver, and erythrocytes to form 6-mercaptopurine, which is responsible for the immunosuppressive and toxic effects of AZA.99 One-tenth of individuals with rheumatic diseases who are treated with AZA may experience infections.100 The likelihood of bacterial infections is heightened when leukopenia is present, while viral infections, notably HZ, can affect up to 6% of patients receiving treatment. Additionally, AZA may worsen chronic viral hepatitis in certain individuals.101
A systematic review and meta-analysis of 9898 participants reported that AZA demonstrated a higher level of safety when compared to glucocorticoids. However, it was observed that the combination of CYC followed by AZA increased the likelihood of experiencing infections. TAC exhibited superior efficacy in preventing serious infections compared to CYC, AZA, and MMF combined with TAC, as well as CYC followed by AZA.3 Currently, there are no established guidelines guiding when to discontinue AZA in the presence of infections. The determination should be personalized, considering factors like the gravity and recurrence of infections, existing medical conditions, and concurrent usage of other medications.
Mycophenolate
Mycophenolate (MMF) mofetil is a prodrug converted to mycophenolic acid (MPA) (active form). MPA inhibits the production of guanine nucleotides, impairs DNA synthesis, and consequently reduces lymphocyte proliferation and antibody production. There is evidence that it reduces fibroblast proliferation, resulting in antifibrotic activity.102
The occurrence of infections in individuals with rheumatic diseases who receive MMF treatment is supported by limited evidence. However, insights from studies in animal models103 and trials in patients with renal allografts104 suggest that MMF can offer protection against Pneumocystis jirovecii. Notably, though, the use of MMF has been linked to HZ in heart transplant recipients,105 and renal allograft recipients have an increased frequency of tissue-invasive CMV infections.106 It is important to acknowledge that these findings may not directly apply to individuals with rheumatologic conditions.
A retrospective study of SLE patients reported that UTIs were associated with the use of prednisone and CYC, while upper airway infections correlated with the use of prednisone, MMF, and cyclosporine. Glucocorticoids were generally associated with increased infection risk.107 In a meta-analysis, MMF showed lower overall infection risk compared to CYC in non-Asian populations treated for LN. Mycophenolate mofetil therapy should be avoided during active systemic or life-threatening infections due to its effects on the immune system and the potential risk of neutropenia.108
Cyclophosphamide
Cyclophosphamide is a DNA-alkylating agent that exerts cytotoxic effects on both replicating and resting lymphocytes. It reduces the number of B and T lymphocytes, leading to decreased lymphocyte proliferation, modulation of cell activation, and reduced antibody production. As a result, it suppresses cellular and humoral immunity.109 It increases the risk of infections by suppressing the bone marrow, which can result in neutropenia and/or lymphopenia. Additionally, CYC interferes with normal neutrophil and lymphocyte function, even in the absence of reductions in cell counts. Patients treated with CYC who develop neutropenia (defined as neutrophil < 1500/µL), especially when combined with high doses of glucocorticoids, are at a high risk of infection.110
A study of 100 patients with SLE found that infection occurred in 45% of CYC-treated patients compared to 12% of those treated with glucocorticoids alone. Bacterial infections were the most common, followed by opportunistic infections and HZ. Patients with infections were more likely to have multiple organ involvement, a lower nadir in the white blood cell count, and a higher maximum dose of corticosteroid than those without infection. Infections were equally prevalent in patients receiving oral or parenteral CYC but were more common when using sequential intravenous and oral therapy.110 The use of CYC therapy should be avoided in the presence of active systemic or potentially life-threatening infections. The use of CYC in neutropenic patients should also be avoided unless neutropenia is likely immune-mediated.
Rituximab
Rituximab is an IgG1 monoclonal antibody that binds to CD20, a B-cell differentiation marker.111 The biologic targets and depletes B cells from the pre-B to the mature B cell stage. Normalization of B cell numbers after treatment typically requires 6-9 months or longer.112
In a meta-analysis113 of 19 observational studies and 2 trials (EXPLORER114 and LUNAR115) which used rituximab to treat SLE patients, adverse reactions were detected in 16.8% of the patients. Among these events, infections were the most frequent, accounting for 63.1% of the cases.
In the EXPLORER trial,114 the group receiving rituximab had a higher occurrence of severe neutropenia events (grade 3 and grade 4) at a rate of 7.7% in comparison to the placebo group, which had a rate of 3.4%, and higher cases of grade 4 neutropenia (6 versus 0). However, there was no significant correlation between neutropenia events and the occurrence of infectious events. The ratio of patients encountering infectious adverse events was comparable between the rituximab and placebo groups, with rates of 82.2% and 83.0%, respectively. The most commonly reported type of infection in both groups was upper respiratory tract infections (URTIs), accounting for 49.1% in rituximab patients and 46.6% in placebo patients. It is worth noting that the rituximab group had an increased frequency of herpesvirus (15.4%) compared to the placebo group (8.0%), including rare cases of oral and genital outbreaks as well as HZ infections (9.5% in the rituximab group and 3.4% in the placebo group). However, most of the herpesvirus infections were resolved within 1 month in approximately 66% of patients. Regarding serious infections, sepsis occurred in 2.3% of patients who received placebo and 1% of patients who received rituximab. Additionally, the placebo group had a higher proportion of patients developing serious infections (17.0%) compared to the rituximab group (9.5%). In the LUNAR trial,115 within the rituximab group, there were 2 instances of death, one of which was attributed to sepsis resulting from a Staphylococcus aureus infection. Infections were reported in 62% of patients receiving rituximab and 64% of patients receiving placebo. Serious infections were observed in 19.2 patients in each group, including 3 cases of opportunistic infections. The hospitalization rates per 100 patients-year were similar between the 2 groups. In these trials, the most frequently observed infections involved the upper respiratory and urinary tracts or were caused by HZ.113
Obinutuzumab
Obinutuzumab is a type II anti-CD20 monoclonal antibody, distinct from the conventional type I antibody, rituximab. It binds to the CD20 antigen in a different manner, leading to enhanced antibody-dependent cellular cytotoxicity and direct elimination of B cells with reduced reliance on complement-dependent cytotoxicity. The NOBILITY trial, a randomized phase 2 study involving patients with LN, did not report a significant increase in serious infectious adverse events.116
Belimumab
Belimumab, an IgG1-lambda monoclonal antibody, neutralizes the B-lymphocyte stimulator (BLyS), inhibits the survival of B lymphocytes, and reduces B-cell-mediated immunity.117 A systematic analysis of 6 studies with 2917 participants aged between 22 and 80 years comparing belimumab to placebo treatment did not report differences in terms of infections between the treatment and control groups.118 Four studies involving 2185 participants119-122 also did not report statistically meaningful differences in the incidence of serious infections between the belimumab and the placebo groups. In the BLISS-76 study,119 a phase 3 randomized trial involving 819 patients, the proportions of serious or severe infections were similar between all treatment groups.
Interestingly, in 2018, Doria et al123 reported lower infection rates in patients treated with belimumab. Similar protective effects were reported in Asian patients with SLE receiving belimumab.124 In 2020, the BLISS-LN study, a 2-year randomized controlled trial of belimumab in LN, reported similar rates of serious infections across the treatment and control groups.124
Anifrolumab
Anifrolumab is a human monoclonal antibody designed to specifically target and inhibit the activity of type I IFN receptors, including IFN-α, IFN-β, and IFN-κ. These particular cytokines are frequently found at elevated levels in patients with lupus. By effectively inhibiting the function of IFNs-I, anifrolumab assists in diminishing the production of proinflammatory and immunomodulatory proteins associated with the immune response. This inhibition extends to the activation of B and T cells, the migration of immune cells, and the release of cytokines. Furthermore, anifrolumab decreases the expression of CD80 and CD83 on dendritic cells, providing additional correction of the aberrant immune responses in patients with SLE.125 Anifrolumab was approved by Food and Drug Administration (FDA) in 2021 for managing moderate to severe SLE patients who are already receiving standard therapy and do not have severe active LN or neuropsychiatric SLE.
Anifrolumab has been evaluated in several pivotal studies, including TULIP-1,126 TULIP-2,127 TULIP-LN,128 and MUSE.129 Across these studies, among patients treated with anifrolumab, the incidence of any adverse event varied from 85% to 89%, while the placebo groups exhibited rates ranging from 77% to 84%. The most commonly observed adverse events included URTI and nasopharyngitis. Notably, there was a higher frequency of HZ in the anifrolumab groups (5%-7%) compared to the placebo groups (1%-2%). However, the majority of HZ cases were not serious and did not necessitate cessation of treatment. All cases showed a favorable response to the appropriate treatment and generally resolved without any long-term complications.125
In the TULIP-LN study, a phase II randomized controlled study evaluating the efficacy and safety of anifrolumab in patients with active LN, the most common adverse events in the combined anifrolumab groups compared to the placebo group were HZ, UTI, and influenza. All HZ cases were cutaneous, with 13 localized and 3 disseminated. HZ cases tended to occur early in the trial and were successfully managed with conventional treatment. The occurrence of other adverse events of special interest was low in all of the treatment groups.128 An evaluation of tolerability in an extension study130 of those who participated in either TULIP 1 or TULIP 2 trials reported as most common adverse events, nasopharyngitis (9.7 versus 5.5 per 100 patient-year), UTI (8 versus 6), and URTI (8 versus 7) in the anifrolumab and placebo groups, respectively. Proportions of latent tuberculosis (2.3 versus 0.8) and influenza (2.2 versus 0.8) were enhanced in anifrolumab patients compared to placebo patients. It is worth noting that, in both groups, there were no occurrences of active tuberculosis, and in the anifrolumab group, no opportunistic infections were noted. During the 3 years, the rates of HZ per 100 patient-years were 3.4 in the anifrolumab group and 2.8 in the placebo group.
Litifilimab
Litifilimab is a humanized monoclonal antibody that specifically binds to the antibody-binding of blood dendritic cell antigen 2 (BDCA2), which is exclusively expressed on plasmacytoid dendritic cells. By targeting BDCA2, litifilimab suppresses the production of IFN-I. A phase II trial did not report alarmingly increased rates of infections.131
Tacrolimus, Cyclosporine, and Voclosporin
Cyclosporine and TAC are 2 immunosuppressive drugs used in the treatment of various immune-mediated diseases. Both drugs belong to the class of calcineurin inhibitors and exhibit similar suppressive effects on cell-mediated and humoral immune responses. While their primary action is on T helper cells, they may also inhibit T suppressor and T cytotoxic cells. Importantly, neither cyclosporine nor TAC causes significant clinical myelosuppression.132
In the context of treating LN or idiopathic membranous nephropathy, these drugs have been found to have a lower infection risk compared to glucocorticoids or other immunosuppressive agents. A meta-analysis of 38 randomized controlled trials involving 2066 patients reported that the combination of TAC and glucocorticoids was associated with a 48% lower risk of infection compared to intravenous CYC plus glucocorticoids. In contrast, intravenous CYC plus glucocorticoids was associated with a significantly higher risk of infection compared to TAC plus glucocorticoids, cyclosporine plus glucocorticoids, and oral CYC plus glucocorticoids.133 Similar results were reported by another meta-analysis,134 in which TAC showed a significantly decreased risk of serious infections when compared to glucocorticoids, CYC, MMF, and AZA. Overall, these studies highlight the relatively lower risk of infections associated with cyclosporine and TAC compared to other immunosuppressive drugs commonly used for treating SLE.
Voclosporin is a next-generation calcineurin inhibitor that shares structural similarities with cyclosporine, with a single amino acid difference that confers superior calcineurin inhibition and reduced plasma concentration variability. This distinction eliminates the need for therapeutic drug monitoring, which is typically required for other calcineurin inhibitors. In addition, voclosporin exhibits a more favorable effect on lipid and glucose concentrations compared to other drugs in the same class.135 It received FDA approval in January 2021 for the treatment of LN in combination with MMF and glucocorticoids.
In the AURORA-I trial, a phase III multicenter randomized controlled study, infections and infestations emerged as the most common adverse events observed in the voclosporin and placebo groups, affecting 65% and 57% of patients, respectively. Most of the reported infections were of mild and moderate severity.135 In the subsequent AURORA-II trial, which focused on evaluating the long-term safety and tolerability of voclosporin versus placebo in patients receiving treatment for an additional 24 months following the conclusion of the AURORA-I study, no unexpected safety concerns were identified in the voclosporin arm when compared to the control group. Moreover, similar proportions of serious adverse effects were reported in both groups.136
Atacicept
Atacicept, a fully human recombinant fusion protein, effectively inhibits B cell-stimulating factors, including APRIL (a proliferation-inducing ligand) and BLyS.137 The APRIL-LN study, a phase II/III, randomized, double-blind, placebo-controlled 52-week trial, was terminated prematurely after enrolling 6 patients due to an unexpected decline in serum immunoglobulin G (IgG) levels and the occurrence of serious infections137. The APRIL-SLE138 and ADDRESS II,139 double-blind, placebo-controlled 52-week trials that evaluated the safety of atacicept and did not alert for serious infections.
Abatacept
Abatacept, a selective costimulation modulator, targets CD80 and CD86 on antigen-presenting cells, leading to the inhibition of T-cell activation. This selectivity blocks the specific interaction between CD80/CD86 receptors and CD28, effectively suppressing T cell proliferation and the immune response of B cells.140,141
A multicenter, exploratory, double-blind, placebo-controlled phase IIb trial conducted over 2 months in SLE patients with polyarthritis, discoid lesions, pleuritis, and/or pericarditis reported a slightly higher rate of infections in the treatment group.140 Similar rates of infections were also reported in another 12-month randomized phase II/III trial.141
Ustekinumab
Ustekinumab is a monoclonal antibody that specifically targets the shared p40 subunit common to interleukins IL-12 and IL-23.142 In a phase II, multicenter, double-blind, randomized controlled trial, infections were the most commonly reported side effects, but they occurred equally in the treatment and control groups.142 Similarly, in the phase III study, which was terminated for lack of efficacy, the rate of infections was comparable in the treatment and control groups.143
Baricitinib
Baricitinib is an oral inhibitor of janus kinases (JAKs) 1 and 2, intracellular enzymes that participate in stimulating hematopoiesis and immune cell functions. Many critical cytokines implicated in the pathophysiology of lupus, including IFNs-I, IL-6, IL-12, and IL-23, depend on the activation of JAKs for intracellular signaling.144
In a 24-week phase II clinical trial evaluating the effectiveness, safety, and tolerance of oral baricitinib in individuals with active SLE, the occurrence of serious infections was increased in the baricitinib 4 mg group (6% of the patients) compared to both the 2 mg (2% of the patients) and the placebo groups (1%).144
Iberdomide
Iberdomide is a cereblon modulator that acts by promoting the degradation of the transcription factors Ikaros and Aiolos, which affect immune-cell development and homeostasis.145 In a phase II trial, the iberdomide groups experienced more frequent urinary, upper respiratory tract infections, and neutropenia in a dose-dependent manner. Infections with herpesvirus, fungi, and varicella-zoster virus were reported in the iberdomide groups.145
Deucravacitinib
Deucravacitinib is a selective, orally administered allosteric inhibitor that targets TYK2, an intracellular kinase that plays a crucial role in mediating the signaling of critical cytokines involved in the pathophysiology of lupus. These cytokines include IFNs-I, as well as IL-10, IL-12, and IL-23.146 A study that evaluated the efficacy of deucravacitinib reported increased rates of URTI and UTI but not significant differences in the incidence of HZ infections. Also, there were no cases of opportunistic infections or tuberculosis.146
Considerations to Mitigate Infections in Patients with Systemic Lupus Erythematosus
Preventive strategies are crucial to reduce the risk of infection in SLE patients. It is strongly advised that lupus patients receive immunizations as part of their preventive care. This includes vaccines against seasonal influenza, pneumococcal infections (both PCV13 and PPSV23), tetanus, HZ, and HPV. While the live vaccine Zostavax is available for HZ, a non-live vaccine known as Shingrix has been approved by the FDA in the United States since 2017. Shingrix is considered safer and more effective for preventing shingles in the general population,147 but there are no reports on its use in patients with SLE. Live attenuated vaccinations for measles, mumps, rubella, varicella zoster, and yellow fever should be evaluated for appropriate administration in select SLE patients before initiating therapy with immunosuppressive drugs.
Before starting treatment with immunosuppressive agents, it is crucial to identify and treat any chronic infections, like HIV, hepatitis B and C, and tuberculosis. This will help minimize the risk of worsening infections while receiving immunosuppressive therapy.
Due to the variable and unknown state of immunosuppression in SLE patients, any suspected infection should be treated promptly. An elevated C-reactive protein level may indicate a bacterial infection rather than a disease flare. Timely recognition and treatment of sepsis are essential. Validated scoring systems can help identify patients at higher risk of poor outcomes, allowing for more targeted interventions in emergency room settings and hospitals.148
Monitoring SLE patients for cytopenias induced by drugs and other adverse effects is crucial. This approach enables physicians to take an engaged participation in minimizing avoidable infections. Blood tests to assess immunoglobulin levels in individuals with SLE may also assist in recognizing patients with a higher risk of infections. In certain cases of severe infection or specific exposure to infectious agents, intravenous immunoglobulin may have a role in treating patients with hypogammaglobulinemia.1
For lupus patients receiving high doses of glucocorticoids (exceeding a daily dose of 30 mg of prednisone or its equivalent), prophylactic therapy to prevent Pneumocystis jirovecii infection is often recommended.
Implementing these preventive strategies and closely monitoring lupus patients can significantly reduce the risk of infections and their associated complications and improve the overall management of the disease.
Footnotes
Peer-review: Externally peer-reviewed.
Author Contributions: Concept – G.T., A.L.F.K.; Design – G.T., A.L.F.K.; Supervision – G.T.; Resources – G.T., A.L.F.K.; Materials – G.T., A.L.F.K.; Data Collection and/or Processing – A.L.F.K.; Analysis and/or Interpretation – G.T., A.L.F.K.; Literature Search – G.T., A.L.F.K.; Writing – G.T., A.L.F.K.; Critical Review – G.T.
Declaration of Interests: The authors have no conflict of interest to declare.
Funding: The authors declared that this study has received no financial support.
References
- 1. Tsokos GC. Autoimmunity and organ damage in systemic lupus erythematosus. Nat Immunol. 2020;21(6):605 614. ( 10.1038/s41590-020-0677-6) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Iliopoulos AG, Tsokos GC. Immunopathogenesis and spectrum of infections in systemic lupus erythematosus. Semin Arthritis Rheum. 1996;25(5):318 336. ( 10.1016/s0049-0172(96)80018-7) [DOI] [PubMed] [Google Scholar]
- 3. Tian J, Luo Y, Wu H, Long H, Zhao M, Lu Q. Risk of adverse events from different drugs for SLE: a systematic review and network meta-analysis. Lupus Sci Med. 2018;5(1):e000253. ( 10.1136/lupus-2017-000253) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Arnaud L, Tektonidou MG. Long-term outcomes in systemic lupus erythematosus: trends over time and major contributors. Rheumatol (Oxf Engl). 2020;59(suppl5):v29 v38. ( 10.1093/rheumatology/keaa382) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Roberts PJ, Isenberg DA, Segal AW. Defective degradation of bacterial DNA by phagocytes from patients with systemic and discoid lupus erythematosus. Clin Exp Immunol. 1987;69(1):68 78. [PMC free article] [PubMed] [Google Scholar]
- 6. Gyimesi E, Kavai M, Kiss E, Csipö I, Szücs G, Szegedi G. Triggering of respiratory burst by phagocytosis in monocytes of patients with systemic lupus erythematosus. Clin Exp Immunol. 1993;94(1):140 144. ( 10.1111/j.1365-2249.1993.tb05991.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Boros P, Muryoi T, Spiera H, Bona C, Unkeless JC. Autoantibodies directed against different classes of Fc gamma R are found in sera of autoimmune patients. J Immunol. 1993;150(5):2018 2024. [PubMed] [Google Scholar]
- 8. Kimberly RP, Salmon JE, Edberg JC. Receptors for immunoglobulin G. Molecular diversity and implications for disease. Arthritis Rheum. 1995;38(3):306 314. ( 10.1002/art.1780380303) [DOI] [PubMed] [Google Scholar]
- 9. Zurier RB. Reduction of phagocytosis and lysosomal enzyme release from human leukocytes by serum from patients with systemic lupus erythematosus. Arthritis Rheum. 1976;19(1):73 78. ( 10.1002/art.1780190112) [DOI] [PubMed] [Google Scholar]
- 10. Keeling DM, Isenberg DA. Haematological manifestations of systemic lupus erythematosus. Blood Rev. 1993;7(4):199 207. ( 10.1016/0268-960x(93)90006-p) [DOI] [PubMed] [Google Scholar]
- 11. Nived O, Linder C, Odeberg H, Svensson B. Reduced opsonisation of protein A containing Staphylococcus aureus in sera with cryoglobulins from patients with active systemic lupus erythematosus. Ann Rheum Dis. 1985;44(4):252 259. ( 10.1136/ard.44.4.252) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Rustagi PK, Currie MS, Logue GL. Complement-activating antineutrophil antibody in systemic lupus erythematosus. Am J Med. 1985;78(6 Pt 1):971 977. ( 10.1016/0002-9343(85)90220-7) [DOI] [PubMed] [Google Scholar]
- 13. Hartman KR, Wright DG. Identification of autoantibodies specific for the neutrophil adhesion glycoproteins CD11b/CD18 in patients with autoimmune neutropenia. Blood. 1991;78(4):1096 1104. [PubMed] [Google Scholar]
- 14. Hartman KR, LaRussa VF, Rothwell SW, Atolagbe TO, Ward FT, Klipple G. Antibodies to myeloid precursor cells in autoimmune neutropenia. Blood. 1994;84(2):625 631. [PubMed] [Google Scholar]
- 15. Yu Y, Su K. Neutrophil extracellular traps and systemic lupus erythematosus. J Clin Cell Immunol. 2013;4:139. ( 10.4172/2155-9899.1000139) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hsieh SC, Wu TH, Tsai CY, et al. Abnormal in vitro CXCR2 modulation and defective cationic ion transporter expression on polymorphonuclear neutrophils responsible for hyporesponsiveness to IL-8 stimulation in patients with active systemic lupus erythematosus. Rheumatol (Oxf Engl). 2008;47(2):150 157. ( 10.1093/rheumatology/kem320) [DOI] [PubMed] [Google Scholar]
- 17. Boumpas DT, Eleftheriades EG, Tsokos GC. Cellular immunity in systemic lupus erythematosus. In vivo (Athens Greece). 1988;2(1):41 45. [PubMed] [Google Scholar]
- 18. Erkeller-Yüsel F, Hulstaart F, Hannet I, Isenberg D, Lydyard P. Lymphocyte subsets in a large cohort of patients with systemic lupus erythematosus. Lupus. 1993;2(4):227 231. ( 10.1177/096120339300200404) [DOI] [PubMed] [Google Scholar]
- 19. Henriques A, Teixeira L, Inês L, et al. NK cells dysfunction in systemic lupus erythematosus: relation to disease activity. Clin Rheumatol. 2013;32(6):805 813. ( 10.1007/s10067-013-2176-8) [DOI] [PubMed] [Google Scholar]
- 20. Nived O, Johansson I, Sturfelt G. Effects of ultraviolet irradiation on natural killer cell function in systemic lupus erythematosus. Ann Rheum Dis. 1992;51(6):726 730. ( 10.1136/ard.51.6.726) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Tsokos GC. Lymphocyte abnormalities in human lupus. Clin Immunol Immunopathol. 1992;63(1):7 9. ( 10.1016/0090-1229(92)90083-z) [DOI] [PubMed] [Google Scholar]
- 22. Winfield JB, Mimura T. Pathogenetic significance of anti-lymphocyte autoantibodies in systemic lupus erythematosus. Clin Immunol Immunopathol. 1992;63(1):13 16. ( 10.1016/0090-1229(92)90085-3) [DOI] [PubMed] [Google Scholar]
- 23. Stohl W. Impaired polyclonal T cell cytolytic activity. A possible risk factor for systemic lupus erythematosus. Arthritis Rheum. 1995;38(4):506 516. ( 10.1002/art.1780380408) [DOI] [PubMed] [Google Scholar]
- 24. Stohl W. Impaired generation of polyclonal T cell-mediated cytolytic activity despite normal polyclonal T cell proliferation in systemic lupus erythematosus. Clin Immunol Immunopathol. 1992;63(2):163 172. ( 10.1016/0090-1229(92)90009-d) [DOI] [PubMed] [Google Scholar]
- 25. Via CS, Tsokos GC, Bermas B, Clerici M, Shearer GM. T cell-antigen-presenting cell interactions in human systemic lupus erythematosus. Evidence for heterogeneous expression of multiple defects. J Immunol. 1993;151(7):3914 3922. [PubMed] [Google Scholar]
- 26. Nalbandian A, Crispín JC, Tsokos GC. Interleukin-17 and systemic lupus erythematosus: current concepts. Clin Exp Immunol. 2009;157(2):209 215. ( 10.1111/j.1365-2249.2009.03944.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kang I, Quan T, Nolasco H, et al. Defective control of latent Epstein-Barr virus infection in systemic lupus erythematosus. J Immunol. 2004;172(2):1287 1294. ( 10.4049/jimmunol.172.2.1287) [DOI] [PubMed] [Google Scholar]
- 28. Lieberman LA, Tsokos GC. Lupus-prone mice fail to raise antigen-specific T cell responses to intracellular infection. PLoS One. 2014;9(10):e111382. ( 10.1371/journal.pone.0111382) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Alcocer-Varela J, Alarcón-Riquelme M, Laffón A, Sánchez-Madrid F, Alarcón-Segovia D. Activation markers on peripheral blood T cells from patients with active or inactive systemic lupus erythematosus. Correlation with proliferative responses and production of IL-2. J Autoimmun. 1991;4(6):935 945. ( 10.1016/0896-8411(91)90056-i) [DOI] [PubMed] [Google Scholar]
- 30. Erkeller-Yuksel FM, Lydyard PM, Isenberg DA. Lack of NK cells in lupus patients with renal involvement. Lupus. 1997;6(9):708 712. ( 10.1177/096120339700600905) [DOI] [PubMed] [Google Scholar]
- 31. Pavón EJ, Muñoz P, Navarro MD, et al. Increased association of CD38 with lipid rafts in T cells from patients with systemic lupus erythematosus and in activated normal T cells. Mol Immunol. 2006;43(7):1029 1039. ( 10.1016/j.molimm.2005.05.002) [DOI] [PubMed] [Google Scholar]
- 32. Aksoy P, White TA, Thompson M, Chini EN. Regulation of intracellular levels of NAD: a novel role for CD38. Biochem Biophys Res Commun. 2006;345(4):1386 1392. ( 10.1016/j.bbrc.2006.05.042) [DOI] [PubMed] [Google Scholar]
- 33. Chini EN. CD38 as a regulator of cellular NAD: a novel potential pharmacological target for metabolic conditions. Curr Pharm Des. 2009;15(1):57 63. ( 10.2174/138161209787185788) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Malavasi F, Deaglio S, Funaro A, et al. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol Rev. 2008;88(3):841 886. ( 10.1152/physrev.00035.2007) [DOI] [PubMed] [Google Scholar]
- 35. Katsuyama E, Suarez-Fueyo A, Bradley SJ, et al. The CD38/NAD/SIRTUIN1/EZH2 axis mitigates cytotoxic CD8 T cell function and identifies patients with SLE prone to infections. Cell Rep. 2020;30(1):112 123.e4. ( 10.1016/j.celrep.2019.12.014) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Chen PM, Katsuyama E, Satyam A, et al. CD38 reduces mitochondrial fitness and cytotoxic T cell response against viral infection in lupus patients by suppressing mitophagy. Sci Adv. 2022;8(24):eabo4271. ( 10.1126/sciadv.abo4271) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Tsokos GC, Smith PL, Christian CB, Lipnick RN, Balow JE, Djeu JY. Interleukin-2 restores the depressed allogeneic cell-mediated lympholysis and natural killer cell activity in patients with systemic lupus erythematosus. Clin Immunol Immunopathol. 1985;34(3):379 386. ( 10.1016/0090-1229(85)90186-2) [DOI] [PubMed] [Google Scholar]
- 38. Zhou P, Chen J, He J, et al. Low-dose IL-2 therapy invigorates CD8+ T cells for viral control in systemic lupus erythematosus. PLoS Pathog. 2021;17(10):e1009858. ( 10.1371/journal.ppat.1009858) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Mitchell SR, Nguyen PQ, Katz P. Increased risk of neisserial infections in systemic lupus erythematosus. Semin Arthritis Rheum. 1990;20(3):174 184. ( 10.1016/0049-0172(90)90058-n) [DOI] [PubMed] [Google Scholar]
- 40. Wilson JG, Ratnoff WD, Schur PH, Fearon DT. Decreased expression of the C3b/C4b receptor (CR1) and the C3d receptor (CR2) on B lymphocytes and of CR1 on neutrophils of patients with systemic lupus erythematosus. Arthritis Rheum. 1986;29(6):739 747. ( 10.1002/art.1780290606) [DOI] [PubMed] [Google Scholar]
- 41. Yoshida K, Yukiyama Y, Miyamoto T. Quantification of the complement receptor function on polymorphonuclear leukocytes: its significance in patients with systemic lupus erythematosus. J Rheumatol. 1987;14(3):490 496. [PubMed] [Google Scholar]
- 42. Mok MY, Ip WKE, Lau CS, Lo Y, Wong WHS, Lau YL. Mannose-binding lectin and susceptibility to infection in Chinese patients with systemic lupus erythematosus. J Rheumatol. 2007;34(6):1270 1276. [PubMed] [Google Scholar]
- 43. Ahearn JM, Fearon DT. Structure and function of the complement receptors, CR1 (CD35) and CR2 (CD21). Adv Immunol. 1989;46:183 219. ( 10.1016/s0065-2776(08)60654-9) [DOI] [PubMed] [Google Scholar]
- 44. Aguado MT, Lambris JD, Tsokos GC, et al. Monoclonal antibodies against complement 3 neoantigens for detection of immune complexes and complement activation. Relationship between immune complex levels, state of C3, and numbers of receptors for C3b. J Clin Invest. 1985;76(4):1418 1426. ( 10.1172/JCI112119) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Yu CL, Tsai CY, Chiu CC, et al. Defective expression of neutrophil C3b receptors and impaired lymphocyte Na(+)-K(+)-ATPase activity in patients with systemic lupus erythematosus. Proc Natl Sci Counc Repub China B. 1991;15(3):178 185. [PubMed] [Google Scholar]
- 46. Alejandro C, Villaseor-Ovies P. Infections and systemic lupus erythematosus [internet] (Almoallim H, ed.). InTech. Accessed 10, 2023. http://www.intechopen.com/books/systemic-lupus-erythematosus/infectious-in-systemic-lupus [Google Scholar]
- 47. Frank MM, Hamburger MI, Lawley TJ, Kimberly RP, Plotz PH. Defective reticuloendothelial system Fc-receptor function in systemic lupus erythematosus. N Engl J Med. 1979;300(10):518 523. ( 10.1056/NEJM197903083001002) [DOI] [PubMed] [Google Scholar]
- 48. Piliero P, Furie R. Functional asplenia in systemic lupus erythematosus. Semin Arthritis Rheum. 1990;20(3):185 189. ( 10.1016/0049-0172(90)90059-o) [DOI] [PubMed] [Google Scholar]
- 49. Anand AJ, Glatt AE. Salmonella osteomyelitis and arthritis in sickle cell disease. Semin Arthritis Rheum. 1994;24(3):211 221. ( 10.1016/0049-0172(94)90076-0) [DOI] [PubMed] [Google Scholar]
- 50. Tektonidou MG, Wang Z, Dasgupta A, Ward MM. Burden of serious infections in adults with systemic lupus erythematosus: a national population-based study, 1996-2011. Arthritis Care Res. 2015;67(8):1078 1085. ( 10.1002/acr.22575) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Singh BK, Singh S. Systemic lupus erythematosus and infections. Reumatismo. 2020;72(3):154 169. ( 10.4081/reumatismo.2020.1303) [DOI] [PubMed] [Google Scholar]
- 52. Dhital R, Pandey RK, Poudel DR, Oladunjoye O, Paudel P, Karmacharya P. All-cause hospitalizations and mortality in systemic lupus erythematosus in the US: results from a national inpatient database. Rheumatol Int. 2020;40(3):393 397. ( 10.1007/s00296-019-04484-5) [DOI] [PubMed] [Google Scholar]
- 53. Wallace DJ, Podell T, Weiner J, Klinenberg JR, Forouzesh S, Dubois EL. Systemic lupus erythematosus—survival patterns: experience with 609 patients. JAMA. 1981;245(9):934 938. ( 10.1001/jama.245.9.934) [DOI] [PubMed] [Google Scholar]
- 54. Rosner S, Ginzler EM, Diamond HS, et al. A multicenter study of outcome in systemic lupus erythematosus. II. Causes of death. Arthritis Rheum. 1982;25(6):612 617. ( 10.1002/art.1780250602) [DOI] [PubMed] [Google Scholar]
- 55. Huicochea Grobet ZL, Berrón R, Ortega Martell JA, Onuma E. Survival up to 5 and 10 years of Mexican pediatric patients with systemic lupus erythematosus. Overhaul of 23 years experience. Allergol Immunopathol, Madr. 1996;24(1):36 38. [PubMed] [Google Scholar]
- 56. Kim WU, Min JK, Lee SH, Park SH, Cho CS, Kim HY. Causes of death in Korean patients with systemic lupus erythematosus: a single center retrospective study. Clin Exp Rheumatol. 1999;17(5):539 545. [PubMed] [Google Scholar]
- 57. Jacobsen S, Petersen J, Ullman S, et al. Mortality and causes of death of 513 Danish patients with systemic lupus erythematosus. Scand J Rheumatol. 1999;28(2):75 80. ( 10.1080/030097499442522) [DOI] [PubMed] [Google Scholar]
- 58. Mok CC, Lee KW, Ho CT, Lau CS, Wong RW. A prospective study of survival and prognostic indicators of systemic lupus erythematosus in a southern Chinese population. Rheumatol (Oxf Engl). 2000;39(4):399 406. ( 10.1093/rheumatology/39.4.399) [DOI] [PubMed] [Google Scholar]
- 59. Rodríguez VE, González-Parés EN. Mortality study in Puerto Ricans with systemic lupus erythematosus. P R Health Sci J. 2000;19(4):335 339. [PubMed] [Google Scholar]
- 60. Bernatsky S, Boivin JF, Joseph L, et al. Mortality in systemic lupus erythematosus. Arthritis Rheum. 2006;54(8):2550 2557. ( 10.1002/art.21955) [DOI] [PubMed] [Google Scholar]
- 61. Wadee S, Tikly M, Hopley M. Causes and predictors of death in South Africans with systemic lupus erythematosus. Rheumatol (Oxf Engl). 2007;46(9):1487 1491. ( 10.1093/rheumatology/kem180) [DOI] [PubMed] [Google Scholar]
- 62. Nossent J, Cikes N, Kiss E, et al. Current causes of death in systemic lupus erythematosus in Europe, 2000-2004: relation to disease activity and damage accrual. Lupus. 2007;16(5):309 317. ( 10.1177/0961203307077987) [DOI] [PubMed] [Google Scholar]
- 63. Al Arfaj AS, Khalil N. Clinical and immunological manifestations in 624 SLE patients in Saudi Arabia. Lupus. 2009;18(5):465 473. ( 10.1177/0961203308100660) [DOI] [PubMed] [Google Scholar]
- 64. Goldblatt F, Chambers S, Rahman A, Isenberg DA. Serious infections in British patients with systemic lupus erythematosus: hospitalisations and mortality. Lupus. 2009;18(8):682 689. ( 10.1177/0961203308101019) [DOI] [PubMed] [Google Scholar]
- 65. Feng X, Zou Y, Pan W, et al. Prognostic indicators of hospitalized patients with systemic lupus erythematosus: a large retrospective multicenter study in China. J Rheumatol. 2011;38(7):1289 1295. ( 10.3899/jrheum.101088) [DOI] [PubMed] [Google Scholar]
- 66. McElhone K, Burnell J, Sutton C, et al. Is the disease-specific Lupusqol sensitive to changes of disease activity in systemic lupus erythematosus patients after treatment of A flare? Value Health. 2014;17(7):A538. ( 10.1016/j.jval.2014.08.1725) [DOI] [PubMed] [Google Scholar]
- 67. Dubula T, Mody GM. Spectrum of infections and outcome among hospitalized South Africans with systemic lupus erythematosus. Clin Rheumatol. 2015;34(3):479 488. ( 10.1007/s10067-014-2847-0) [DOI] [PubMed] [Google Scholar]
- 68. Chen D, Xie J, Chen H, et al. Infection in Southern Chinese patients with systemic lupus erythematosus: spectrum, drug resistance, outcomes, and risk factors. J Rheumatol. 2016;43(9):1650 1656. ( 10.3899/jrheum.151523) [DOI] [PubMed] [Google Scholar]
- 69. Budhoo A, Mody GM, Dubula T, Patel N, Mody PG. Comparison of ethnicity, gender, age of onset and outcome in South Africans with systemic lupus erythematosus. Lupus. 2017;26(4):438 446. ( 10.1177/0961203316676380) [DOI] [PubMed] [Google Scholar]
- 70. Padjen I, Cerovec M, Erceg M, Mayer M, Stevanović R, Anić B. Disease characteristics and causes of early and late death in a group of Croatian patients with systemic lupus erythematosus deceased over a 10-year period. Croat Med J. 2018;59(1):3 12. ( 10.3325/cmj.2018.59.3) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Wu XY, Yang M, Xie YS, et al. Causes of death in hospitalized patients with systemic lupus erythematosus: a 10-year multicenter nationwide Chinese cohort. Clin Rheumatol. 2019;38(1):107 115. ( 10.1007/s10067-018-4259-z) [DOI] [PubMed] [Google Scholar]
- 72. Ingvarsson RF, Landgren AJ, Bengtsson AA, Jönsen A. Good survival rates in systemic lupus erythematosus in southern Sweden, while the mortality rate remains increased compared with the population. Lupus. 2019;28(12):1488 1494. ( 10.1177/0961203319877947) [DOI] [PubMed] [Google Scholar]
- 73. Gonzalez Lucero L, Barbaglia AL, Bellomio VI, et al. Prevalence and incidence of systemic lupus erythematosus in Tucumán, Argentina. Lupus. 2020;29(13):1815 1820. ( 10.1177/0961203320957719) [DOI] [PubMed] [Google Scholar]
- 74. Bultink IEM, de Vries F, van Vollenhoven RF, Lalmohamed A. Mortality, causes of death and influence of medication use in patients with systemic lupus erythematosus vs matched controls. Rheumatology. 2021;60(1):207 216. ( 10.1093/rheumatology/keaa267) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Moghaddam B, Marozoff S, Li L, Sayre EC. Zubieta JAA-. All-cause and cause-specific mortality in systemic lupus erythematosus: a population-based study. Rheumatol (Oxf Engl). 2021;61(1):367 376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Al-Rayes H, Al-Swailem R, Arfin M, Sobki S, Rizvi S, Tariq M. Systemic lupus erythematosus and infections: a retrospective study in Saudis. Lupus. 2007;16(9):755 763. ( 10.1177/0961203307079943) [DOI] [PubMed] [Google Scholar]
- 77. Houman MH, Smiti-Khanfir M, Ben Ghorbell I, Miled M. Systemic lupus erythematosus in Tunisia: demographic and clinical analysis of 100 patients. Lupus. 2004;13(3):204 211. ( 10.1191/0961203303lu530xx) [DOI] [PubMed] [Google Scholar]
- 78. Battaglia M, Garrett-Sinha LA. Bacterial infections in lupus: roles in promoting immune activation and in pathogenesis of the disease. J Transl Autoimmun. 2021;4:100078. ( 10.1016/j.jtauto.2020.100078) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Kang TY, Lee HS, Kim TH, Jun JB, Yoo DH. Clinical and genetic risk factors of herpes zoster in patients with systemic lupus erythematosus. Rheumatol Int. 2005;25(2):97 102. ( 10.1007/s00296-003-0403-3) [DOI] [PubMed] [Google Scholar]
- 80. Su CF, Lai CC, Li TH, et al. Epidemiology and risk of invasive fungal infections in systemic lupus erythematosus: a nationwide population-based cohort study. Ther Adv Musculoskelet Dis. 2021;13:1759720X211058502. ( 10.1177/1759720X211058502) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Fernandez-Ruiz R, Paredes JL, Niewold TB. COVID-19 in patients with systemic lupus erythematosus: lessons learned from the inflammatory disease. Transl Res. 2021;232:13 36. ( 10.1016/j.trsl.2020.12.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Strangfeld A, Schäfer M, Gianfrancesco MA, et al. Factors associated with COVID-19-related death in people with rheumatic diseases: results from the COVID-19 Global Rheumatology Alliance physician-reported registry. Ann Rheum Dis. 2021;80(7):930 942. ( 10.1136/annrheumdis-2020-219498) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ugarte-Gil MF, Alarcón GS, Izadi Z, et al. Characteristics associated with poor COVID-19 outcomes in individuals with systemic lupus erythematosus: data from the COVID-19 Global Rheumatology Alliance. Ann Rheum Dis. 2022;81(7):970 978. ( 10.1136/annrheumdis-2021-221636) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Slavikova M, Schmeisser H, Kontsekova E, Mateička F, Borecky L, Kontsek P. Incidence of autoantibodies against type I and Type II interferons in a cohort of systemic lupus erythematosus patients in Slovakia. J Interferon Cytokine Res. 2003;23(3):143 147. ( 10.1089/107999003321532475) [DOI] [PubMed] [Google Scholar]
- 85. Morimoto AM, Flesher DT, Yang J, et al. Association of endogenous anti-interferon-α autoantibodies with decreased interferon-pathway and disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63(8):2407 2415. ( 10.1002/art.30399) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Gupta S, Tatouli IP, Rosen LB, et al. Distinct functions of autoantibodies against interferon in systemic lupus erythematosus: A comprehensive analysis of anticytokine autoantibodies in common rheumatic diseases. Arthritis Rheumatol. 2016;68(7):1677 1687. ( 10.1002/art.39607) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. von Wussow P, Jakschies D, Hartung K, Deicher H. Presence of interferon and anti-interferon in patients with systemic lupus erythematosus. Rheumatol Int. 1988;8(5):225 230. ( 10.1007/BF00269199) [DOI] [PubMed] [Google Scholar]
- 88. Mathian A, Breillat P, Dorgham K, et al. Lower disease activity but higher risk of severe COVID-19 and herpes zoster in patients with systemic lupus erythematosus with pre-existing autoantibodies neutralising IFN-α. Ann Rheum Dis. 2022;81(12):1695 1703. ( 10.1136/ard-2022-222549) [DOI] [PubMed] [Google Scholar]
- 89. Cohen J, Pinching AJ, Rees AJ, Peters DK. Infection and immunosuppression. A study of the infective complications of 75 patients with immunologically-mediated disease. Q J Med. 1982;51(201):1 15. [PubMed] [Google Scholar]
- 90. Al-Hadithy H, Isenberg DA, Addison IE, Goldstone AH, Snaith ML. Neutrophil function in systemic lupus erythematosus and other collagen diseases. Ann Rheum Dis. 1982;41(1):33 38. ( 10.1136/ard.41.1.33) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Salmon JE, Kapur S, Meryhew NL, Runquist OA, Kimberly RP. High-dose, pulse intravenous methylprednisolone enhances Fc gamma receptor-mediated mononuclear phagocyte function in systemic lupus erythematosus. Arthritis Rheum. 1989;32(6):717 725. ( 10.1002/anr.1780320609) [DOI] [PubMed] [Google Scholar]
- 92. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol. 2020;16(3):155 166. ( 10.1038/s41584-020-0372-x) [DOI] [PubMed] [Google Scholar]
- 93. Ruiz-Irastorza G, Olivares N, Ruiz-Arruza I, Martinez-Berriotxoa A, Egurbide MV, Aguirre C. Predictors of major infections in systemic lupus erythematosus. Arthritis Res Ther. 2009;11(4):R109. ( 10.1186/ar2764) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Sisó A, Ramos-Casals M, Bové A, et al. Previous antimalarial therapy in patients diagnosed with lupus nephritis: influence on outcomes and survival. Lupus. 2008;17(4):281 288. ( 10.1177/0961203307086503) [DOI] [PubMed] [Google Scholar]
- 95. Bultink IE, Hamann D, Seelen MA, et al. Deficiency of functional mannose-binding lectin is not associated with infections in patients with systemic lupus erythematosus. Arthritis Res Ther. 2006;8(6):R183. ( 10.1186/ar2095) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Cronstein BN. Molecular therapeutics. Methotrexate and its mechanism of action. Arthritis Rheum. 1996;39(12):1951 1960. ( 10.1002/art.1780391203) [DOI] [PubMed] [Google Scholar]
- 97. Ibrahim A, Ahmed M, Conway R, Carey JJ. Risk of infection with methotrexate therapy in inflammatory diseases: a systematic review and meta-analysis. J Clin Med. 2018;8(1):15. ( 10.3390/jcm8010015) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. McLean-Tooke A, Aldridge C, Waugh S, Spickett GP, Kay L. Methotrexate, rheumatoid arthritis and infection risk: what is the evidence? Rheumatol (Oxf Engl). 2009;48(8):867 871. ( 10.1093/rheumatology/kep101) [DOI] [PubMed] [Google Scholar]
- 99. Huskisson EC. Azathioprine. Clin Rheum Dis. 1984;10(2):325 332. ( 10.1016/S0307-742X(21)00505-1) [DOI] [PubMed] [Google Scholar]
- 100. McKendry RJR. Pruine analogues. Second line agents in the treatment of rheumatic diseases. New York: Marcel Decker; 1991. [Google Scholar]
- 101. Singh G, Fries JF, Spitz P, Williams CA. Toxic effects of azathioprine in rheumatoid arthritis. A national post-marketing perspective. Arthritis Rheum. 1989;32(7):837 843. ( 10.1002/j.2326-5205.1989.tb00014.x) [DOI] [PubMed] [Google Scholar]
- 102. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 2000;47(2-3):85 118. ( 10.1016/s0162-3109(00)00188-0) [DOI] [PubMed] [Google Scholar]
- 103. Oz HS, Hughes WT. Novel anti-pneumocystis carinii effects of the immunosuppressant mycophenolate mofetil in contrast to provocative effects of tacrolimus, sirolimus, and dexamethasone. J Infect Dis. 1997;175(4):901 904. ( 10.1086/513988) [DOI] [PubMed] [Google Scholar]
- 104. Husain S, Singh N. The impact of novel immunosuppressive agents on infections in organ transplant recipients and the interactions of these agents with antimicrobials. Clin Infect Dis. 2002;35(1):53 61. ( 10.1086/340867) [DOI] [PubMed] [Google Scholar]
- 105. Kobashigawa J, Miller L, Renlund D, et al. A randomized active-controlled trial of mycophenolate mofetil in heart transplant recipients. Mycophenolate mofetil investigators. Transplantation. 1998;66(4):507 515. ( 10.1097/00007890-199808270-00016) [DOI] [PubMed] [Google Scholar]
- 106. Sarmiento JM, Dockrell DH, Schwab TR, Munn SR, Paya CV. Mycophenolate mofetil increases cytomegalovirus invasive organ disease in renal transplant patients. Clin Transplant. 2000;14(2):136 138. ( 10.1034/j.1399-0012.2000.140206.x) [DOI] [PubMed] [Google Scholar]
- 107. Skare TL, Dagostini JS, Zanardi PI, Nisihara RM. Infections and systemic lupus erythematosus. Einstein (Sao Paulo). 2016;14(1):47 51. ( 10.1590/S1679-45082016AO3490) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Thong KM, Chan TM. Infectious complications in lupus nephritis treatment: a systematic review and meta-analysis. Lupus. 2019;28(3):334 346. ( 10.1177/0961203319829817) [DOI] [PubMed] [Google Scholar]
- 109. Hall AG, Tilby MJ. Mechanisms of action of, and modes of resistance to, alkylating agents used in the treatment of haematological malignancies. Blood Rev. 1992;6(3):163 173. ( 10.1016/0268-960x(92)90028-o) [DOI] [PubMed] [Google Scholar]
- 110. Pryor BD, Bologna SG, Kahl LE. Risk factors for serious infection during treatment with cyclophosphamide and high-dose corticosteroids for systemic lupus erythematosus. Arthritis Rheum. 1996;39(9):1475 1482. ( 10.1002/art.1780390906) [DOI] [PubMed] [Google Scholar]
- 111. Reff ME, Carner K, Chambers KS, et al. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood. 1994;83(2):435 445. [PubMed] [Google Scholar]
- 112. Cragg MS, Walshe CA, Ivanov AO, Glennie MJ. The biology of CD20 and its potential as a target for mAb therapy. Curr Dir Autoimmun. 2005;8:140 174. ( 10.1159/000082102) [DOI] [PubMed] [Google Scholar]
- 113. Lan L, Han F, Chen JH. Efficacy and safety of rituximab therapy for systemic lupus erythematosus: a systematic review and meta-analysis. J Zhejiang Univ Sci B. 2012;13(9):731 744. ( 10.1631/jzus.B1200057) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Merrill JT, Neuwelt CM, Wallace DJ, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010;62(1):222 233. ( 10.1002/art.27233) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Rovin BH, Furie R, Latinis K, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the lupus Nephritis Assessment with rituximab study. Arthritis Rheum. 2012;64(4):1215 1226. ( 10.1002/art.34359) [DOI] [PubMed] [Google Scholar]
- 116. Furie RA, Aroca G, Cascino MD, et al. B-cell depletion with obinutuzumab for the treatment of proliferative lupus nephritis: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2022;81(1):100 107. ( 10.1136/annrheumdis-2021-220920) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Furie R, Stohl W, Ginzler EM, et al. Biologic activity and safety of Belimumab, a neutralizing anti-B-lymphocyte stimulator (BLyS) monoclonal antibody: a phase I trial in patients with systemic lupus erythematosus. Arthritis Res Ther. 2008;10(5):R109. ( 10.1186/ar2506) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Singh JA, Shah NP, Mudano AS. Belimumab for systemic lupus erythematosus. Cochrane Database Syst Rev. 2021;2(2):CD010668. ( 10.1002/14651858.CD010668.pub2) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Furie R, Petri M, Zamani O, et al. A phase III, randomized, placebo-controlled study of Belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63(12):3918 3930. ( 10.1002/art.30613) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Navarra SV, Guzmán RM, Gallacher AE, et al. Efficacy and safety of Belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9767):721 731. ( 10.1016/S0140-6736(10)61354-2) [DOI] [PubMed] [Google Scholar]
- 121. Stohl W, Schwarting A, Okada M, et al. Efficacy and safety of subcutaneous Belimumab in systemic lupus erythematosus: a fifty-two-week randomized, double-blind, placebo-controlled study. Arthritis Rheumatol. 2017;69(5):1016 1027. ( 10.1002/art.40049) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Wallace DJ, Stohl W, Furie RA, et al. A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of Belimumab in patients with active systemic lupus erythematosus. Arthritis Rheum. 2009;61(9):1168 1178. ( 10.1002/art.24699) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Doria A, Bass D, Schwarting A, et al. A 6-month open-label extension study of the safety and efficacy of subcutaneous Belimumab in patients with systemic lupus erythematosus. Lupus. 2018;27(9):1489 1498. ( 10.1177/0961203318777634) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Zhang F, Bae SC, Bass D, et al. A pivotal phase III, randomised, placebo-controlled study of Belimumab in patients with systemic lupus erythematosus located in China, Japan and South Korea. Ann Rheum Dis. 2018;77(3):355 363. ( 10.1136/annrheumdis-2017-211631) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Riggs JM, Hanna RN, Rajan B, et al. Characterisation of anifrolumab, a fully human anti-interferon receptor antagonist antibody for the treatment of systemic lupus erythematosus. Lupus Sci Med. 2018;5(1):e000261. ( 10.1136/lupus-2018-000261) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Furie RA, Morand EF, Bruce IN, et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial. Lancet Rheumatol. 2019;1(4):e208 e219. ( 10.1016/S2665-9913(19)30076-1) [DOI] [PubMed] [Google Scholar]
- 127. Morand EF, Furie R, Tanaka Y, et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med. 2020;382(3):211 221. ( 10.1056/NEJMoa1912196) [DOI] [PubMed] [Google Scholar]
- 128. Jayne D, Rovin B, Mysler EF, et al. Phase II randomised trial of type I interferon inhibitor anifrolumab in patients with active lupus nephritis. Ann Rheum Dis. 2022;81(4):496 506. ( 10.1136/annrheumdis-2021-221478) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Furie R, Khamashta M, Merrill JT, et al. Anifrolumab, an anti-interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol. 2017;69(2):376 386. ( 10.1002/art.39962) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Kalunian KC, Furie R, Morand EF, et al. A randomized, placebo-controlled Phase III extension trial of the long-term safety and tolerability of anifrolumab in active systemic lupus erythematosus. Arthritis Rheumatol. 2023;75(2):253 265. ( 10.1002/art.42392) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Furie RA, van Vollenhoven RF, Kalunian K, et al. Trial of anti-BDCA2 antibody Litifilimab for systemic lupus erythematosus. N Engl J Med. 2022;387(10):894 904. ( 10.1056/NEJMoa2118025) [DOI] [PubMed] [Google Scholar]
- 132. Wiederrecht G, Lam E, Hung S, Martin M, Sigal N. The mechanism of action of FK-506 and cyclosporin A. Ann N Y Acad Sci. 1993;696:9 19. ( 10.1111/j.1749-6632.1993.tb17137.x) [DOI] [PubMed] [Google Scholar]
- 133. Liu D, Yang Y, Kuang F, Qing S, Hu B, Yu X. Risk of infection with different immunosuppressive drugs combined with glucocorticoids for the treatment of idiopathic membranous nephropathy: A pairwise and network meta-analysis. Int Immunopharmacol. 2019;70:354 361. ( 10.1016/j.intimp.2019.03.002) [DOI] [PubMed] [Google Scholar]
- 134. Singh JA, Hossain A, Kotb A, Wells G. Risk of serious infections with immunosuppressive drugs and glucocorticoids for lupus nephritis: a systematic review and network meta-analysis. BMC Med. 2016;14(1):137. ( 10.1186/s12916-016-0673-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Rovin BH, Teng YKO, Ginzler EM, 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. 2021;397(10289):2070 2080. ( 10.1016/S0140-6736(21)00578-X) [DOI] [PubMed] [Google Scholar]
- 136. Saxena A, Teng YKO, Collins C, England N, Leher H. Voclosporin for Lupis Nephritis: results of the two-year Aurora 2 continuation study. Ann Rheum Dis. 2022;81:325. [Google Scholar]
- 137. Ginzler EM, Wax S, Rajeswaran A, et al. Atacicept in combination with MMF and corticosteroids in lupus nephritis: results of a prematurely terminated trial. Arthritis Res Ther. 2012;14(1):R33. ( 10.1186/ar3738) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Isenberg D, Gordon C, Licu D, Copt S, Rossi CP, Wofsy D. Efficacy and safety of atacicept for prevention of flares in patients with moderate-to-severe systemic lupus erythematosus (SLE): 52-week data (APRIL-SLE randomised trial). Ann Rheum Dis. 2015;74(11):2006 2015. ( 10.1136/annrheumdis-2013-205067) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Wallace DJ, Isenberg DA, Morand EF, et al. Safety and clinical activity of atacicept in the long-term extension of the phase 2b ADDRESS II study in systemic lupus erythematosus. Rheumatol (Oxf Engl). 2021;60(11):5379 5389. ( 10.1093/rheumatology/keab115) [DOI] [PubMed] [Google Scholar]
- 140. Merrill JT, Burgos-Vargas R, Westhovens R, et al. The efficacy and safety of abatacept in patients with non-life-threatening manifestations of systemic lupus erythematosus: results of a twelve-month, multicenter, exploratory, phase IIb, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2010;62(10):3077 3087. ( 10.1002/art.27601) [DOI] [PubMed] [Google Scholar]
- 141. Furie R, Nicholls K, Cheng TT, et al. Efficacy and safety of abatacept in lupus nephritis: A twelve-month, randomized, double-blind study. Arthritis Rheumatol. 2014;66(2):379 389. ( 10.1002/art.38260) [DOI] [PubMed] [Google Scholar]
- 142. van Vollenhoven RF, Hahn BH, Tsokos GC, et al. Efficacy and safety of ustekinumab, an IL-12 and IL-23 inhibitor, in patients with active systemic lupus erythematosus: results of a multicentre, double-blind, phase 2, randomised, controlled study. Lancet. 2018;392(10155):1330 1339. ( 10.1016/S0140-6736(18)32167-6) [DOI] [PubMed] [Google Scholar]
- 143. Vollenhoven van RF, Kalunian KC, Dörner T, et al. Phase 3, multicentre, randomised, placebo-controlled study evaluating the efficacy and safety of ustekinumab in patients with systemic lupus erythematosus. Ann Rheum Dis. 2022;81(11):1556 1563. ( 10.1136/ard-2022-222858) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Wallace DJ, Furie RA, Tanaka Y, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2018;392(10143):222 231. ( 10.1016/S0140-6736(18)31363-1) [DOI] [PubMed] [Google Scholar]
- 145. Merrill JT, Werth VP, Furie R, et al. Phase 2 trial of Iberdomide in systemic lupus erythematosus. N Engl J Med. 2022;386(11):1034 1045. ( 10.1056/NEJMoa2106535) [DOI] [PubMed] [Google Scholar]
- 146. Morand E, Pike M, Merrill JT, et al. Deucravacitinib, a tyrosine kinase 2 inhibitor, in systemic lupus erythematosus: A Phase II, randomized, double-blind, placebo -controlled trial. Arthritis Rheumatol. 2023;75 (2):242 252. ( 10.1136/annrheumdis-2020-218272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Cunningham AL, Lal H, Kovac M, et al. Efficacy of the herpes zoster subunit vaccine in adults 70 years of age or older. N Engl J Med. 2016;375(11):1019 1032. ( 10.1056/NEJMoa1603800) [DOI] [PubMed] [Google Scholar]
- 148. Fanouriakis A, Tziolos N, Bertsias G, Boumpas DT. Update οn the diagnosis and management of systemic lupus erythematosus. Ann Rheum Dis. 2021;80(1):14 25. ( 10.1136/annrheumdis-2020-218272) [DOI] [PubMed] [Google Scholar]