Next Article in Journal
Prevalence of Irritable Bowel Syndrome in Ankylosing Spondylitis and Its Association with Clinical and Demographic Findings and Gut Pathology
Previous Article in Journal
Multi-Scale Analysis of Lyme Disease Ecology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

In the Pursuit of Precision: Novel Target Therapies Revolutionizing SLE Care

by
Tsvetelina Velikova
1,*,
Dimitrina Miteva
1,2,
Maria Kokudeva
3,
Georgi H. Vasilev
1,4,
Simeon Monov
5 and
Russka Shumnalieva
1,5
1
Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
2
Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
3
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medical University of Sofia, ul. Dunav 2, 1000 Sofia, Bulgaria
4
Laboratory of Hematopathology and Immunology, National Specialized Hospital for Active Treatment of Hematological Diseases, 6 Plovdivsko pole Str., 1756 Sofia, Bulgaria
5
Clinic of Rheumatology, Department of Rheumatology, Faculty of Medicine, Medical University-Sofia, 13 Urvich Str., 1612 Sofia, Bulgaria
*
Author to whom correspondence should be addressed.
Rheumato 2024, 4(3), 120-136; https://doi.org/10.3390/rheumato4030009
Submission received: 14 March 2024 / Revised: 17 June 2024 / Accepted: 24 June 2024 / Published: 29 June 2024

Abstract

:
Systemic lupus erythematosus (SLE) is a chronic, autoimmune, immune complex-mediated disease affecting mainly females at a young age. The disease etiology is still unknown, and different genetic and epigenetic factors related to disease onset and manifestations are being explored. The standard treatment regimen for SLE includes the long-term use of corticosteroids and non-specific immunosuppressive agents, often limited by co-morbidities or related side effects. However, recent advances in disease pathogenesis clarifying the role of inflammatory cytokines, chemokines, immune cells, and co-stimulation molecules have made a more practical, targeted approach possible, leading to personalized treatment strategies. This review summarizes current knowledge about SLE-targeted therapies in clinical practice.

1. Introduction

Systemic lupus erythematosus (SLE) is an autoimmune condition involving multiple organ systems and affecting predominantly women of childbearing age [1,2], with African Americans commonly affected [3]. As per the latest European Alliance of Associations for Rheumatology (EULAR) 2023 update [4], the current standard treatment of SLE includes nonsteroidal anti-inflammatory drugs (NSAIDs) for symptomatic relief and the antimalarial drug hydroxychloroquine (Plaquenil), which is recommended for all patients and corticosteroids (CS). Additional options are immunosuppressants and biologics that provide precise, targeted therapy, which opens doors for new treatment options [5,6]. However, the trend in terms of SLE management has turned progressively towards the research and development of novel targeted therapies, explicitly expanding their availability in the process [7,8,9]. Additionally, there is a current need for a holistic approach to SLE treatment that combines lifestyle modifications (e.g., smoking cessation, exercise) [10,11,12] with expanded therapeutic options, where new targeted therapies come in handy, providing wide-ranging opportunities to accomplish optimal disease outcomes. Since SLE is characterized by its heterogeneity [13], no single treatment plan can always apply to all patients. This issue renders the need for more individualized and novel targeted therapies that can address this concern precisely. For example, the tendency in recent years has been to aim for a weaning off CS [14] and other immunosuppressive drugs for long-term usage in SLE [15,16]. Therefore, biologic agents are an excellent alternative to control the disease progression and reduce flares effectively [17], while having fewer adverse effects than immunosuppressants [18]. This brings hope to the currently difficult management of SLE when it comes to accomplishing the treat-to-target strategy [19,20], which ultimately assures adequate disease remission. Moreover, targeted therapies have shown promising results in handling one of SLE’s most serious manifestations: lupus nephritis (LN) [21,22].
Furthermore, the efficacy of various SLE medicines has been observed to vary between ancestries, ethnicity, and genetic background, which may influence disease development and course and shape therapeutic responses. However, it is unclear if these differences are pharmacogenetic [23,24]. Moreover, a phenotypic and genomics approach is preferable in a multi-ethnic cohort to subtype SLE [25].
Still, there is a need for more precise and personalized therapy for SLE patients. This comprehensive review provides a concise overview of the latest advancements in SLE management, focusing on targeted therapies in clinical settings, including B-cell therapies, anti-cytokines, small molecules, co-stimulatory therapies, and CAR T cell therapies.

2. SLE Overview: Etiology and Pathogenesis as a Basis for Novel Tailored Therapy

SLE is a chronic, autoimmune, immune complex-mediated disease affecting mainly females at a young age. The etiology is still unknown, with genetic and epigenetic factors contributing to disease pathogenesis and clinical presentations [26]. The disease is characterized by the production of autoantibodies against nuclear antigens, immune complex depositions in various tissues, and subsequent complement activation leading to cell migration, tissue damage, and organ failure.
The exact cause of SLE remains unknown; however, various genetic, epigenetic, environmental, and hormonal factors are believed to contribute to its development [27]. Although genetic factors play a significant role, they are not solely responsible for SLE onset and need additional environmental triggers for the disease to manifest [28]. Generally speaking, many autoimmune diseases arise from the intricate interaction between genetic susceptibility and environmental risk factors. Genetic factors that contribute to the pathogenesis of SLE are being studied thoroughly, and new ones are being identified [29]. Additionally, hormonal factors may contribute to the development of SLE. Conditions characterized by elevated estrogen levels, such as the use of oral contraceptives, hormone replacement therapy in postmenopausal women, and endometriosis, are linked to an increased risk of SLE [30]. It is important to note that drugs such as procainamide or hydralazine can cause drug-induced lupus erythematosus [31].
The interplay between genetic susceptibility and environmental exposures can lead to epigenetic changes that dysregulate immune responses and contribute to the development and progression of SLE [32,33]. Many immunological factors concerning SLE pathogenesis, such as TNFa, IFN, IL-17, and other cytokines, have been investigated. Furthermore, meta-analyses show that gene polymorphism, such as that in the TNF-α gene, could be associated with SLE predisposition [34,35]. However, TNF-α is a proinflammatory cytokine that plays a role in the pathogenesis of various autoimmune disorders and is not limited to SLE. Similarly, interferon (INF) type I is central to SLE development and progression [36,37,38,39]. By activating dendritic and B-cells, type I INF facilitates immune dysregulation, which is a significant propelling element in the pathogenesis of SLE [40].
Furthermore, INF is also responsible for heightened inflammation and tissue damage in SLE as well as the amplification of the immune response by enhancing the expression of major histocompatibility complex (MHC) molecules on antigen-presenting cells and promoting the activation and differentiation of T cells, which further exacerbate the ongoing inflammation and tissue damage [40]. The deficiency in classical complement proteins (C1q, C4, C2) also leads to macrophages’ inability to phagocytose immune complexes and apoptotic cell material, such as plasma and nuclear antigens. This failure results in dysregulated and intolerant lymphocytes targeting intracellular antigens, usually hidden. This ultimately leads to the production of autoantibodies, such as antinuclear antibodies (ANAs) and anti-double-stranded DNA (anti-dsDNA) antibodies as well as anti-Smith, anti-Ro/SSA, and anti-La/SSB antibodies [41].
Lastly, type III hypersensitivity reactions are common in SLE, involving the formation of antibody–antigen complexes in the microvasculature. This leads to complement activation and inflammation, damaging various organs [42]. In contrast, type II hypersensitivity reactions involve IgG and IgM antibodies targeting antigens on cells, leading to different cytopenias, like anemia [43].
All advancements in understanding the pathogenesis of SLE have paved the way for developing novel targeted therapies designed to manage autoimmune responses effectively. In Figure 1, we present an overview of the factors related to SLE pathogenesis.

3. Treatment Options for SLE

Compared to other rheumatic disease treatments, SLE is still lagging in replacing traditional therapeutic approaches with novel venues. Traditional treatment agents include CS, antimalarials (hydroxychloroquine), and conventional immunomodulating/ immunosuppressive agents (azathioprine, methotrexate, cyclophosphamide, cyclosporine A, mycophenolate) [44]. Recently, the “treat-to-target” treatment strategy initially developed for rheumatoid arthritis (RA) management was conceptually extended and applied to SLE patient management [45]. Adhering to the latest EULAR and ACR recommendations [4] for the application of conventional disease-modifying drugs (csDMARDs), hydroxychloroquine could be used for all SLE patients at a target dosage regimen of 5 mg/kg body weight unless clear contraindication is present. However, this target regimen could be individualized upon considering the risk of flare or higher risk factors (i.e., kidney disease, preexisting macular or retinal disease, tamoxifen) predisposing patients to retinal toxicity. Due to its proven efficacy in reducing disease flares, safety, and low cost, hydroxychloroquine resembles the cornerstone of SLE therapy. It is eligible for long-term usage in combination with other therapeutic regimens [46,47].
Regarding CS usage, the latest recommendations set the maximal acceptable threshold to 5 mg/day prednisone equivalent for maintenance therapy in comparison to 7.5 mg/day in the 2019 recommendations. Furthermore, complete CS withdrawal should be considered as the optimal target when possible, which conceptually reorientates CS usage for SLE as a “bridging therapy” tool [4].
A lack of therapeutic response to hydroxychloroquine as a single-line therapy or in combination with CS or the inability to reduce the dosage regimen below the maintenance dosage often determines a need to add an immunosuppressive agent as a second-line therapy for mild non-renal cases of SLE. In those cases, methotrexate, azathioprine, or mycophenolate can be added. Moderate or severe cases of non-renal SLE and LN, on the other hand, could require more complex treatment strategies involving the usage of cyclophosphamide (a low dose often combined with belimumab for LN or a high dose), a calcineurin inhibitor (often combined with mycophenolate), and target therapies, such as belimumab, anifrolumab (for non-renal SLE), and rituximab (for severe or refractory patients) [4].
However, conventional immunosuppressive agents have adverse effects and limitations, such as a broad spectrum of target organ injuries or disease relapses after the discontinuation of therapy. Since long-term disease remission should be achieved in therapeutic approaches that target the establishment of self-tolerance to SLE-associated autoantigens, many SLE patients seem to be trapped in a vicious cycle of disease exacerbation followed by immunosuppressive intervention. Thus, it was shown that long-standing disease remission could not be achieved using broad immunosuppressive agents alone [48]. Therefore, the need for novel SLE-tailored therapeutic strategies centered around fundamental SLE dysregulation mechanisms comes even more to the fore.

3.1. B Cell-Directed Therapy for SLE

B cell-directed therapy for SLE has been investigated in multiple studies, systematic reviews, and meta-analyses that have contributed to the understanding of the efficacy and safety of this form of treatment [49]. Representatives of B cell-directed therapy include rituximab, ocrelizumab, and ublituximab, monoclonal antibodies that target CD20 antigens on the surface of B cells [50], and belimumab, which targets B lymphocyte stimulator-BlyS.
Unfavorably, the LUNAR study (2012) found that rituximab therapy did not enhance clinical outcomes after one year in individuals with active proliferative LN who were also treated with mycophenolate mofetil and CS [51]. The study demonstrated that although rituximab reduced CD19 + B cells and anti-dsDNA antibody levels while increasing serum levels of complement factors (C3 and C4), renal response rates were comparable across the treatment and placebo groups (45.8% and 56.9%, respectively). Furthermore, patients receiving rituximab had a higher incidence of side events such as hypotension and neutropenia [51].
However, a metanalysis of Wu et al. (2020) that included two randomized controlled trials (RCTs) and 13 observational studies, with a total of 742 patients, demonstrated notably decreased peripheral CD19 + B cells and increased C3 and C4, resulting in a net change in disease activity score and confirming rituximab effectiveness and safety for SLE [52].
The results of the latter meta-analysis overlap with the outcomes from another systematic review conducted by Cobo-Ibáñez et al. 10 years ago, which demonstrated rituximab safety and effectiveness in treating non-renal SLE, particularly when it comes to disease activity, immunologic markers, and the sparing of CS. However, the authors concluded that rituximab could only be advised for organ-specific symptoms like thrombocytopenia and arthritis [53].
On the contrary, a systematic review by Weidenbusch et al. (2013) indicated that in SLE with LN patients who have not obtained remission from traditional therapy, rituximab is an effective way to induce remission [54]. On the other hand, Borba et al. demonstrated that despite its good safety profile, rituximab showed no superiority over a placebo in terms of efficacy in SLE [55].
Belimumab is currently the only B-cell-directed therapy for SLE that the FDA and EMA have approved. Belimumab is a monoclonal antibody that blocks B lymphocyte stimulator-BLyS, essential for activating B cell function [49]. BLISS-52 and BLISS-76 are two phase III studies investigating belimumab’s effectiveness (a biologic agent that targets B cells) in SLE. The results showed that the administration of belimumab, in addition to standard therapy, led to improvements in inflammatory indices and clinical symptoms of SLE in some subgroups of patients [49,56].
Another phase III trial focused on the long-term safety and efficacy of belimumab in SLE. The results showed improved inflammatory parameters and reduced SLE activity in patients receiving belimumab [57]. In their meta-analysis, Wei et al. (2016) estimated the efficacy and safety of therapy with belimumab compared with a placebo plus standard therapy in patients with SLE. The results suggested that treatment with belimumab plus standard therapy is more effective than a placebo plus standard therapy in SLE patients, representing major progress in the treatment of SLE. Regardless of the statistical analyses, further research is necessary to optimize treatment effects [58].
Similarly, in their systematic review and meta-analysis, Borba et al. (2014) evaluated the efficacy, safety, and tolerability of biologic drugs compared with a placebo for SLE. Belimumab exhibited a satisfactory profile regarding efficacy, safety, and tolerability. Biologic agents exhibited a good safety profile for SLE treatment, indicating that these agents are promising therapies and should be further investigated [55]. Regarding adverse reactions and loss of efficacy, despite the positive results of these studies and meta-analyses, B cell-directed therapy may be associated with various adverse reactions and loss of effectiveness. Adverse reactions related to the use of belimumab include nausea, diarrhea, pyrexia, nasopharyngitis, bronchitis, insomnia, limb pain, depression, migraine, pharyngitis, cystitis, leukopenia, viral gastroenteritis, serious infections, depression-related events, suicidal ideation, fatal infections, and serious psychiatric events [59].
In conclusion, B cell-directed therapy aims to regulate or reduce B cell activity through various therapeutic strategies, such as monoclonal antibodies, selective inhibitors, and other drugs that target specific molecules or signaling pathways on B cells. Furthermore, this therapy controls unwanted immune responses associated with SLE and RA.

3.2. Co-Stimulatory Molecules for SLE

Co-stimulatory molecules are essential signaling molecules that activate the immune system and control the immune response. They play a crucial role in regulating the activity of T cells and other cellular elements of the immune system [60]. No medications are approved explicitly as co-stimulatory molecules for treating SLE yet. However, considering the pathogenesis of SLE, some of the most essential co-stimulatory molecules include the following. CD28 is a crucial molecule for T-cell activation. It binds to B7 molecules (CD80 and CD86) that are found on the surface of other cells, such as antigen-presenting cells (APCs) like macrophages or dendritic cells [61].
Similarly, CTLA-4 (cytologic T lymphocyte antigen 4) is an inhibitory receptor expressed on the surface of activated T cells. It competes with CD28 to bind to B7 molecules (CD80 and CD86), but with higher affinity, inhibiting T-cell activation [62]. Another molecule is ICOS, a co-stimulatory receptor that is expressed on activated T cells. It regulates the immune response by binding to ICOS-L (ICOS-Ligand), found on the surface of other cells, such as B cells and dendritic cells [63]. CD40 and CD40L are molecules expressed on the surface of B cells (CD40) and activated T cells (CD40L). They are involved in the stimulation and differentiation of B cells and the immune response [64].
For the therapy of SLE, various co-stimulatory molecules are being investigated as potential targets for new drugs. Since CD40 is a co-stimulatory molecule that plays a vital role in activating the immune system, studies suggest that the inhibition of the interaction between CD40 and its ligand (CD40L) may benefit the inflammatory characteristic of SLE [65].
Dapirolizumab pegol is composed of an anti-CD40 ligand Fab’antibody fragment connected to polyethylene glycol, included in phase IIb clinical trials that demonstrated clinical improvement, declined anti-DNA antibodies, and normalization of C3 and C4 [66].
Additionally, dapirolizumab pegol showed a good safety profile with a low risk of thromboembolic events [67]. Ramanujam et al. also rediscovered the role of the CD40-CD40L pathway in SLE and LN, emphasizing the blocking of this pathway to ameliorate systemic inflammation and nephritis [68,69]. Furthermore, the role of CD40-CD40L was confirmed in murine SLE models and cultures of cells in SLE patients [70,71].
Given the fact that B7 (B7-1 and B7-2) molecules interact with T cell receptors and play a vital role in regulating the immune response, modulating this interaction may be a potential strategy for treating SLE [72]. Similarly, a ligand for 4-1BB (CD137) is a co-stimulatory molecule expressed on activated T cells. Some studies have shown that blocking or stimulating 4-1BB may have anti-inflammatory or immunomodulatory effects that may be useful in controlling SLE [73].

3.3. Targeting Intracellular Signaling via JAK Inhibitors in SLE

The discovery of targeted small-molecule therapies, including Janus kinase (JAK) inhibitors, changed the therapy paradigm for some disorders. Through suppressing intracellular signaling (i.e., JAK protein–signal transducer and activator of transcription (STAT) transcription factors pathways), JAK inhibitors exert the potential for effectiveness in many autoimmune diseases, including RA, inflammatory bowel disease, spondylarthritis, alopecia areata, and some allergic diseases [74].
Through JAK-STAT, cytokines from the innate immune system communicate with adaptive immunological systems, such as autoreactive T cells, B cell activation, and autoantibody formation. These processes may be used as therapeutic targets for SLE [74,75]. Furthermore, convenient administration, reduced production costs, and non-immunogenicity benefit these targeted molecules [76,77].
In line with this, Nikolopoulos et Parodis focused on the clinical application of JAK inhibitors in SLE [78]. Data showed that in individuals with SLE, tofacitinib, a JAK1/3 inhibitor, decreased the type I interferon signature, improved vascular function, and lowered cholesterol levels [79]. Baricitinib, a JAK1/2 inhibitor, has been tested in one phase 2 trial, which showed notable improvements in lupus rashes and arthritis, and two phase 3 trials, which did not confirm these findings [80,81,82].
More extensive phase 3 trials will examine deucravacitinib, a selective tyrosine kinase 2 (TYK2) inhibitor, which produced higher response rates than a placebo in a phase 2 trial in SLE [83]. However, using JAK inhibitors necessitates carefully considering their multi-target effects, including sufficient screening and regular monitoring for infection, cardiovascular problems, thrombosis, and cancer [84,85,86].
A systematic review and meta-analysis on the efficacy and safety of JAK inhibitors in SLE and cutaneous lupus by Ma et al. highlighted that although JAK inhibitors were superior to a placebo in managing SLE and treating musculoskeletal and mucocutaneous involvement, they are not approved for SLE yet [87].
Therefore, why do we need JAK inhibitors in SLE? Richter et al. [88] answered this question by pointing out that none of these treatments has been approved for clinical practice despite encouraging outcomes from clinical trials. So far, using tofacitinib and baricitinib has shown the best outcomes. Owing to its intricate etiology, the treatment of SLE is evolving. Although JAK inhibitors are therapeutically helpful, how they relate to SLE is unclear. Although the safety and efficacy profiles indicate promising outcomes, more clinical trials are required. However, all the data released thus far are a foundation for upcoming research that should yield new and helpful insights into treating SLE patients [88].

3.4. Targeting Cytokines and Interferon and Kinoid Vaccines in SLE

Because cytokine production is different in SLE patients and may change during the disease [89], interferons and some inflammatory cytokines have been investigated for therapeutic purposes. A recent systematic review showed that more than 15 cytokine-targeting molecules had been discovered for the targeted therapy of SLE in clinical practice [20]. Here, we will focus on the most promising targeted therapies currently in clinical development.

3.4.1. IL-2

Interleukin-2 (IL-2) and regulatory T cells (Tregs) are essential in promoting and maintaining immune tolerance. In patients with SLE, there are low levels of this interleukin, which leads to defects in Treg cells [90]. With low-dose IL-2 therapy, this deficiency can be reversed and restore immune tolerance [91,92,93]. In an open-label phase I/II trial with SLE patients, more than 80% showed a significant immune response and increased Treg cells [94]. Although this trial is in its initial phases, it has demonstrated that low dosages of IL-2 can improve Treg abnormalities linked to IL-2 deficiency in SLE patients. Furthermore, low-dose IL-2 therapy restores endogenous immunological tolerance mechanisms, thereby proposing a targeted biological treatment method that directly targets the pathophysiology of SLE [94].
The role of IL-2 has been evaluated in two RCTs with a good safety profile and clinical response [95,96]. He et al. demonstrated declined SLE Responder Index-4 in an IL-2-treated group vs. a placebo at week 12 (p = 0.052) and week 24 (p = 0.027). Furthermore, low-dose IL-2 treatment resulted in remission in 53.8% of patients with LN. No severe infections were recorded in the IL-2 group and two were recorded in the placebo group [95]. Similarly, Humrich et al. conducted a post-hoc hierarchical analysis of the primary and critical secondary endpoints in a per-protocol population, together with an exploratory analysis of many other secondary endpoints. The outcomes supported that low-dose IL-2 is advantageous in active SLE [96,97].
Placebo-controlled trials of low-dose IL-2 are Aldesleukin (NCT03312335), Efavaleukin Alfa (NCT03451422), LUPIL-2 trial with ILT-101 (NCT02955615), IL-2 with Telitacicept (NCT05339217), and CNTY-101 (NCT06255028) [98].

3.4.2. IL-6

Interleukin 6 (IL-6) is a cytokine with pleiotropic effects and has been found to be elevated in SLE patients [99,100,101,102,103]. PF-04236921, a human monoclonal IL-6 antibody, was investigated in SLE clinical trials but failed to demonstrate efficacy [104,105]. Another promising antibody, sirukumab, failed in the phase II trial [106]. Tocilizumab, a human monoclonal against the IL-6 receptor (IL-6R), was evaluated in an open-label phase I study [107]. The monoclonal antibodies MRA003US (NCT00046774) and CNTO 136 (NCT01702740) have no published results.

3.4.3. IL-17

Interleukin 17 (IL-17) is involved in the pathogenesis of rheumatic and musculoskeletal diseases. In SLE patients, increased Th17 cells and high levels of IL-17 in serum have been observed [108]. A human monoclonal antibody, secukinumab, was in a phase III clinical trial [109]. However, the study was terminated due to a futility analysis. There is one more trial in the database for IL-17, which is from 2021, but it has an unknown status (NCT05045417). The outcomes for SLE have been unfavorable so far; however, case reports have demonstrated the efficacy of secukinumab for LN refractory treatment, as reported by Costa et al. [110]. Nevertheless, other authors reported secukinumab-induced SLE in psoriatic arthritis [111] and ankylosing spondylitis [112].

3.4.4. IL-12/23 Axis

In patients with active SLE, there are increased serum levels of IL-23 compared to healthy individuals, which limits the in vitro production of IL-2 [93]. IL-12 is also involved in the pathophysiology of SLE [113]. Activation of the IL-23/IL-17A axis leads to an increased amount of Th17 cells, which ultimately contributes to the pathogenesis of LN [114,115]. In connection with these data, a monoclonal anti-IL-12/23 antibody, ustekinumab, targeting the p40 subunit shared by the cytokines IL-12 and IL-23, has been reported [116]. This is an up-and-coming option for the terminated phase III trial, and safety and adverse effects are similar to those reported by other studies.

3.4.5. IL-10

Interleukin 10 (IL-10) has anti-inflammatory and proinflammatory effects, which are pathogenic in SLE [117]. A study shows that IL-10 serum levels are increased in SLE patients. The correlation with disease status is robust [118]. The anti-IL-10 antibody has completed a phase II trial (NCT02554019) and has results [119].
Hannon et al. included sixty-one RCTs involving 11,232 participants and reported 43 different interventions for cutaneous disease in SLE, including the trial mentioned above in their analysis. They concluded that evidence supports the commonly used treatment hydroxychloroquine and the combination of chloroquine and methotrexate. However, further data analyses and more trials designed to detect differences in the efficacy of anti-cytokines are needed [120].

3.4.6. Interferons

Interferon is a key cytokine in the pathogenesis of SLE, which is supported by numerous animal and human findings [121,122,123,124]. SLE patients have increased levels of type I IFN, and the genes are highly expressed in the peripheral blood [125,126,127]. Given the potential of IFNs to initiate or enhance immune responses leading to organ damage in lupus, many biologic therapies targeting IFNs are under investigation. Various strategies to develop interferon-targeted treatment have been tried over the years but have been largely unsuccessful. Subsequently, several molecules have been identified that target the IFN pathways and the IFNAR receptor, which signals intracellularly through JAK1/Tyk2 [128,129,130].
Anifrolumab (MEDI-546) is a monoclonal antibody against IFNAR1. After a successful phase, two open-label trials (MUSE) were approved for fast-track status by the FDA in 2021 [131,132,133]. Two other phase 3 studies were conducted in active SLE (TULIP-1 and TULIP-2) [134,135]. The first trial reported a negative primary endpoint (SRI-4) [134] and TULIP-2 reported positive results [135]. The discrepancies between the two trials are related to the selection of SRI-4 and a subgroup of lower disease activity SLE [136]. The use of anifrolumab in LN was investigated in the phase 2 TULIP-LN study [137]. A phase 3 trial (IRIS, NCT05138133) has started and is expected to end in 2027. It was shown that only phase 4 studies can accurately assess the safety of new SLE drugs.
Since anifrolumab was approved for moderate and severe SLE treatment, the post-hoc analysis confirmed its better efficacy in patients with a high type I IFN signature than in patients with a low type I IFN signature [138]. At the moment, anifrolumab is indicated for SLE without renal or CNS involvement.
Another anti-IFNα monoclonal antibody, sifalimumab, has been studied in a phase II trial in patients with moderate to severe SLE [133]. The most common complication reported was herpes zoster in patients receiving the high dose. The study reported positive results, although the effect was relatively modest. Rontalizumab is another human anti-IFNα antibody and was evaluated in a placebo-controlled phase II trial [139]. The severe side effects were relatively acceptable and classified as unrelated to the drug. The higher doses would be more effective, but the trials were stopped.
It was shown that IFNα kinoid, i.e., IFNα immunogen appropriately adjuvanted, produced short-lived neutralizing antibodies (Abs) but had no discernible adverse effects and no cellular immunological response to the cytokine in a murine model. Clinically, kinoid-vaccinated NZB/W mice showed reduced lupus manifestations, including proteinuria, histological renal lesions, and death triggered by IFNα adjuvant challenge [140].
A phase IIb, randomized, double-blind, placebo-controlled study was conducted for IFNα-kinoid (NCT02665364) in patients with active SLE [127,141]. IFN-K had a significant corticoid-sparing effect, but the evidence was limited and of moderate quality. Despite all the potential improvements in the understanding of the pathogenesis of SLE, the need for accurate treatment is still high. The development of new therapeutic agents in SLE will allow for the discontinuation of CS and the achievement of remission or low-disease activity and remission.

3.5. CAR T Cell Therapy in SLE

A revolutionary new approach for SLE treatment is using genetically modified treatments with chimeric antigen receptor (CAR) T-cells designed to specifically target other cells or molecules with a main pathogenic role for disease onset and progression [142]. CARs are recombinant receptors coupled to autologous T cells to target a specific antigen. Targeting the CD19 antigen in SLE aims to induce a rapid and prolonged depletion of the circulating B cells in patients who failed to achieve clinical and/or serological remission on standard treatment protocols [143,144].
CAR Tcell therapy was first introduced by Schett et al. in a patient with refractory-to-standard treatment SLE, which achieved clinical drug-free remission for the whole follow-up period [145,146]. The authors recently reported a case series with 15 patients with refractory autoimmune connective tissue diseases (eight with SLE, three with idiopathic inflammatory Myopathies —IIM, and four with systemic sclerosis–SSc patients) who were treated with CAR T cells to achieve clinical and laboratory drug-free remission [147]. Research on CAR T cell therapy in refractory or relapsing autoimmune diseases, such as SLE, is progressing rapidly to improve treatment strategies and clinical outcomes in this kind of patient. Currently, several clinical trials are exploring the safety and efficacy of CAR T cell therapy in SLE or LN [148].
All of the novel targeted therapies in SLE discussed in this review are presented in Table 1.

4. Conclusions

The evolving landscape of SLE management underscores the need for precise and targeted therapies. While CS and immunosuppressive agents have long been the mainstay, their limitations prompt exploration into novel approaches. Recent elucidations of SLE pathogenesis, particularly the genetics, epigenetics, and involvement of inflammatory mediators and immune dysregulation, have paved the way for personalized treatment modalities. By targeting specific molecular pathways, such as inflammatory cytokines and co-stimulation molecules, tailored interventions offer promising avenues for improved outcomes. Based on their efficacy and safety in the clinical trials, we can speculate that the subsequent approved treatments will be JAK inhibitors and members of CAR T cell therapy.

Author Contributions

Conceptualization, T.V. and R.S.; methodology, M.K.; software, G.H.V.; validation, R.S. and T.V.; formal analysis, D.M.; investigation, D.M.; resources, M.K. and S.M.; data curation, G.H.V. and S.M.; writing—original draft preparation, T.V., D.M., M.K., G.H.V. and R.S.; writing—review and editing, S.M. and T.V.; visualization, R.S.; supervision, T.V.; project administration, T.V.; funding acquisition, T.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financed by the European Union project NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project no. BG-RRP-2.004-0008.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tsokos, G.C. Systemic lupus erythematosus. N. Engl. J. Med. 2011, 365, 2110–2121. [Google Scholar] [CrossRef] [PubMed]
  2. Durcan, L.; O’Dwyer, T.; Petri, M. Management strategies and future directions for systemic lupus erythematosus in adults. Lancet 2019, 393, 2332–2343. [Google Scholar] [CrossRef]
  3. Kiriakidou, M.; Ching, C.L. Systemic Lupus Erythematosus. Ann. Intern. Med. 2020, 172, ITC81–ITC96. [Google Scholar] [CrossRef]
  4. Fanouriakis, A.; Kostopoulou, M.; Andersen, J.; Aringer, M.; Arnaud, L.; Bae, S.-C.; Boletis, J.; Bruce, I.N.; Cervera, R.; Doria, A.; et al. EULAR recommendations for the management of systemic lupus erythematosus: 2023 update. Ann. Rheum. Dis. 2024, 83, 15–29. [Google Scholar] [CrossRef] [PubMed]
  5. Lazar, S.; Kahlenberg, J.M. Systemic lupus erythematosus: New diagnostic and therapeutic approaches. Annu. Rev. Med. 2023, 74, 339–352. [Google Scholar] [CrossRef]
  6. Katarzyna, P.-B.; Wiktor, S.; Ewa, D.; Piotr, L. Current treatment of systemic lupus erythematosus: A clinician’s perspective. Rheumatol. Int. 2023, 43, 1395–1407. [Google Scholar] [CrossRef]
  7. Wang, D.H.; Wallace, D.J.M. New Insights Into Systemic Lupus Erythematosus Therapies: 2010–2020. JCR J. Clin. Rheumatol. 2022, 28, e217–e221. [Google Scholar] [CrossRef]
  8. Jordan, N.; Lutalo, P.M.; D’cruz, D.P. Novel therapeutic agents in clinical development for systemic lupus erythematosus. BMC Med. 2013, 11, 120. [Google Scholar] [CrossRef]
  9. Fanouriakis, A.; Tziolos, N.; Bertsias, G.; Boumpas, D.T. Update οn the diagnosis and management of systemic lupus erythematosus. Ann. Rheum. Dis. 2021, 80, 14–25. [Google Scholar] [CrossRef]
  10. The Lancet Rheumatology. Embracing holistic management in SLE. Lancet Rheumatol. 2023, 5, e639. [Google Scholar] [CrossRef]
  11. Parodis, I.; Gomez, A.; Tsoi, A.; Chow, J.W.; Pezzella, D.; Girard, C.; Stamm, T.A.; Boström, C. Systematic literature review informing the EULAR recommendations for the non-pharmacological management of systemic lupus erythematosus and systemic sclerosis. RMD Open 2023, 9, e003297. [Google Scholar] [CrossRef]
  12. Parodis, I.; Girard-Guyonvarc’h, C.; Arnaud, L.; Distler, O.; Domján, A.; Ende, C.H.M.V.D.; Fligelstone, K.; Kocher, A.; Larosa, M.; Lau, M.; et al. EULAR recommendations for the non-pharmacological management of systemic lupus erythematosus and systemic sclerosis. Ann. Rheum. Dis. 2023, 83, 720–729. [Google Scholar] [CrossRef] [PubMed]
  13. Dörner, T.; Furie, R. Novel paradigms in systemic lupus erythematosus. Lancet 2019, 393, 2344–2358. [Google Scholar] [CrossRef] [PubMed]
  14. Fanouriakis, A.; Kostopoulou, M.; Alunno, A.; Aringer, M.; Bajema, I.; Boletis, J.N.; Cervera, R.; Doria, A.; Gordon, C.; Govoni, M.; et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann. Rheum. Dis. 2019, 78, 736–745. [Google Scholar] [CrossRef]
  15. Jourde-Chiche, N.; Costedoat-Chalumeau, N.; Baumstarck, K.; Loundou, A.; Bouillet, L.; Burtey, S.; Caudwell, V.; Chiche, L.; Couzi, L.; Daniel, L.; et al. Weaning of maintenance immunosuppressive therapy in lupus nephritis (WIN-Lupus): Results of a multicentre randomised controlled trial. Ann. Rheum. Dis. 2022, 81, 1420–1427. [Google Scholar] [CrossRef]
  16. Jourde-Chiche, N.; Chiche, L. An era of immunosuppressant withdrawal in systemic lupus erythematosus: Winning through weaning. Lancet Rheumatol. 2024, 6, e133–e134. [Google Scholar] [CrossRef]
  17. Tsang-A-Sjoe, M.W.P.; Bultink, I.E.M. New developments in systemic lupus erythematosus. Rheumatology 2021, 60, vi21–vi28. [Google Scholar] [CrossRef] [PubMed]
  18. Durcan, L.; Petri, M. Why targeted therapies are necessary for systemic lupus erythematosus. Lupus 2016, 25, 1070–1079. [Google Scholar] [CrossRef]
  19. Sánchez, A.R.P.; Voskuyl, A.E.; van Vollenhoven, R.F. Treat-to-target in systemic lupus erythematosus: Advancing towards its implementation. Nat. Rev. Rheumatol. 2022, 18, 146–157. [Google Scholar] [CrossRef]
  20. Felten, R.; Scherlinger, M.; Mertz, P.; Chasset, F.; Arnaud, L. New biologics and targeted therapies in systemic lupus: From new molecular targets to new indications. A systematic review. Jt. Bone Spine 2023, 90, 105523. [Google Scholar] [CrossRef]
  21. Thakare, S.B.; So, P.N.; Rodriguez, S.; Hassanein, M.; Lerma, E.; Wiegley, N.; GlomCon Editorial Team. Novel Therapeutics for Management of Lupus Nephritis: What Is Next? Kidney Med. 2023, 5, 100688. [Google Scholar] [CrossRef]
  22. Jia, X.; Lu, Y.; Zheng, X.; Tang, R.; Chen, W. Targeted therapies for lupus nephritis: Current perspectives and future directions. Chin. Med. J. 2023, 137, 34–43. [Google Scholar] [CrossRef] [PubMed]
  23. Demkova, K.; Morris, D.L.; Vyse, T.J. Genetics of SLE: Does this explain susceptibility and severity across racial groups? Rheumatology 2023, 62 (Suppl. 1), i15–i21. [Google Scholar] [CrossRef] [PubMed]
  24. González, L.A.; Toloza, S.M.; McGwin, G., Jr.; Alarcón, G.S. Ethnicity in systemic lupus erythematosus (SLE): Its influence on susceptibility and outcomes. Lupus 2013, 22, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  25. Lanata, C.M.; Paranjpe, I.; Nititham, J.; Taylor, K.E.; Gianfrancesco, M.; Paranjpe, M.; Andrews, S.; Chung, S.A.; Rhead, B.; Barcellos, L.F.; et al. A phenotypic and genomics approach in a multi-ethnic cohort to subtype systemic lupus erythematosus. Nat. Commun. 2019, 10, 3902, Erratum in Nat. Commun. 2020, 11, 1164. [Google Scholar] [CrossRef] [PubMed]
  26. Shumnalieva, R.; Kachakova, D.; Shoumnalieva-Ivanova, V.; Miteva, P.; Kaneva, R.; Monov, S. Whole peripheral blood miR-146a and miR-155 expression levels in Systemic lupus erythematosus patients. Acta Reumatol. Port. 2018, 43, 217–225. [Google Scholar] [PubMed]
  27. Kamen, D.L. Environmental influences on systemic lupus erythematosus expression. Rheum. Dis. Clin. North Am. 2014, 40, 401–412, vii. [Google Scholar] [CrossRef] [PubMed]
  28. Woo, J.M.P.; Parks, C.G.; Jacobsen, S.; Costenbader, K.H.; Bernatsky, S. The role of environmental exposures and gene-environment interactions in the etiology of systemic lupus erythematous. J. Intern. Med. 2022, 291, 755–778. [Google Scholar] [CrossRef] [PubMed]
  29. Guga, S.; Wang, Y.; Graham, D.C.; Vyse, T.J. A review of genetic risk in systemic lupus erythematosus. Expert Rev. Clin. Immunol. 2023, 19, 1247–1258. [Google Scholar] [CrossRef]
  30. Barbhaiya, M.; Costenbader, K.H. Environmental exposures and the development of systemic lupus erythematosus. Curr. Opin. Rheumatol. 2016, 28, 497–505. [Google Scholar] [CrossRef]
  31. Solhjoo, M.; Goyal, A.; Chauhan, K. Drug-Induced Lupus Erythematosus. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  32. Long, H.; Yin, H.; Wang, L.; Gershwin, M.E.; Lu, Q. The critical role of epigenetics in systemic lupus erythematosus and autoimmunity. J. Autoimmun. 2016, 74, 118–138. [Google Scholar] [CrossRef] [PubMed]
  33. Hedrich, C.M. Epigenetics in SLE. Curr. Rheumatol. Rep. 2017, 19, 58. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, Y.H.; Harley, J.B.; Nath, S.K. Meta-analysis of TNF-alpha promoter -308 A/G polymorphism and SLE susceptibility. Eur. J. Hum. Genet. 2006, 14, 364–371. [Google Scholar] [CrossRef] [PubMed]
  35. Zou, Y.-F.; Feng, X.-L.; Pan, F.-M.; Su, H.; Tao, J.-H.; Ye, D.-Q. Meta-analysis of TNF-alpha promoter—238A/G polymorphism and SLE susceptibility. Autoimmunity 2010, 43, 264–274. [Google Scholar] [CrossRef] [PubMed]
  36. Ghorbaninezhad, F.; Leone, P.; Alemohammad, H.; Najafzadeh, B.; Nourbakhsh, N.S.; Prete, M.; Malerba, E.; Saeedi, H.; Tabrizi, N.J.; Racanelli, V.; et al. Tumor necrosis factor-α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). Int. J. Mol. Med. 2022, 49, 43. [Google Scholar] [CrossRef] [PubMed]
  37. Rönnblom, L.; Leonard, D. Interferon pathway in SLE: One key to unlocking the mystery of the disease. Lupus Sci. Med. 2019, 6, e000270. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Postal, M.; Vivaldo, J.F.; Fernandez-Ruiz, R.; Paredes, J.L.; Appenzeller, S.; Niewold, T.B. Type I interferon in the pathogenesis of systemic lupus erythematosus. Curr. Opin. Immunol. 2020, 67, 87–94. [Google Scholar] [CrossRef] [PubMed]
  39. Crow, M.K. Pathogenesis of systemic lupus erythematosus: Risks, mechanisms and therapeutic targets. Ann. Rheum. Dis. 2023, 82, 999–1014. [Google Scholar] [CrossRef] [PubMed]
  40. Caielli, S.; Wan, Z.; Pascual, V. Systemic Lupus Erythematosus Pathogenesis: Interferon and Beyond. Annu. Rev. Immunol. 2023, 41, 533–560. [Google Scholar] [CrossRef]
  41. Han, S.; Zhuang, H.; Shumyak, S.; Yang, L.; Reeves, W.H. Mechanisms of autoantibody production in systemic lupus erythematosus. Front. Immunol. 2015, 6, 228. [Google Scholar] [CrossRef]
  42. Pan, L.; Lu, M.-P.; Wang, J.-H.; Xu, M.; Yang, S.-R. Immunological pathogenesis and treatment of systemic lupus erythematosus. World J. Pediatr. 2020, 16, 19–30. [Google Scholar] [CrossRef] [PubMed]
  43. Bashal, F. Hematological disorders in patients with systemic lupus erythematosus. Open Rheumatol. J. 2013, 7, 87–95. [Google Scholar] [CrossRef] [PubMed]
  44. Basta, F.; Fasola, F.; Triantafyllias, K.; Schwarting, A. Systemic Lupus Erythematosus (SLE) Therapy: The Old and the New. Rheumatol. Ther. 2020, 7, 433–446. [Google Scholar] [CrossRef] [PubMed]
  45. Zucchi, D.; Cardelli, C.; Elefante, E.; Tani, C.; Mosca, M. Treat-to-Target in Systemic Lupus Erythematosus: Reality or Pipe Dream. J. Clin. Med. 2023, 12, 3348. [Google Scholar] [CrossRef] [PubMed]
  46. Almeida-Brasil, C.C.; Hanly, J.G.; Urowitz, M.; Clarke, A.E.; Ruiz-Irastorza, G.; Gordon, C.; Ramsey-Goldman, R.; Petri, M.; Ginzler, E.M.; Wallace, D.J.; et al. Flares after hydroxychloroquine reduction or discontinuation: Results from the Systemic Lupus International Collaborating Clinics (SLICC) inception cohort. Ann. Rheum. Dis. 2022, 81, 370–378. [Google Scholar] [CrossRef] [PubMed]
  47. Shinjo, S.K.; Bonfá, E.; Wojdyla, D.; Borba, E.F.; Ramirez, L.A.; Scherbarth, H.R.; Brenol, J.C.T.; Chacón-Diaz, R.; Neira, O.J.; Berbotto, G.A.; et al. Antimalarial treatment may have a time-dependent effect on lupus survival: Data from a multinational Latin American inception cohort. Arthritis Rheum. 2010, 62, 855–862. [Google Scholar] [CrossRef]
  48. Mathias, L.M.; Stohl, W. Systemic lupus erythematosus (SLE): Emerging therapeutic targets. Expert Opin. Ther. Targets 2020, 24, 1283–1302. [Google Scholar] [CrossRef] [PubMed]
  49. Navarra, S.V.; Guzmán, R.M.; Gallacher, A.E.; Hall, S.; Levy, R.A.; Jimenez, R.E.; Li, E.K.-M.; Thomas, M.; Kim, H.-Y.; León, M.G.; et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: A randomised, placebo-controlled, phase 3 trial. Lancet 2011, 377, 721–731. [Google Scholar] [CrossRef] [PubMed]
  50. Dörner, T.; Jacobi, A.M.; Lipsky, P.E. B cells in autoimmunity. Arthritis Res. Ther. 2009, 11, 247. [Google Scholar] [CrossRef]
  51. Negative results for the LUNAR phase III study of rituximab. Nat. Rev. Rheumatol. 2012, 8, 122. [CrossRef]
  52. Wu, S.; Wang, Y.; Zhang, J.; Han, B.; Wang, B.; Gao, W.; Zhang, N.; Zhang, C.; Yan, F.; Li, Z. Efficacy and safety of rituximab for systemic lupus erythematosus treatment: A meta-analysis. Afr. Health Sci. 2020, 20, 871–884. [Google Scholar] [CrossRef]
  53. Cobo-Ibáñez, T.; Loza-Santamaría, E.; Pego-Reigosa, J.M.; Marqués, A.O.; Rúa-Figueroa, I.; Fernández-Nebro, A.; Cáliz, R.C.; Longo, F.J.L.; Muñoz-Fernández, S. Efficacy and safety of rituximab in the treatment of non-renal systemic lupus erythematosus: A systematic review. Semin. Arthritis Rheum. 2014, 44, 175–185. [Google Scholar] [CrossRef]
  54. Weidenbusch, M.; Römmele, C.; Schröttle, A.; Anders, H.-J. Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol. Dial. Transplant. 2013, 28, 106–111. [Google Scholar] [CrossRef]
  55. Borba, H.H.; Wiens, A.; de Souza, T.T.; Correr, C.J.; Pontarolo, R. Efficacy and safety of biologic therapies for systemic lupus erythematosus treatment: Systematic review and meta-analysis. BioDrugs 2014, 28, 211–228. [Google Scholar] [CrossRef] [PubMed]
  56. Furie, R.; Petri, M.; Zamani, O.; Cervera, R.; Wallace, D.J.; Tegzová, D.; Sanchez-Guerrero, J.; Schwarting, A.; Merrill, J.T.; Chatham, W.W.; et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011, 63, 3918–3930. [Google Scholar] [CrossRef] [PubMed]
  57. Furie, R.A.; Wallace, D.J.; Aranow, C.; Fettiplace, J.; Wilson, B.; Mistry, P.; Roth, D.A.; Gordon, D. Long-term safety and efficacy of belimumab in patients with systemic lupus erythematosus: A continuation of a seventy-six-week phase III parent study in the United States. Arthritis Rheumatol. 2018, 70, 868–877. [Google Scholar] [CrossRef]
  58. Wei, L.-Q.; Liang, Y.-G.; Zhao, Y.; Liang, H.-T.; Qin, D.-C.; She, M.-C. Efficacy and Safety of Belimumab Plus Standard Therapy in Patients With Systemic Lupus Erythematosus: A Meta-analysis. Clin. Ther. 2016, 38, 1134–1140. [Google Scholar] [CrossRef]
  59. Medscape, Belimumab. Available online: https://reference.medscape.com/drug/benlysta-belimumab-999632 (accessed on 4 March 2024).
  60. Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef] [PubMed]
  61. Esensten, J.H.; Helou, Y.A.; Chopra, G.; Weiss, A.; Bluestone, J.A. CD28 co-stimulation: From Mechanism to Therapy. Immunity 2016, 44, 973–988. [Google Scholar] [CrossRef]
  62. Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
  63. Gonzalo, J.A.; Tian, J.; Delaney, T.; Corcoran, J.; Rottman, J.B.; Lora, J.; Al-Garawi, A.; Kroczek, R.; Gutierrez-Ramos, J.C.; Coyle, A.J. ICOS is critical for T helper cell-mediated lung mucosal inflammatory responses. Nat. Immunol. 2001, 2, 597–604. [Google Scholar] [CrossRef] [PubMed]
  64. Banchereau, J.; Bazan, F.; Blanchard, D.; Briè, F.; Galizzi, J.P.; van Kooten, C.; Liu, Y.J.; Rousset, F.; Saeland, S. The CD40 antigen and its ligand. Annu. Rev. Immunol. 1994, 12, 881–926. [Google Scholar] [CrossRef] [PubMed]
  65. Ramanujam, M.; Wang, X.; Huang, W.; Schiffer, L.; Grimaldi, C.; Akkerman, A.; Diamond, B.; Madaio, M.P.; Davidson, A. Mechanism of action of transmembrane activator and calcium modulator ligand interactor-Ig in murine systemic lupus erythematosus. J. Immunol. 2004, 173, 3524–3534. [Google Scholar] [CrossRef] [PubMed]
  66. Furie, R.A.; Bruce, I.N.; Dörner, T.; Leon, M.G.; Leszczyński, P.; Urowitz, M.; Haier, B.; Jimenez, T.; Brittain, C.; Liu, J.; et al. Phase 2, randomized, placebo-controlled trial of dapirolizumab pegol in patients with moderate-to-severe active systemic lupus erythematosus. Rheumatology 2021, 60, 5397–5407. [Google Scholar] [CrossRef] [PubMed]
  67. Soybilgic, A. Biologic agents and other emerging therapies for childhood SLE. Pediatr. Ann. 2022, 51, E63–E71. [Google Scholar] [CrossRef] [PubMed]
  68. Ramanujam, M.; Steffgen, J.; Visvanathan, S.; Mohan, C.; Fine, J.S.; Putterman, C. Phoenix from the flames: Rediscovering the role of the CD40-CD40L pathway in systemic lupus erythematosus and lupus nephritis. Autoimmun. Rev. 2020, 19, 102668. [Google Scholar] [CrossRef]
  69. Marken, J.; Muralidharan, S.; Giltiay, N.V. Correction to: Anti-CD40 antibody KPL-404 inhibits T cell-mediated activation of B cells from healthy donors and autoimmune patients. Arthritis Res. Ther. 2021, 23, 36, Erratum in Arthritis Res. Ther. 2021, 23, 5. https://doi.org/10.1186/s13075-020-02372-z. [Google Scholar] [CrossRef] [PubMed]
  70. Perper, S.J.; Westmoreland, S.V.; Karman, J.; Twomey, R.; Seagal, J.; Wang, R.; McRae, B.L.; Clarke, S.H. Treatment with a CD40 Antagonist Antibody Reverses Severe Proteinuria and Loss of Saliva Production and Restores Glomerular Morphology in Murine Systemic Lupus Erythematosus. J. Immunol. 2019, 203, 58–75. [Google Scholar] [CrossRef] [PubMed]
  71. Samant, M.; Ziemniak, J.; Paolini, J.F. First-in-Human Phase 1 Randomized Trial with the Anti-CD40 Monoclonal Antibody KPL-404: Safety, Tolerability, Receptor Occupancy, and Suppression of T-Cell-Dependent Antibody Response. J. Pharmacol. Exp. Ther. 2023, 387, 306–314. [Google Scholar] [CrossRef]
  72. Doyle, A.M.; Mullen, A.C.; Villarino, A.V.; Hutchins, A.S.; High, F.A.; Lee, H.W.; Thompson, C.B.; Reiner, S.L. Induction of cytotoxic T lymphocyte antigen 4 (CTLA-4) restricts clonal expansion of helper T cells. J. Exp. Med. 2001, 194, 893–902. [Google Scholar] [CrossRef]
  73. Seo, S.K.; Choi, J.H.; Kim, Y.H.; Kang, W.J.; Park, H.Y.; Suh, J.H.; Choi, B.K.; Vinay, D.S.; Kwon, B.S. 4-1BB-mediated immunotherapy of rheumatoid arthritis. Nat. Med. 2004, 10, 1088–1094. [Google Scholar] [CrossRef] [PubMed]
  74. Tanaka, Y.; Luo, Y.; O’Shea, J.J.; Nakayamada, S. Janus kinase-targeting therapies in rheumatology: A mechanisms-based approach. Nat. Rev. Rheumatol. 2022, 18, 133–145. [Google Scholar] [CrossRef] [PubMed]
  75. Nakayamada, S.; Tanaka, Y. Novel JAK inhibitors under investigation for systemic lupus erythematosus: Where are we now? Expert Opin. Investig. Drugs 2023, 32, 901–908. [Google Scholar] [CrossRef] [PubMed]
  76. Mok, C.C. Targeted Small Molecules for Systemic Lupus Erythematosus: Drugs in the Pipeline. Drugs 2023, 83, 479–496. [Google Scholar] [CrossRef] [PubMed]
  77. Huo, R.; Huang, X.; Yang, Y.; Lin, J. Potential Use of Janus Kinase Inhibitors in the Treatment of Systemic Lupus Erythematosus. J. Inflamm. Res. 2023, 16, 1471–1478. [Google Scholar] [CrossRef] [PubMed]
  78. Nikolopoulos, D.; Parodis, I. Janus kinase inhibitors in systemic lupus erythematosus: Implications for tyrosine kinase 2 inhibition. Front. Med. 2023, 10, 1217147. [Google Scholar] [CrossRef] [PubMed]
  79. Hasni, S.A.; Gupta, S.; Davis, M.; Poncio, E.; Temesgen-Oyelakin, Y.; Carlucci, P.M.; Wang, X.; Naqi, M.; Playford, M.P.; Goel, R.R.; et al. Phase 1 double-blind randomized safety trial of the Janus kinase inhibitor tofacitinib in systemic lupus erythematosus. Nat. Commun. 2021, 12, 3391. [Google Scholar] [CrossRef] [PubMed]
  80. Wallace, D.J.; Furie, R.A.; Tanaka, Y.; Kalunian, K.C.; Mosca, M.; Petri, M.A.; Dörner, T.; Cardiel, M.H.; Bruce, I.N.; Gomez, E.; et al. Baricitinib for systemic lupus erythematosus: A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2018, 392, 222–231. [Google Scholar] [CrossRef] [PubMed]
  81. Morand, E.F.; Vital, E.M.; Petri, M.; van Vollenhoven, R.; Wallace, D.J.; Mosca, M.; Furie, R.A.; Silk, M.E.; Dickson, C.L.; Meszaros, G.; et al. Baricitinib for systemic lupus erythematosus: A double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-I). Lancet 2023, 401, 1001–1010. [Google Scholar] [CrossRef]
  82. Petri, M.; Bruce, I.N.; Dörner, T.; Tanaka, Y.; Morand, E.F.; Kalunian, K.C.; Cardiel, M.H.; Silk, M.E.; Dickson, C.L.; Meszaros, G.; et al. Baricitinib for systemic lupus erythematosus: A double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-II). Lancet 2023, 401, 1011–1019. [Google Scholar] [CrossRef]
  83. Morand, E.; Pike, M.; Merrill, J.T.; van Vollenhoven, R.; Werth, V.P.; Hobar, C.; Delev, N.; Shah, V.; Sharkey, B.; Wegman, T.; et al. Deucravacitinib, a tyrosine kinase 2 inhibitor, in systemic lupus Erythematosus: A phase II, randomized, double-blind. Placebo-Control Trial Arthritis Rheumatol. 2023, 75, 242–252. [Google Scholar] [CrossRef] [PubMed]
  84. Ytterberg, S.R.; Bhatt, D.L.; Mikuls, T.R.; Koch, G.G.; Fleischmann, R.; Rivas, J.L.; Germino, R.; Menon, S.; Sun, Y.; Wang, C.; et al. Cardiovascular and cancer risk with Tofacitinib in rheumatoid arthritis. N. Engl. J. Med. 2022, 386, 316–326. [Google Scholar] [CrossRef]
  85. Khosrow-Khavar, F.; Kim, S.C.; Lee, H.; Lee, S.B.; Desai, R.J. Tofacitinib and risk of cardiovascular outcomes: Results from the safety of TofAcitinib in routine care patients with rheumatoid arthritis (STAR-RA) study. Ann. Rheum. Dis. 2022, 81, 798–804. [Google Scholar] [CrossRef] [PubMed]
  86. Molander, V.; Bower, H.; Frisell, T.; Delcoigne, B.; Di Giuseppe, D.; Askling, J. Venous thromboembolism with JAK inhibitors and other immune-modulatory drugs: A Swedish comparative safety study among patients with rheumatoid arthritis. Ann. Rheum. Dis. 2023, 82, 189–197. [Google Scholar] [CrossRef]
  87. Ma, L.; Peng, L.; Zhao, J.; Bai, W.; Jiang, N.; Zhang, S.; Wu, C.; Wang, L.; Xu, D.; Leng, X.; et al. Efficacy and safety of Janus kinase inhibitors in systemic and cutaneous lupus erythematosus: A systematic review and meta-analysis. Autoimmun. Rev. 2023, 22, 103440. [Google Scholar] [CrossRef]
  88. Richter, P.; Cardoneanu, A.; Burlui, A.M.; Macovei, L.A.; Bratoiu, I.; Buliga-Finis, O.N.; Rezus, E. Why Do We Need JAK Inhibitors in Systemic Lupus Erythematosus? Int. J. Mol. Sci. 2022, 23, 11788. [Google Scholar] [CrossRef]
  89. Dean, G.S.; Tyrrell-Price, J.; Crawley, E.; Isenberg, D.A. Cytokines and systemic lupus erythematosus. Ann. Rheum. Dis. 2000, 59, 243–251. [Google Scholar] [CrossRef]
  90. Riaz, M.F.; Garg, G.; Umeano, L.; Iftikhar, S.; Alhaddad, S.F.; Paulsingh, C.N.; Hamid, P. Comparison of Low-Dose Interleukin 2 Therapy in Conjunction With Standard Therapy in Patients With Systemic Lupus Erythematosus vs Rheumatoid Arthritis: A Systematic Review. Cureus 2024, 16, e56704. [Google Scholar] [CrossRef] [PubMed]
  91. von Spee-Mayer, C.; Siegert, E.; Abdirama, D.; Rose, A.; Klaus, A.; Alexander, T.; Enghard, P.; Sawitzki, B.; Hiepe, F.; Radbruch, A.; et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann. Rheum. Dis. 2016, 75, 1407–1415. [Google Scholar] [CrossRef]
  92. Comte, D.; Karampetsou, M.P.; Tsokos, G.C. T cells as a therapeutic target in SLE. Lupus 2015, 24, 351–363. [Google Scholar] [CrossRef]
  93. Dai, H.; He, F.; Tsokos, G.C.; Kyttaris, V.C. IL-23 Limits the production of IL-2 and promotes autoimmunity in lupus. J. Immunol. 2017, 199, 903–910. [Google Scholar] [CrossRef] [PubMed]
  94. He, J.; Zhang, X.; Wei, Y.; Sun, X.; Chen, Y.; Deng, J.; Jin, Y.; Gan, Y.; Hu, X.; Jia, R.; et al. Low-dose interleukin-2 treatment selectively modulates CD4(+) T cell subsets in patients with systemic lupus erythematosus. Nat. Med. 2016, 22, 991–993. [Google Scholar] [CrossRef] [PubMed]
  95. He, J.; Zhang, R.; Shao, M.; Zhao, X.; Miao, M.; Chen, J.; Liu, J.; Zhang, X.; Zhang, X.; Jin, Y.; et al. Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: A randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2020, 79, 141–149. [Google Scholar] [CrossRef] [PubMed]
  96. Humrich, J.Y.; Cacoub, P.; Rosenzwajg, M.; Pitoiset, F.; Pham, H.P.; Guidoux, J.; Leroux, D.; Vazquez, T.; Riemekasten, G.; Smolen, J.S.; et al. Low-dose interleukin-2 therapy in active systemic lupus erythematosus (LUPIL-2): A multicentre, double-blind, randomised and placebo-controlled phase II trial. Ann. Rheum. Dis. 2022, 81, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  97. Humrich, J.Y.; Riemekasten, G. Clinical trials: The rise of IL-2 therapy—A novel biologic treatment for SLE. Nat. Rev. Rheumatol. 2016, 12, 695–696. [Google Scholar] [CrossRef] [PubMed]
  98. Clinical Trials. Available online: https://clinicaltrials.gov/search?cond=Lupus%20Erythematosus,%20Systemic&term=IL-2%20&intr=IL2 (accessed on 14 March 2024).
  99. Gröndal, G.; Gunnarsson, I.; Rönnelid, J.; Rogberg, S.; Klareskog, L.; Lundberg, I. Cytokine production, serum levels and disease activity in systemic lupus erythematosus. Clin. Exp. Rheumatol. 2000, 18, 565–570. [Google Scholar] [PubMed]
  100. Ohl, K.; Tenbrock, K. Inflammatory cytokines in systemic lupus erythematosus. J. Biomed. Biotechnol. 2011, 2011, 432595. [Google Scholar] [CrossRef] [PubMed]
  101. Ripley, B.J.; Goncalves, B.; Isenberg, D.A.; Latchman, D.S.; Rahman, A. Raised levels of interleukin 6 in systemic lupus erythematosus correlate with anemia. Ann. Rheum. Dis. 2005, 64, 849–853. [Google Scholar] [CrossRef] [PubMed]
  102. Maeda, K.; Mehta, H.; Drevets, D.A.; Coggeshall, K.M. IL-6 increases B-cell IgG production in a feed-forward proinflammatory mechanism to skew hematopoiesis and elevate myeloid production. Blood 2010, 115, 4699–4706. [Google Scholar] [CrossRef]
  103. Kimura, A.; Kishimoto, T. IL-6: Regulator of Treg/Th17 balance. Eur. J. Immunol. 2010, 40, 1830–1835. [Google Scholar] [CrossRef]
  104. Wallace, D.J.; Strand, V.; Merrill, J.T.; Popa, S.; Spindler, A.J.; Eimon, A.; Petri, M.; Smolen, J.S.; Wajdula, J.; Christensen, J.; et al. Efficacy and safety of an interleukin 6 monoclonal antibody for the treatment of systemic lupus erythematosus: A phase II dose-ranging randomized controlled trial. Ann. Rheum. Dis. 2017, 76, 534–542. [Google Scholar] [CrossRef] [PubMed]
  105. Robert, M.; Miossec, P. Interleukin-17 and lupus: Enough to be a target? For which patients? Lupus 2020, 29, 6–14. [Google Scholar] [CrossRef] [PubMed]
  106. Szepietowski, J.C.; Nilganuwong, S.; Wozniacka, A.; Kuhn, A.; Nyberg, F.; van Vollenhoven, R.F.; Bengtsson, A.A.; Reich, A.; de Vries, D.E.; van Hartingsveldt, B.; et al. Phase I, randomized, double-blind, placebo-controlled, multiple intravenous, dose-ascending study of sirukumab in cutaneous or systemic lupus erythematosus. Arthritis Rheum. 2013, 65, 2661–2671. [Google Scholar] [CrossRef] [PubMed]
  107. Illei, G.G.; Shirota, Y.; Yarboro, C.H.; Daruwalla, J.; Tackey, E.; Takada, K.; Fleisher, T.; Balow, J.E.; Lipsky, P.E. Tocilizumab in systemic lupus erythematosus: Data on safety, preliminary efficacy, and impact on circulating plasma cells from an open-label phase I dosage-escalation study. Arthritis Rheum. 2010, 62, 542–552. [Google Scholar] [CrossRef] [PubMed]
  108. Koga, T.; Ichinose, K.; Kawakami, A.; Tsokos, G.C. The role of IL-17 in systemic lupus erythematosus and its potential as a therapeutic target. Expert Rev. Clin. Immunol. 2019, 15, 629–637. [Google Scholar] [CrossRef] [PubMed]
  109. US National Library of Medicine. Study of Safety, Efficacy and Tolerability of Secukinumab versus Placebo, in Combination with SoC Therapy, in Patients with Active Lupus Nephritis (SELUNE). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04181762 (accessed on 20 June 2024).
  110. Costa, R.; Antunes, P.; Salvador, P.; Oliveira, P.; Marinho, A. Secukinumab on Refractory Lupus Nephritis. Cureus 2021, 13, e17198. [Google Scholar] [CrossRef] [PubMed]
  111. Avila-Ribeiro, P.; Lopes, A.R.; Martins-Martinho, J.; Nogueira, E.; Antunes, J.; Romeu, J.C.; Cruz-Machado, A.R.; Vieira-Sousa, E. Secukinumab-induced systemic lupus erythematosus in psoriatic arthritis. ARP Rheumatol. 2023, 2, 265–268. [Google Scholar] [PubMed]
  112. Koller-Smith, L.; Oakley, S. Secukinumab-induced systemic lupus erythematosus occurring in a patient with ankylosing spondylitis. Rheumatology 2022, 61, e146–e147. [Google Scholar] [CrossRef] [PubMed]
  113. Ueno, H. The IL-12-STAT4 axis in the pathogenesis of human systemic lupus erythematosus. Eur. J. Immunol. 2020, 50, 10–16. [Google Scholar] [CrossRef]
  114. Wong, C.K.; Lit, L.C.W.; Tam, L.S.; Li, E.K.M.; Wong, P.T.Y.; Lam, C.W.K. Hyperproduction of IL- 23 and IL-17 in patients with systemic lupus erythematosus: Implications for Th17-mediated inflammation in auto-immunity. Clin. Immunol. 2008, 127, 385–393. [Google Scholar] [CrossRef]
  115. Zickert, A.; Amoudruz, P.; Sundström, Y.; Rönnelid, J.; Malmström, V.; Gunnarsson, I. IL-17 and IL-23 in lupus nephritis—Association to histopathology and response to treatment. BMC Immunol. 2015, 16, 7. [Google Scholar] [CrossRef]
  116. van Vollenhoven, R.F.; Hahn, B.H.; Tsokos, G.C.; Wagner, C.L.; Lipsky, P.; Touma, Z.; Werth, V.P.; Gordon, R.M.; Zhou, B.; Hsu, B.; 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, 1330–1339. [Google Scholar] [CrossRef]
  117. Facciotti, F.; Larghi, P.; Bosotti, R.; Vasco, C.; Gagliani, N.; Cordiglieri, C.; Mazzara, S.; Ranzani, V.; Rottoli, E.; Curti, S.; et al. Evidence for a pathogenic role of extrafollicular, IL-10-producing CCR6+B helper T cells in systemic lupus erythematosus. Proc. Natl. Acad. Sci. USA 2020, 117, 7305–7316. [Google Scholar] [CrossRef]
  118. Godsell, J.; Rudloff, I.; Kandane-Rathnayake, R.; Hoi, A.; Nold, M.F.; Morand, E.F.; Harris, J. Clinical associations of IL-10 and IL-37 in systemic lupus erythematosus. Sci. Rep. 2016, 6, 34604. [Google Scholar] [CrossRef] [PubMed]
  119. Available online: https://clinicaltrials.gov/study/NCT02554019 (accessed on 14 March 2024).
  120. Hannon, C.W.; McCourt, C.; Lima, H.C.; Chen, S.; Bennett, C. Interventions for cutaneous disease in systemic lupus erythematosus. Cochrane Database Syst. Rev. 2021, 3, CD007478. [Google Scholar] [CrossRef] [PubMed]
  121. Kirou, K.A.; Lee, C.; George, S.; Louca, K.; Papagiannis, I.G.; Peterson, M.G.E.; Ly, N.; Woodward, R.N.; Fry, K.E.; Lau, A.Y.; et al. Coordinate overexpression of interferon-alpha-induced genes in systemic lupus erythematosus. Arthritis Rheum. 2004, 50, 3958–3967. [Google Scholar] [CrossRef] [PubMed]
  122. Niewold, T.B.; Hua, J.; Lehman, T.J.A.; Harley, J.B.; Crow, M.K. High serum IFN-alpha activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007, 8, 492–502. [Google Scholar] [CrossRef]
  123. Baechler, E.C.; Batliwalla, F.M.; Karypis, G.; Gaffney, P.M.; Ortmann, W.A.; Espe, K.J.; Shark, K.B.; Grande, W.J.; Hughes, K.M.; Kapur, V.; et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc. Natl. Acad. Sci. USA 2003, 100, 2610–2615. [Google Scholar] [CrossRef]
  124. Zhuang, H.; Szeto, C.; Han, S.; Yang, L.; Reeves, W.H. Animal Models of Interferon Signature Positive Lupus. Front. Immunol. 2015, 6, 291. [Google Scholar] [CrossRef]
  125. Li, Q.Z.; Zhou, J.; Lian, Y.; Zhang, B.; Branch, V.K.; Carr-Johnson, F.; Karp, D.R.; Mohan, C.; Wakeland, E.K.; Olsen, N.J. Interferon signature gene expression is correlated with autoantibody profiles in patients with incomplete lupus syndromes. Clin. Exp. Immunol. 2010, 159, 281–291. [Google Scholar] [CrossRef]
  126. Brohawn, P.Z.; Streicher, K.; Higgs, B.W.; Morehouse, C.; Liu, H.; Illei, G.; Ranade, K. Type I interferon gene signature test–low and–high patients with systemic lupus erythematosus have distinct gene expression signatures. Lupus 2019, 28, 1524–1533. [Google Scholar] [CrossRef] [PubMed]
  127. Chasset, F.; Mathian, A.; Dorgham, K.; Ribi, C.; Trendelenburg, M.; Huynh-Do, U.; Roux-Lombard, P.; Courvoisier, D.S.; Amoura, Z.; Gorochov, G.; et al. Serum interferon-α levels and IFN type I- stimulated genes score perform equally to assess systemic lupus erythematosus disease activity. Ann. Rheum. Dis. 2022, 81, 901–903. [Google Scholar] [CrossRef] [PubMed]
  128. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef] [PubMed]
  129. Sim, T.M.; Ong, S.J.; Mak, A.; Tay, S.H. Type I Interferons in Systemic Lupus Erythematosus: A Journey from Bench to Bedside. Int. J. Mol. Sci. 2022, 23, 2505. [Google Scholar] [CrossRef] [PubMed]
  130. Mathian, A.; Mouries-Martin, S.; Dorgham, K.; Devilliers, H.; Yssel, H.; Castillo, L.G.; Cohen-Aubart, F.; Haroche, J.; Hié, M.; de Chambrun, M.P.; et al. Ultrasensitive serum interferon-α quantification during SLE remission identifies patients at risk for relapse. Ann. Rheum. Dis. 2019, 78, 1669–1676. [Google Scholar] [CrossRef] [PubMed]
  131. Furie, R.; Khamashta, M.; Merrill, J.T.; Werth, V.P.; Kalunian, K.; Brohawn, P.; Illei, G.G.; Drappa, J.; Wang, L.; Yoo, S.; et al. Anifrolumab, an anti–interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol. 2017, 69, 376–386. [Google Scholar] [CrossRef] [PubMed]
  132. Jones, S.A.; Morand, E.F. Targeting Interferon Signalling in Systemic Lupus Erythematosus: Lessons Learned. Drugs 2024, 84, 625–635. [Google Scholar] [CrossRef] [PubMed]
  133. Khamashta, M.; Merrill, J.T.; Werth, V.P.; Furie, R.; Kalunian, K.; Illei, G.G.; Drappa, J.; Wang, L.; Greth, W. Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: A randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016, 75, 1909–1916. [Google Scholar] [CrossRef]
  134. Furie, R.A.; Morand, E.F.; Bruce, I.N.; Manzi, S.; Kalunian, K.C.; Vital, E.M.; Ford, T.L.; Gupta, R.; Hiepe, F.; Santiago, M.; et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): A randomised, controlled, phase 3 trial. Lancet Rheumatol. 2019, 1, e208–e219. [Google Scholar] [CrossRef]
  135. Morand, E.F.; Furie, R.; Tanaka, Y.; Bruce, I.N.; Askanase, A.D.; Richez, C.; Bae, S.-C.; Brohawn, P.Z.; Pineda, L.; Berglind, A.; et al. Trial of anifrolumab in active systemic lupus erythematosus. N. Engl. J. Med. 2020, 382, 211–221. [Google Scholar] [CrossRef]
  136. Bruce, I.N.; Furie, R.A.; Morand, E.F.; Manzi, S.; Tanaka, Y.; Kalunian, K.C.; Merrill, J.T.; Puzio, P.; Maho, E.; Kleoudis, C.; et al. Concordance and discordance in SLE clinical trial outcome measures: Analysis of three anifrolumab phase 2/3 trials. Ann. Rheum. Dis. 2022, 81, 962–969. [Google Scholar] [CrossRef] [PubMed]
  137. Jayne, D.; Rovin, B.; Mysler, E.F.; Furie, R.A.; Houssiau, F.A.; Trasieva, T.; Knagenhjelm, J.; Schwetje, E.; Chia, Y.L.; Tummala, R.; et al. Phase II randomized trial of type I interferon inhibitor anifrolumab in patients with active lupus nephritis. Ann. Rheum. Dis. 2022, 81, 496–506. [Google Scholar] [CrossRef] [PubMed]
  138. Jayne, D.; Rovin, B.; Mysler, E.F.; Furie, R.A.; Houssiau, F.A.; Trasieva, T.; Knagenhjelm, J.; Schwetje, E.; Chia, Y.L.; Tummala, R.; 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. 2022, 4, e282–e292. [Google Scholar]
  139. Kalunian, K.C.; Merrill, J.T.; Maciuca, R.; McBride, J.M.; Townsend, M.J.; Wei, X.; Davis, J.C.; Kennedy, W.P. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-alpha) in patients with systemic lupus erythematosus (ROSE). Ann. Rheum. Dis. 2016, 75, 196–202. [Google Scholar] [CrossRef]
  140. Zagury, D.; Le Buanec, H.; Mathian, A.; Larcier, P.; Burnett, R.; Amoura, Z.; Emilie, D.; Peltre, G.; Bensussan, A.; Bizzini, B.; et al. IFNalpha kinoid vaccine-induced neutralizing antibodies prevent clinical manifestations in a lupus flare murine model. Proc. Natl. Acad. Sci. USA 2009, 106, 5294–5299. [Google Scholar] [CrossRef] [PubMed]
  141. Houssiau, F.A.; Thanou, A.; Mazur, M.; Ramiterre, E.; Mora, D.A.G.; Misterska-Skora, M.; Perich-Campos, R.A.; Smakotina, S.A.; Cruz, S.C.; Louzir, B.; et al. IFN-α kinoid in systemic lupus erythematosus: Results from a phase IIb, randomized, placebo-controlled study. Ann. Rheum. Dis. 2020, 79, 347–355. [Google Scholar] [CrossRef] [PubMed]
  142. Schett, G.; Mackensen, A.; Mougiakakos, D. CAR T-cell therapy in autoimmune diseases. Lancet 2023, 402, 2034–2044. [Google Scholar] [CrossRef] [PubMed]
  143. Kretschmann, S.; Völkl, S.; Reimann, H.; Krönke, G.; Schett, G.; Achenbach, S.; Lutzny-Geier, G.; Müller, F.; Mougiakakos, D.; Dingfelder, J.; et al. Successful Generation of CD19 Chimeric Antigen Receptor T Cells from Patients with Advanced Systemic Lupus Erythematosus. Transpl. Cell Ther. 2023, 29, 27–33. [Google Scholar] [CrossRef] [PubMed]
  144. Taubmann, J.; Müller, F.; Mutlu, M.Y.; Völkl, S.; Aigner, M.; Bozec, A.; Mackensen, A.; Grieshaber-Bouyer, R.; Schett, G. CD19 Chimeric Antigen Receptor T Cell Treatment: Unraveling the Role of B Cells in Systemic Lupus Erythematosus. Arthritis Rheumatol. 2023, 76, 497–504. [Google Scholar] [CrossRef]
  145. Mougiakakos, D.; Krönke, G.; Völkl, S.; Kretschmann, S.; Aigner, M.; Kharboutli, S.; Böltz, S.; Manger, B.; Mackensen, A.; Schett, G. CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus. N. Engl. J. Med. 2021, 385, 567–569. [Google Scholar] [CrossRef] [PubMed]
  146. Mackensen, A.; Mackensen, A.; Müller, F.; Müller, F.; Mougiakakos, D.; Mougiakakos, D.; Böltz, S.; Böltz, S.; Wilhelm, A.; Wilhelm, A.; et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat. Med. 2022, 28, 2124–2132, Erratum in Nat. Med. 2023, 29, 2956. https://doi.org/10.1038/s41591-022-02091-9. [Google Scholar] [CrossRef] [PubMed]
  147. Müller, F.; Taubmann, J.; Bucci, L.; Wilhelm, A.; Bergmann, C.; Völkl, S.; Aigner, M.; Rothe, T.; Minopoulou, I.; Tur, C.; et al. CD19 CAR T-Cell Therapy in Autoimmune Disease—A Case Series with Follow-up. N. Engl. J. Med. 2024, 390, 687–700. [Google Scholar] [CrossRef] [PubMed]
  148. Clinical Trials. Available online: www.clinicaltrials.gov (accessed on 14 March 2024).
Figure 1. Factors for SLE development and pathogenesis, including immune mechanisms, targeted organs, and targeted therapies.
Figure 1. Factors for SLE development and pathogenesis, including immune mechanisms, targeted organs, and targeted therapies.
Rheumato 04 00009 g001
Table 1. Novel targeted therapies for SLE patients.
Table 1. Novel targeted therapies for SLE patients.
Molecular Pathway and Corresponding Agent Tested in Clinical Trials in SLE
Molecular TargetTherapeutic AgentTypeResults
B-cell inhibition
BAFF/APRILBelimumab, BenlystaFully humanized IgG1 monoclonal antibodyApproved for active, antibody-positive, non-renal SLE
TabalumabFully human IgG4 monoclonal antibodyFailed to meet its primary endpoints
BlisibimodFusion proteinFailed to meet its primary endpoints
AtaciceptFully human recombinant fusion proteinAPRIL-SLE terminated; ADDRESSII had a better safety profile
CD20RituximabChimeric monoclonal antibodyLUNAR/EXPLORER-negative; accoring to EULAR it could be used aslast choice for severe refractory lupus
OcrelizumabFully humanized monoclonal antibodyTrials were terminated because of severe infections
ObinutuzumabFully humanized monoclonal antibodyPhase III ongoing
CD22EpratuzumabFully humanized monoclonal antibodyPhase III failed
CD19XmAb5871Monoclonal antibodyUnder investigation
Proteasome inhibitorsBortezomibA modified dipeptidyl boronic acidTerminated phase II trial because of adverse reactions
Co-stimulation
CD28/B7AbataceptA fusion protein (CTLA4-Ig)Ineffective in phase II in nephritis and general SLE
CD40/CD154DapirolizumabPEG-conjugated antiCD40L Fab fragmentOngoing
Intracellular signaling
Bruton’s tyrosine kinaseFenebrutinibSmall molecule, tyrosine kinaseGood safety profile, failed to meet its primary endpoint
Calcineurin inhibitorVoclosporinCalcineurin inhibitorApproved by FDA for lupus nephritis
JAK1UpadacitinibSmall moleculePhase III trial in SLE
JAK1/2BaricitinibSmall moleculePhase III discontinued
JAK1/3TofacitinibSmall moleculeUnder investigation
Tyk2 (JAK4)DeucravacitinibSmall moleculeUnder investigation
Cytokines
Interferon-αSifalimumabFully human monoclonal antibodyNo effect; discontinued
RontalizumabHumanized monoclonal antibodyWell tolerated, but no additional trials planned
AnifrolumabFully human monoclonal antibodyApproved for moderate and severe SLE and SLE nephritis
IFN-KInactivated IFN-α coupled with proteinFailed to meet its primary endpoint
IL-2Aldesleukin; AMG592CytokineUnder investigation
IL-12/23UstekinumabFully human monoclonal antibodySuspended phase III over efficacy data
IL-6SirukumabFully human monoclonal antibodyFailed
IL-17Secukinumab Fully human monoclonal antibodyPhase III clinical trial
IL-10Anti-IL-10 antibody Fully human monoclonal antibodyHas completed a phase II trial (NCT02554019)
CAR T cell therapyChimeric–antigen receptors are recombinant receptors that are coupled to autologous T cells to target a specific antigen-In clinical trials for autoimmune diseases, incl. SLE
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Velikova, T.; Miteva, D.; Kokudeva, M.; Vasilev, G.H.; Monov, S.; Shumnalieva, R. In the Pursuit of Precision: Novel Target Therapies Revolutionizing SLE Care. Rheumato 2024, 4, 120-136. https://doi.org/10.3390/rheumato4030009

AMA Style

Velikova T, Miteva D, Kokudeva M, Vasilev GH, Monov S, Shumnalieva R. In the Pursuit of Precision: Novel Target Therapies Revolutionizing SLE Care. Rheumato. 2024; 4(3):120-136. https://doi.org/10.3390/rheumato4030009

Chicago/Turabian Style

Velikova, Tsvetelina, Dimitrina Miteva, Maria Kokudeva, Georgi H. Vasilev, Simeon Monov, and Russka Shumnalieva. 2024. "In the Pursuit of Precision: Novel Target Therapies Revolutionizing SLE Care" Rheumato 4, no. 3: 120-136. https://doi.org/10.3390/rheumato4030009

Article Metrics

Back to TopTop