Back to Journals » ImmunoTargets and Therapy » Volume 14
Immunotargets and Therapy for Systemic Lupus Erythematosus
Authors Mok CC
Received 22 February 2025
Accepted for publication 16 June 2025
Published 24 June 2025 Volume 2025:14 Pages 605—629
DOI https://doi.org/10.2147/ITT.S485650
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Michael Shurin
Chi Chiu Mok
Department of Medicine and Geriatrics, Tuen Mun Hospital, Hong Kong
Correspondence: Chi Chiu Mok, Department of Medicine & Geriatrics, Tuen Mun Hospital, Tsing Chung Koon Road, New Territories, Hong Kong, SAR, People’s Republic of China, Tel +85224685386, Fax +85224569100, Email [email protected]
Abstract: The pathophysiology of systemic lupus erythematosus (SLE) is complex and involves most cell types of the innate and adaptive immune system. Impaired clearance of apoptotic bodies and self-antigens, dysregulated cytokine network and aberrated functions of the immune cells lead to overproduction of autoantibodies, activation of complements, immune complex deposition and tissue injury. Novel biological and newer generation immunosuppressive agents have been developed to target the B cells, T cells, T/B cell interaction, plasmacytoid dendritic cells and the cytokines. With the advances in the knowledge about the intracellular pathways, small molecules that inhibit the downstream signal transduction from surface receptors and intracellular protein degradation by the ubiquitin-proteasome system are being developed in the pipeline. This article summarizes the evidence of various immunotargets for the treatment of SLE. These novel agents target specific cellular mechanisms, and further works are necessary to stratify patients according to biomarkers to receive individualized therapies that could help maximize the clinical response. With the availability of more therapeutic choices, a combination approach to achieve synergistic effects while reducing adverse events by dosage reduction of individual drugs is being explored for SLE patients at risk of disease progression or refractory to conventional therapies.
Keywords: target, immune, therapeutics, novel, biologics, small molecules, lupus
Introduction
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that predominantly affects women in their childbearing age. The illness is characterized by periods of quiescence and flares, which are notoriously unpredictable The onset of SLE cannot be prevented and there is no cure. Despite the availability of new drugs and improvement in adjunctive therapies, the survival of SLE patients fails to improve further after the mid-1990s’.1 Conventional immunomodulatory drugs, except for hydroxychloroquine, have not been shown to confer survival benefit in SLE,2 probably related to their adverse effects, such as infective complications, which still account for more than 50% of the mortality in more recent cohorts of SLE patients.3 This calls for the development of novel therapeutic approaches with a better efficacy-to-toxicity balance.4
The pathogenesis of SLE remains obscure.5 A plethora of genetic, epigenetic and environmental factors, including infective agents, hormonal factors and ultraviolet light, are involved and lead to dysregulation of the innate and adaptive immune responses.6 Genome- wide association studies (GWAS) have identified more than 200 risk loci in SLE,7 many of which are related to antigen presentation, immune regulation and interferon (IFN) signalling.8 Abnormal epigenetic mechanisms such as DNA methylation, histone modification, noncoding RNAs and RNA methylation also contribute to the modification of gene expression.9 Pathological processes that are probably involved in the initiation and perpetuation of the immune perturbations in SLE include: (1) loss of self-tolerance and generation of autoreactive B cells and T cells, leading to the production of antinuclear (ANA) and other autoantibodies10 (2) defective clearance of apoptotic materials, nuclear antigens, nucleosomes and immune complexes (ICs) by macrophages and complements6,11,12 (3) impaired neutrophil apoptosis and degradation of DNA, histones, cytoplasmic granules and other mediators inside the neutrophil extracellular traps (NETs).13–16 These antigens activate the plasmacytoid dendritic cells (pDCs) via the toll-like receptors (TLR7/9) to produce type I interferons (IFNs) and interleukin (IL)-6, which in turn accelerate monocyte maturation, inhibit T cell apoptosis, increase proliferation and autoantibody production by B cells.10,12 Increased activity of the myeloid dendritic cells17 and defective functions of the regulatory B (Bregs) and T cells (Tregs)18–20 polarizes the T cells to differentiate into the proinflammatory phenotypes (Th1/Th17). The augmented autoantibody production and IC formation mediates tissue inflammation and organ damage in patients with SLE.
Despite the negative results from multiple clinical trials of SLE drugs, newer biologic and targeted small molecules continue to emerge from the pipeline21 (Table 1). This article reviews the various immune targets for novel SLE therapies (Figure 1).
![]() |
Table 1 Immunotargets and Therapy of Systemic Lupus Erythematosus |
Targeting the B Cells
B cells are central in the pathophysiology of SLE because they produce pathogenic antibodies. In addition, they are capable of presenting self-antigens to activate the autoreactive T cells22,23 and produce cytokines that amplify the inflammatory response.20 A subset of IL-10 secreting Bregs that suppresses the inflammatory response are altered in SLE, which is coupled with an expansion of transitional B cells which are dependent on B cell activating factor (BAFF) for maturation and a subpopulation of autoreactive age-associated B cells (ABCs).23,24
B cells can be targeted by direct depletion with monoclonal antibodies (anti-CD19, anti-CD20), engineered T cells (Chimeric Antigen Receptor T cells [CAR-T]) and dual action monoclonal antibodies, or indirect modulation via inhibition of growth factors (eg BAFF), costimulatory molecules and intracellular signalling pathways (eg Bruton’s tyrosine kinase [BTK]).25
B-Cell Depletion
Anti-CD20
The chimeric anti-CD20 monoclonal antibody, rituximab, was first studied in SLE. In the pivotal randomized controlled trial (RCT) of active non-renal SLE, two courses of rituximab (1gm for 2 doses) on background standard of care (SOC) did not report benefit in disease activity reduction at week 52 compared with placebo (PBO).26 Another RCT (LUNAR) of rituximab in lupus nephritis (LN) also failed to meet its primary end point of renal response at week 52 when added to glucocorticoid (GC) and mycophenolate mofetil (MMF).27 While rituximab was more effective than PBO in improving anti-dsDNA and complement levels, neutropenia, leukopenia, hypotension, infusion reactions, herpes zoster, and opportunistic infections were more common. Despite the futility of these RCTs, rituximab continued to be used off-label to treat refractory SLE and LN, with clinical responses reported in 67–86% of patients.28
Ocrelizumab is a fully humanized anti-CD20 biologic that was subsequently studied in SLE.29,30 In two RCTs of non-renal SLE and LN, patients were randomly assigned to receive two courses of ocrelizumab (400 mg or 1000 mg for 2 doses 2-weekly and after 4 months) or PBO. Patients in the LN trial received the SOC with GC combined with either MMF (3g/day) or low-dose cyclophosphamide (CYC) followed by azathioprine (AZA). Both studies were terminated prematurely because of severe serious infections. An analysis of the data from patients who received study drug for ≥32 weeks showed that the overall renal response rate of the combined ocrelizumab groups was non-significantly higher than that of PBO.30 The effect size was higher with ocrelizumab combining with CYC than MMF whereas serious infections were more common in the MMF/ocrelizumab group.
As the serious infections in the ocrelizumab studies were potentially related to the protocol-based high-dose of MMF being used in Asian patients, subsequent RCTs of anti-CD20 biologics in SLE adopted a lower dose of MMF and less intense background immunosuppression.31 Obinutuzumab is a recombinant humanized type II anti-CD20 monoclonal antibody that is glyco-engineered for greater affinity for the FcγRIII on effector cells, leading to enhanced antibody dependent cellular cytotoxicity (ADCC), direct B-cell killing, and less reliance on complement dependent cytotoxicity (CDC).32 Obinutuzumab is more effective than rituximab in depleting B cells and ameliorating LN in the lupus mice.33
A Phase II RCT (NOBILITY) of 125 patients with active proliferative LN who were receiving a SOC of GC and MMF showed that the complete renal response (CRR) rate at week 52 through week 104 in the obinutuzumab arm was superior to PBO.31 Obinutuzumab was not associated with an increase in serious adverse events (SAEs), serious infections or deaths. Post-hoc analyses demonstrated that the anti-CD20 agent reduced the incidence of LN flares, first estimated glomerular filtration rate (eGFR) decline by 30% or 40%, and a composite outcome of treatment failure, doubling of serum creatinine, or death.34 The Phase III RCT of obinutuzumab in 271 patients with active LN (REGENCY) confirmed its efficacy compared to PBO, in conjunction with GC and MMF, at week 76 in terms of CRR or CRR together with a maintenance prednisone dose of ≤7.5mg/day.35 Despite the absence of unexpected safety signals, SAEs and infections were more frequent in obinutuzumab-treated patients. Greater adjusted between-group differences in the rate of CRR were observed in those patients with higher serological SLE activity, uP/Cr>3.0 and histological class IV±V disease.
A GC-free regimen consisting of rituximab (1 gram for 2 doses) and MMF (dosage adjusted by drug level) has been explored in a single arm prospective study of 50 patients with LN.36 Complete or partial renal response was reported in 90% of patients treated with this regimen (RITUXILUP regimen) without concomitant oral prednisone over a median time of 37 weeks. As a planned RCT of this regimen was aborted (NCT01773616), it is hoped that the ongoing RCT of obinutuzumab/MMF versus GC/MMF in LN (OBILUP; NCT04702256) would provide new insights on the efficacy of this GC-free regimen in LN.
Anti-CD22
Epratuzumab is a human monoclonal antibody that targets the CD22 antigen on mature B cells, which is involved in the modulation of B-cell receptor (BCR) signalling, cellular activation and survival.37 Compared to CD20, CD22 blockade is less cytotoxic. Epratuzumab diminished B cell proliferation, migration and production of inflammatory cytokines.38 Early clinical trials showed that epratuzumab induced a modest depletion of peripheral B cells without significant effects on T cells, autoantibody or immunoglobulin levels.39,40 Following a phase IIb PBO-controlled RCT of epratuzumab (ENBLEM) demonstrating safety and preliminary efficacy in SLE,41,42 two subsequent phase III RCTs were conducted (EMBODY 1/2).43 Unfortunately, the primary clinical improvement end point, BILAG-based combined lupus assessment [BICLA] at week 48, was not met, although adverse events (AEs) and treatment-emergent AEs (TEAEs) were not increased with treatment.
Anti-CD19 and T-Cell Engagers
Obexelimab is dual function, non-cytolytic monoclonal antibody that binds to CD19 and FcyRIIb to inhibit B cells and plasmablasts.44 A phase II PBO-controlled RCT of obexelimab in 104 SLE patients with non-organ-threatening manifestations did not meet the primary end point of reaching week 32 without loss of improvement, although the time to this end point was prolonged with obexelimab compared to PBO.45 B cells decreased by approximately 50% after anti-CD19 treatment and the presence of gene expression clusters with high B cell pathway modules were associated with longer time to loss of improvement. Another anti-CD19 monoclonal, inebilizumab, approved for treatment of neuromyelitis optica spectrum disorder (NMOSD) in adult patients, is registered for a study in refractory SLE (NCT06570798).
The anti-CD3/anti-CD19 bispecific T cell engager (BiTE), blinatumomab, instigates CD3-positive T cells to target CD19-positive B cells,46 leading to B cell elimination and concomitant reduction of T cells. Blinatumomab is effective against acute lymphoblastic leukemia and other B cell malignancies. As the drug targets both malignant and non-malignant lymphocytes, it may potentially cause the cytokine release syndrome and neurotoxicity similar to CAR-T therapy. Blinatumomab has been shown to be effective in refractory rheumatoid arthritis47 and systemic sclerosis.48 As the effect of blinatumomab mimics CAR-T but does not require preconditioning with lymphodepletion and chemotherapy, it is a more acceptable option in younger SLE patients.49 Two studies of blinatumomab in refractory SLE have been registered (NCT06789107 and NCT06570798).
Teclistamab is a CD3/B-cell maturation antigen (BCMA) BiTE that is used to treat refractory myeloma.50 Unlike CD3/CD19 BiTE, it has a capacity to deplete the long-lived plasma cells. Successful use of teclistamab has been reported in a patient with refractory SLE.51 Teclistamab led to a rapid depletion of peripheral-blood B cells by week 1 and eradication of marrow B cells and plasma cells by week 8. However, no clinical trials in SLE have been registered yet.
Car-t
CAR-T therapy involves the use of engineered T cells to target CD19 on B cells or BCMA on myeloma cells.25,52 CAR-T depletes B cells profoundly, including the memory B cells, plasmablasts and plasma cells. Pilot studies of CD19-targeted CAR-T therapy showed good response in patients with severe or refractory SLE.53 However, the procedure involves pre-conditioning with cyclophosphamide and fludarabine, which is undesirable for fertility and oncogenicity reasons. Moreover, it carries the risk of cytokine release and immune effector cell-associated neurotoxicity syndromes. Publication bias, relatively small number of SLE patients treated with CAR-T, lack of long-term data on safety and efficacy, and the general unavailability of this procedure are the major limitations of CAR-T in SLE.5
Long-Lived Plasma Cells
Long-lived plasma cells are capable of generating autoreactive immunologic memory and autoantibodies that are relevant for the pathogenesis of SLE.54 In contrast to short-lived plasmablasts, they are resistant to conventional immunosuppressive and B cell depletion therapies because of the lack of CD19 and CD20 expression.55 However, these plasma cells have upregulation of the autophagy pathways and the unfolded protein response, leading to the susceptibility to proteasome inhibition.25 Targeting the immunoproteasomes and the cereblon E3 ligase in the ubiquitin-proteasome system (UPS), which is a major pathway for intracellular protein degradation, is a novel therapeutic approach in SLE.56,57
Bortezomib is a non-selective inhibitor of both the constitutive and immune proteasomes. Case series have reported efficacy of bortezomib in conjunction with GCs for the treatment of refractory SLE manifestations, including LN.58–62 Bortezomib depleted both short-lived and long-lived plasma cells in the peripheral blood and bone marrow, which was associated with improvement of anti-dsDNA level and proteinuria in LN.58,60–62 However, the drug is limited by gastrointestinal toxicities, peripheral neuropathy and drug resistance on prolonged use.63
Zetomipzomib is a highly selective immunoproteasome inhibitor with enhanced safety.64 A phase Ib/II open study in 33 patients with active SLE despite stable background immunosuppression showed that subcutaneous zetomipzomib for 13 weeks improved disease activity, which was maintained for 12 weeks in 94% patients.65 The drug was well tolerated and no neuropathy, prolonged gastrointestinal AEs were reported. Patients with LN recruited in this study also showed improvement in proteinuria after treatment for 24 weeks.66 Unfortunately, a PBO-controlled phase IIb RCT (NCT05781750) to evaluate the efficacy and safety of zetomipzomib with SOC in LN was prematurely terminated for safety signals.
The protein cereblon is a substrate receptor of the cullin-ring ligase-4 E3 ubiquitin ligase complex, which tags polyubiquitin chains to degrade neo-substrate proteins that are disease promoting, such as transcription factors and tumor promoting proteins.67 Cereblon E3 ligase modulators (CELMoDs) such as thalidomide, lenalidomide and pomalidomide synergize with the UPS to degrade Ikaros and Aiolos, which are transcriptional factors that regulate differentiation of lymphocytes, plasma cells and pDCs.57,68 Moreover, as Ikaros and Aiolos are transcriptional suppressors of IL-2, their degradation leads to enhanced IL-2 and NK cell activity.69
Thalidomide and lenalidomide have been used to treat refractory cutaneous lupus erythematosus (CLE), particularly subacute and chronic lesions.70 Most published studies were small, retrospective and uncontrolled.70–75 Moreover, thalidomide is limited by its poor intestinal absorption and the risk of peripheral polyneuropathy, which was reported in 15–80% of patients with no clear dose relationship.70 Lenalidomide appeared to be less neurotoxic, but disease flares were common after drug discontinuation. Teratogenicity, potential cardiovascular AEs and thromboembolism,76 are major deterrent of CELMoD use in SLE.
Iberdomide is a high affinity CELMoD. In a phase IIb RCT,77 iberdomide treatment for 24 weeks in SLE patients significantly reduced the number of peripheral B cells and pDCs but increased Tregs and IL-2 production in a dose-dependent manner.78 However, reduction in type I IFN gene signature occurred only in patients with high expression at baseline. These findings are consistent with the known immunomodulatory effects of iberdomide through enhanced degradation of Ikaros and Aiolos.
In a phase IIa PBO-controlled dose-escalating RCT of iberdomide in 42 patients with active SLE,79 improvement in physicians’ global assessment (PGA) and Cutaneous Lupus Area and Severity Index Activity (CLASI-A) scores were observed with treatment. Safety was established and a subsequent phase II RCT of 288 active SLE patients confirmed efficacy of iberdomide 0.45mg compared to PBO in terms of SRI-4 response at week 24.80 Subgroup analyses revealed higher effectiveness in patients with baseline SLEDAI ≥10, high Aiolos and IFN gene signature.78,80 However, meaningful differences in most secondary end points, including CLASI-A score and joint counts, were not observed between iberdomide and PBO. AEs and SAEs were non-significantly higher in iberdomide-treated patients, with neutropenia and infections being the commonest. No peripheral neuropathy was reported. In patients with active CLE who participated in the above RCT,80 significantly more iberdomide-treated (0.45mg) patients achieved ≥50% CLASI-A improvement from baseline to week 24 compared with PBO for subacute and chronic but not acute CLE lesions.81 Further clinical trials are necessary to confirm the efficacy of iberdomide in SLE, particularly for different subtypes of CLE.
There are alternative ways of targeting the plasma cells. Daratumumab is a monoclonal antibody that binds to CD38 expressed on plasma cells and plasmablasts. It depletes plasma cells and is approved for upfront combination therapy of myeloma.82 Successful treatment of refractory SLE by daratumumab has been reported.83,84 However, there are no controlled trials in SLE yet. Felzartamab is another anti-CD38 monoclonal antibody that is being studied in primary membranous nephropathy.85 A phase Ib study of felzartamab in LN has been registered (NCT06064929).
Targeting B-Cell Growth Factor
BAFF Inhibition
BAFF binds to three receptors on B cell surface, namely transmembrane activator and calcium modulator ligand interactor (TACI), BCMA and BAFF-R, which is essential for B cell maturation, survival, proliferation and immunoglobulin class switching.86 BAFF mRNA and serum levels were elevated in SLE and correlated with disease activity.87,88 APRIL (a proliferation-inducing ligand), a homolog of BAFF, is also important factor for survival and activation of B cells.89 APRIL binds to TACI and BCMA receptors with a higher affinity than BAFF. Inhibition of the BAFF and/or APRIL promotes apoptosis and prevents maturation of the B cells.90
Belimumab is a monoclonal antibody against BAFF that is approved for the treatment of SLE and LN. Pivotal RCTs in non-renal SLE confirmed benefit of belimumab when added to SOC in achieving the SRI-4 response.91–94 Extension of the BLISS-52 and BLISS-72 RCTs91,92 for up to 8 years showed sustained efficacy of belimumab and stable organ damage score in 88% of these patients.95 Pooled data from RCTs confirmed superiority of belimumab to PBO when combined with SOC in the treatment of SLE96 and a GC-sparing effect.97 However, the efficacy of belimumab in reducing renal flare in patients treated for extra-renal activity appeared to be dependent on concomitant anti-malarial therapy.98 For the treatment of LN, a large RCT reported significantly higher rate of primary end point renal response (PERR) when belimumab was combined to the SOC of either MMF or low-dose CYC when compared to PBO.99 Efficacy was consistent in the Asian subpopulation100 and across patients with newly diagnosed or relapsed LN.101 Open-label extension of this study for 28 weeks showed maintenance of the treatment response without new safety signals.102 A secondary analysis of the BLISS-LN trial showed that belimumab significantly reduced the risk of kidney-related events or death, renal flares and a sustained 30% or 40% decline in eGFR versus SOC alone.103 However, kidney response to belimumab was not observed in patients with sub-epithelial deposits or a baseline uP/Cr of ≥3.0.
Tabalumab is a human monoclonal antibody against both soluble and membrane-bound BAFF.104 Two identical PBO-controlled phase III RCTs of tabalumab in SLE with moderate to severe activity were conducted.105,106 In the ILLUMINATE-1 study, the primary end point (SRI-5 rate) was not met at week 52, although significant improvement in anti-dsDNA and complements, reduction in total B cells, immunoglobulins and BAFF levels were achieved with tabalumab treatment.105 In ILLUMINATE-2 study, the primary end point (SRI-5 at week 52) was met in the 120 mg (every 2 week) tabalumab arm.106 In both RCTs, none of the secondary end points, which included time to first severe SLE flare and GC-sparing effect, supported efficacy of tabalumab. SAEs, TEAEs and mortality were not increased across the tabalumab arms, although depression and suicidal ideation was more common. A pooling of data of the two RCTs did not reveal significant benefit of tabalumab on various renal parameters.107 Although the primary end point could have been met if SRI-4 had been used, further development of the drug was not pursued.
Blisibimod is a fusion protein comprising four BAFF binding domains linked to the N-terminus of the fragment crystallizable region (Fc) of a human antibody.108 Following a phase II RCT showing efficacy of blisibimod at week 24 in terms of the SRI-5 response and a significant improvement in SLE serology and reduction in B cell count,109 a phase III PBO-controlled RCT were performed (CHABLIS-SC1).110 This study recruited 442 SLE patients with high disease activity despite GCs and SOC. Unfortunately, the primary outcome, SRI-6 response at week 52, was not met. Exploratory end points that included SRI-4 and SRI-8 were also not significantly different between the blisibimod and PBO arms, including AEs and SAEs.
Atacicept (TACI-Ig) is a fusion protein that blocks the activity of both BAFF (soluble and membrane-bound) and APRIL.111 Atacicept treatment in SLE patients reduced peripheral mature B cells and immunoglobulin levels in a dose-dependent manner.112 A phase II/III PBO-controlled RCT of atacicept in patients with active LN was prematurely terminated for safety concerns.113 Another 52-week phase II/III PBO-controlled RCT of atacicept in 461 patients with active SLE did not meet the primary outcome of reduction in the incidence of a new BILAG A or B flare.114 The atacicept 150 mg arm was terminated prematurely because of increased serious infections. Atacicept reduced immunoglobulin levels and improved lupus serology. Patients with elevated serum levels of BAFF and APRIL showed a greater response in the reduction of SLE flares.115 Another 24-week phase IIb PBO-controlled RCT (ADDRESS II) involving 306 active SLE patients was repeated and did not show an increased incidence of TEAEs or SAEs as compared to PBO.116 Although the primary end point (SRI-4 at week 24) was not met, subgroups of patients with higher disease activity at baseline showed a significantly higher SRI-4 rates than PBO. However, further studies of atacicept were not pursued.
Telitacicept is another fusion protein that inhibits the binding of BAFF and APRIL to the TACI receptor.117 A multicenter phase II RCT involving 249 patients with active SLE conducted in China met the primary end point of having a significantly higher SRI-4 response at week 48 in all the 3 doses of telitacicept studied as compared to PBO. The drug was well tolerated with no increase in the incidence of AEs or SAEs.118 Telitacicept is currently licensed for SLE treatment in mainland China.
Anti-BAFF/Anti-CD20 Combination
The rebound increase in serum BAFF level after B cell depletion promotes reconstitution of autoreactive B cells that may be associated with SLE flares.119 Administration of a BAFF inhibitor after anti-CD20 therapy may retard full B-cell repopulation and production of SLE-specific autoantibodies.120 A phase II open RCT (CALIBRATE) of 43 patients with recurrent or refractory LN showed that the addition of belimumab to the background GC/CYC/rituximab regimen caused a greater reduction in autoreactive naive B cells from baseline to week 48 compared to the no belimumab group without increasing AEs.121 However, the renal response rates were not significantly improved with belimumab/rituximab combination. Another phase II PBO-controlled RCT (Beat-Lupus) confirmed safety of rituximab and belimumab combination in 52 patients with refractory SLE manifestations (38% with LN).122 Belimumab-rituximab combination resulted in greater suppression of B-cell repopulation, anti-dsDNA levels, and reduced frequency of severe flares at week 52 compared to PBO.
In a more recent RCT (BLISS-BELIEVE) which recruited 263 patients with active SLE, combining rituximab with a background of SC belimumab while stopping concomitant immunosuppression led to a more profound drop in most B cell subsets, including memory B cells, and anti-dsDNA levels compared to PBO at week 52.123 However, there was no significant difference between the combination group and the belimumab only group in the proportion of patients who achieved disease control at this time point, defined as a SLEDAI-2K ≤2 without immunosuppressants and a prednisone-equivalent dose of ≤5 mg/day. Of concern was an increased incidence of serious infections in the biologic combination group.
As the efficacy and safety of rituximab/belimumab combination is conflicting, further works are needed to re-evaluate this approach in different patient subgroups. A phase III open-label 2-year RCT (Synbiose-2) of rituximab/belimumab in patients with severe SLE, including LN, is in progress (NCT03747159).
Dual Mechanism Anti-BAFF
Ianalumab (VAY-736) is a human, decarboxylated antibody against the BAFF receptor (BAFF-R). It eliminates B cells via a dual mechanism of actions through enhancing ADCC and induction of B cell apoptosis via blockade of the BAFF/BAFF-R interaction.124 A phase III PBO-controlled RCT of ianalumab in LN (SIRIUS-LN) is in progress (NCT05126277).
Rozibafusp alfa is a bispecific IgG2-peptide fusion designed to inhibit both BAFF and inducible T-cell costimulator ligand (ICOSL). A phase Ib RCT in rheumatoid arthritis showed safety and efficacy of rozibafusp as compared to PBO.125 However, a phase IIb RCT in SLE was terminated because of futility of the interim results (NCT04058028).
Targeting B-Cell Signalling Pathway
The BTK is expressed in most hematopoietic cells, including the B cells and terminally differentiated plasma cells.126 In B cells, BTK plays an essential role in the downstream signal pathways through the B-cell receptor127 and enhances the sensitivity of the B cells to the toll-like TLR-mediated events that include cytokine and autoantibody production.128 BTK expression was increased in peripheral blood mononuclear cells (PBMCs) of SLE patients and correlated with disease activity, including renal.129 BTK inhibitors (BTKis) do not deplete B cells and the effects of BTK blockade in other immune cell types may enhance their therapeutic effects in SLE.
Evobrutinib, a highly selective oral BTKi, was studied in a phase II RCT of patients with active SLE despite SOC.130 However, the study did not meet the primary end points of SRI-4 and SRI-6 at week 52 compared to PBO in the high disease activity subpopulation. No clinically meaningful differences in other secondary outcomes were observed either between evobrutinib and PBO, although the drug was well-tolerated. Another phase II dose-ranging study of fenebrutinib, a second-generation highly selective, reversible oral BTKi, was conducted in 260 SLE patients with moderate/severe activity who were receiving SOC.131 Although fenebrutinib reduced the BTK-dependent plasmablast RNA signature, anti-dsDNA, and IgG/IgM levels relative to PBO, the proportion of patients who achieved the SRI-4 and BICLA responses was not significantly higher in the treatment groups at week 48. However, SAEs were numerically more common in BTKi users. Orelabrutinib is a highly selective, irreversible oral BTKi that has been studied in a phase Ib/II RCT of 60 SLE patients in China.132 The primary outcome, SRI-4 response rate at week 12, was significantly higher in the treatment than PBO groups. Overall, AEs were mild to moderate and the majority of TEAEs were not severe. As the results of these three RCTs are discrepant, further phase III RCTs of the BTKis in SLE are needed.
Lipocalimab is a monoclonal antibody that binds with high affinity to block the neonatal Fc receptor (FcRn). It inhibits IgG recycling and reduces circulating IgG levels, including autoantibodies in a dose-dependent manner.133 The biologic agent is being studied in primary Sjogren’s syndrome (NCT04969812),134 rheumatoid arthritis, autoimmune hemolytic anemia, myasthenia gravis,135 chronic inflammatory demyelinating polyneuropathy and severe hemolytic disease of the fetus and newborn.136 Two studies of lipocalimab in SLE and LN have been registered (NCT04882878; NCT04883619).
Targeting T Cells
Calcineurin Inhibitors
The calcineurin inhibitors (CNIs) block T-cell activation through suppression of the calcium/calmodulin-dependent phosphatase calcineurin.137 Cyclosporin A (CSA) and tacrolimus (TAC) have long been used in the treatment of SLE and LN.138,139 TAC is preferred to CSA for SLE treatment because of fewer cosmetic, hypertensive and dyslipidemic AEs. Recent RCTs have established non-inferiority of TAC to MMF or intravenous pulse CYC as initial therapy of LN in terms of renal response at 6 months.140,141 Moreover, low-dose combination of TAC and MMF has been shown to be more effective than intravenous CYC as initial therapy of LN at 6 months.142 Over 18 months, the rate of renal flares was similar between the MMF-TAC and CYC-AZA groups but the former was associated with a lower withdrawal rate due to AEs.143 Addition of low-dose TAC to background MMF has also been shown to be effective in reducing proteinuria in refractory LN.144,145 TAC does not affect the cell counts and is relatively safe during pregnancy.146 Although long-term data on CNI nephrotoxicity are awaiting, TAC is commonly used for initial or maintenance treatment of SLE and LN in Asian countries.147,148 In the latest 2024 Asia Pacific League of Association for Rheumatology (APLAR) consensus, CNI is one of the first-line agents recommended for initial treatment of LN.149
Voclosporin is a chemical analog of CSA that has a stronger binding capacity to cyclophilin A, leading to a higher potency of calcineurin inhibition, faster elimination and less variability in plasma concentration.150 A phase III PBO-controlled RCT of voclosporin in active LN showed efficacy of voclosporin in enhancing the CRR rate at week 52 when used in conjunction with MMF and GCs.151 SAE, serious infection and mortality was not significantly different between the voclosporin and PBO groups of patients. The results of this RCT corroborates that of the multi-target regimen in China in which upfront combination of CNI (TAC) with MMF was more effective than CYC in LN.142 While the cost-effectiveness of combined MMF and voclosporin for upfront combination therapy of LN has to be further evaluated,152 it is recommended for patients at risk of renal progression,149,153 including refractory and frequently relapsing disease. Moreover, the CNIs may be more effective in ameliorating LN patients with heavy proteinuria and/or podocytopathy in kidney biopsy.154
The mTOR Inhibitors
The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that is essential for cell metabolism, growth and survival. Activation of the mTOR pathway in kidney tissues is reported in patients with active LN.155 Inhibitors of mTOR, such as sirolimus and everolimus, are newer immunosuppressive agents that are used in kidney transplantation to prevent allograft rejection to facilitate sparing of the CNIs.156 An open single-arm phase I/II study demonstrated efficacy of sirolimus in 40 patients with active SLE who were unresponsive or intolerant to SOC therapies in reducing disease activity and the dosage requirement of GCs at month 12.157 Sirolimus was well tolerated, with only mild reduction in hemoglobin, neutrophil count and HDL-cholesterol level reported. Other case series of Caucasian and Asian patients with SLE also demonstrated efficacy and tolerability of sirolimus.158,159 Meta-analysis of observational studies showed that sirolimus (1–3mg/day) treatment led to remission of SLE manifestations in 74% of patients.160 Discontinuation of sirolimus occurred in 9.3% of patients due to reversible and mild AEs.
Inhibition of B/T Cell Co-Stimulation
Abatacept (CTLA4-Ig) is a recombinant fusion protein consisting of the extracellular domain of CTLA4 and a fragment of Fc domain of human IgG1. It binds to CD80 or CD86 with a higher affinity than CD28, thus hindering the co-stimulatory signal for T cell activation.161 In a phase IIb exploratory RCT, 175 patients with active non-renal SLE were randomized to receive abatacept or PBO,162 in addition to prednisone. The primary outcome of absence of new BILAG A or B flare at month 12 was not met, although treatment difference was greatest in patients with polyarthritis. Another PBO-controlled phase II/III multicenter RCT of abatacept in active proliferative LN on a background of GC and MMF also did not meet the primary end point of CRR at week 52.163 In both RCTs, serious AEs and AEs such as herpes zoster and gastroenteritis, and withdrawal due to SAEs, were numerically more frequent in abatacept users. Another phase II RCT of active proliferative LN (ACCESS) also did not show benefit of abatacept over PBO in the CRR rate at week 24 when combined with prednisone and low-dose CYC as initial treatment.164 However, the incidence of AEs and SAEs was not significantly increased with abatacept.
Interaction between CD40 ligand (CD40L or CD154) on activated T cells and CD40 on B cells is important for maintaining B cell functions such as B cell differentiation, germinal center formation, and antibody isotype switching.165 Abnormal expression of CD40L in SLE patients might be associated with the generation of autoantibodies.166,167 An anti-CD40L antibody, BG9588, has halted development because of increased risk of thromboembolism related to platelet activation.168 Dapirolizumab pegol, a polyethylene glycol-conjugated antigen-binding (Fab0) fragment that lacks a functional Fc domain to reduce the risk of platelet activation, was developed to target the CD40L.169 A phase IIb, PBO-controlled RCT of dapirolizumab in 182 SLE patients moderate-to-severe disease activity (RISE)170 did not meet the primary end point of a dose-responsive BICLA responder rates at week 24, although the drug was well tolerated.
Targeting Dendritic Cells
The pDCs are the main producers of type I interferons (IFNs) which are important for the pathogenesis of SLE. Although pDCs constitute a small population of leukocytes in the peripheral blood, they accumulate in lesions of skin and other organs in patients with SLE.171,172 Litifilimab is a monoclonal antibody that directs against blood dendritic cell antigen-2 (BDCA2) uniquely expressed on pDCs.173 This interaction results in suppression of TLR-7/9-induced production of type I IFNs, other cytokines and chemokine by the pDCs.173,174 Litifilimab also exhibits a dual effect on pDCs by internalization of surface low-affinity Fc gamma receptor (CD32a), thus preventing stimulation of the pDCs by DNA or RNA-containing immune complexes. A phase II PBO-controlled RCT (LILAC-A) of SLE patients with active arthritis or skin lesions showed a significantly greater reduction in active joint counts in the litifilimab 450mg group compared to PBO at week 24.175 In the second part of this RCT (LILAC-B) that involved patients with active CLE, litifilimab was shown to be superior to PBO in the reduction of the CLASI-A score.176 In both studies, most of the secondary end points did not support the results of the primary end point analyses. Litifilimab was associated with increased herpes zoster and other viral infections.
Daxdilimab is an IgG1 monoclonal antibody that binds to immunoglobulin-like transcript 7 (ILT7) on the surface of pDCs, leading to their depletion through the ADCC mechanism.177 A Phase I study showed that daxdilimab profoundly reduced both circulating and tissue-resident pDCs, which was associated with reduced type I IFN activity and improvement in the CLASI-A score in patients with CLE.177 No increased viral infection was observed with daxdilimab. A phase II trial in discoid lupus is in progress (NCT05591222).
Afimetoran is an antagonist of TLR7 and 8. A phase Ib study of afimetoran in 13 patients with CLE showed efficacy of the drug in suppressing the CLASI-A score, which was coupled with a reduction in expression of the TLR7/8 and IFN pathway genes and other cytokines.178
Targeting Cytokines
Cytokines are secreted by immune cells for mutual communication and orchestration of the immune response.179 They may exhibit pro-inflammatory or anti-inflammatory properties, or both, depending on the micro-environment. Cytokine production is dysregulated in SLE, which may the primary or secondary to the imbalance of the Th1/Th2 and Th17/Treg pathways.180 A number of cytokines are over-expressed in patients with SLE, including the IFNs (IFNα, IFNγ), interleukins (IL-6/10/12/17/21/23) and BAFF.181 Targeting the cytokines by monoclonal antibodies and their downstream intracellular pathway by the Janus kinase inhibitors (JAKis) is an important approach in the treatment of SLE.
Anti-Interferon
IFNα promotes dendritic cell development, T cell activation, and autoantibody production by B cells.182 Levels of IFN-α, IFN-driven chemokines, and expression of IFN-regulated genes were elevated in SLE patients and correlated with clinical and serological disease activity and serum IL-10 levels.183–185 Moreover, gain-of-function genetic variants in the type I IFN pathway have been associated with increased susceptibility to SLE.186
Rontalizumab is a human monoclonal antibody that neutralizes 12 subtypes of IFNα but does not bind to IFNβ or IFNω.187 A phase II PBO-controlled RCT did not meet the clinical end points of BILAG improvement and SRI response at week 24 in patients with active SLE.188 Sifalimumab is another human IgG1κ monoclonal antibody that neutralizes most subtypes of IFNα. A phase II RCT in 431 SLE patients showed that the SRI-4 response rate at week 52 was superior in the 1200mg sifalimumab arm than PBO.189 Improvement in joint counts and CLASI-A skin scores was also documented with sifalimumab treatment, although no changes in anti-dsDNA or C3/4 levels were observed. AEs were not increased with treatment except herpes zoster. Despite the encouraging results, the drug was not further developed.
Anifrolumab is a human monoclonal antibody that directs against the type I IFN receptor, thus blocking the signals from all type I IFNs, including IFNα, IFNβ, IFNε, IFNκ and IFNω.190 Following a phase IIb RCT showing promising results and safety of anifrolumab,191 two pivotal phase III PBO-controlled RCTs were conducted.192,193 In the TULIP-1 study, 457 patients with active non-renal SLE were randomized to receive two doses of anifrolumab and PBO in addition to the SOC.192 At week 52, the SRI-4 response rate was similar between anifrolumab and PBO groups. However, a secondary analysis of the BICLA response and the abolition of the treatment failure criterion of new prescription of non-steroidal anti-inflammatory drugs (NSAIDs) during the trial showed benefit of anifrolumab compared to PBO. In the TULIP-2 study, a modified primary end point of BICLA at week 52 was adopted.193 Results showed a significant higher proportion of SLE patients achieved the BICLA response than PBO. Anifrolumab was subsequently approved for the treatment of moderate to severe SLE in addition to SOC.
Pooled data of the TULIP studies showed that anifrolumab treatment was associated with earlier, more frequent, more prolonged and sustained achievement of the low disease activity state (LLDAS)194 and a sustained GC tapering effect compared to PBO.195 Patient subgroups with larger treatment differences relative to PBO included those with baseline high IFN gene signature, abnormal serological markers and Asian ethnicity.196 A 3-year extension of the TULIP studies revealed sustained efficacy of anifrolumab in the reduction of SLE activity and the cumulative GC doses without new safety signals.197 However, anifrolumab treatment was associated with an increased incidence of herpes zoster and respiratory tract infections.198
Anifrolumab has also been studied in LN. A phase II PBO-controlled RCT of 147 patients with active LN did not meet the primary outcome of improvement in the geometric mean of proteinuria at week 52 with anifrolumab.199 However, secondary analysis indicated that using an intensified anifrolumab regimen (with loading doses), the CRR rate was significantly higher than that of PBO. While the incidence of SAEs was similar across all groups, herpes zoster was more common in anifrolumab users. An extension of this study for a further of 12 months demonstrated similar efficacy of anifrolumab over PBO without new safety signals.200 A phase III study of anifrolumab in LN is in progress (NCT05138133).
There are alternative ways of targeting IFNα in SLE. IFN-α-kinoid (IFN-K) is a recombinant human IFNα conjugated to an immunogenic carrier protein.201 Active immunization of IFN-K disrupts B cell tolerance and generates neutralizing antibodies against all subtypes of IFNα. A phase I/II dose-escalating RCT of IFN-K in 28 patients with active SLE showed safety and immunogenicity of the drug.202 IFN-K administration induced anti-IFNα antibodies in all subjects, with significantly higher anti-IFNα titers in IFN signature positive than negative patients. IFN-K significantly diminished the expression of IFN-induced genes, which was associated with improvement in C3 levels. A significant correlation was found between the neutralizing anti-IFNα titers and reduction in IFN scores compared to baseline.203
On the other hand, IFNα can also be antagonized by targeting the pDCs (Targeting Dendritic Cells above) or the intracellular downstream signal pathway by the JAKis (see below).
Anti-IL6
IL-6 is mainly secreted by activated macrophages and T cells and acts synergistically with the type I IFNs to activate B cells, leading to production of protective and pathogenic antibodies. Serum IL-6 levels were elevated in SLE patients and correlated with disease activity and anti-dsDNA level.204 A phase I study of tocilizumab in 16 SLE patients showed efficacy in improving disease activity, particularly arthritis, which was associated with reduction in anti-dsDNA,205 number of activated T and B cells, plasmablasts and post-switched memory B cells.206 Another anti-IL6 monoclonal antibody (sirukumab) was studied in a phase I PBO-controlled trial of 46 patients with SLE or CLE.207 Both tocilizumab and sirukumab treatment were associated with a dose-dependent reduction in neutrophil count and increase in infective complications.205,207 In a phase II proof-of-concept PBO-controlled RCT of 25 patients with refractory LN, the renal response was not found to be superior in the treatment group at week 24.208 Moreover, in the sirukumab group, SAEs developed in 48% of patients through week 40, most of which were infective complications.
Anti-IL12/23
IL12 and IL23 play an essential role in inflammation and autoimmunity.209 In patients with SLE, a disease activity-related upregulation of serum IL12, IL-23 and IL-23 receptor levels, as well as the expression of the IL12/23 shared common p40 subunit as compared to healthy subjects has been demonstrated.210–212 Ustekinumab is a fully human monoclonal antibody that inhibits the IL-12/23 p40 subunit and is approved for patients with skin psoriasis, psoriatic arthritis and inflammatory bowel disease. Despite the promising results from a phase II PBO-controlled RCT of ustekinumab in SLE,213,214 a phase III RCT of 516 patients with active SLE was prematurely terminated due futility of the efficacy end point on interim analysis.215 The AEs were consistent with the known safety profile of ustekinumab.
Guselkumab is a monoclonal antibody that targets IL-23p19 subunit. A phase II RCT in adult patients with active LN (histologic class III/IV) was prematurely terminated due to enrollment challenges.216 At week 24, no differences in the primary renal efficacy endpoint and other secondary endpoints could be demonstrated between the guselkumab and PBO group of patients.
Low-Dose IL-2
IL-2 is a cytokine produced by activated CD4+ T cells that induces growth and proliferation of the T cells, NK and B cells.217 High-dose IL-2 augments the anti-tumor effects of the cytotoxic T cells but also lead to undesirable effects due to the activation of other immune cells. However, low-dose IL-2 preferentially activates the Tregs, leading to immune suppression and tolerance.218 Binding of IL-2 to the IL-2 receptor (IL-2R) triggers several downstream signalling pathways that include the JAK-STAT, phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) and the mitogen activated protein kinase (MAPK) that increase Treg proliferation and functional activity.219 In addition, murine studies also indicated low-dose IL-2 could effectively deplete autoreactive follicular T helper cells and abolish the autoantibody response.220
A phase II PBO-controlled RCT of low-dose IL-2 in 100 SLE patients who were receiving SOC did not meet the primary outcome of SRI-4 response at week 12.221 However, efficacy of low-dose IL-2 was shown after excluding 2 centers with 100% SRI-4 rate in the PBO arms. Another RCT of 60 SLE patients in China also did not meet the same primary end point at week 12.222 However, at week 24, a significantly higher rate of SRI-4 response was observed with IL-2 treatment without increased serious infections. Another phase II PBO-controlled RCT of IL-2 (efavaleukin alfa) in patients with active non-renal SLE was prematurely terminated due to futility (NCT04680637).
The limitations of low-dose IL-2 include the short serum half-life, potential dose-dependent adverse effects, great variability in patients’ response at different dosages, as well as the lack of long-term safety data. Future study design should refine the dosage regimens, explore the prospect of combination with other novel agents, and nest the most appropriate patient subsets by biomarkers to receive IL-2 therapy.
Targeting Cytokine Downstream Pathways
Baricitinib
Baricitinib is a JAK1/2 inhibitor that has shown efficacy in a phase II RCT of SLE patients with active joint and/or skin disease.223 Both the primary efficacy endpoint (resolution of skin disease or arthritis at week 24) and secondary end points that included SRI-4 response and LLDAS were achieved in the baricitinib (4mg) group compared to PBO. Although baricitinib did not lead to a greater improvement in anti-dsDNA and C3,224 levels of IL-12 p40, IL-6, mRNA expression of STAT1-target, STAT2-target, and STAT4-target and multiple IFN responsive genes were reduced.225 Serious infections were more frequent in the baricitinib 4mg group relative to PBO. Two identical phase III RCTs of baricitinib in non-renal SLE (BRAVE 1/2) were subsequently performed.226,227 The primary end point, SRI-4 response at week 52, was met in BRAVE-1 but not in BRAVE-2. All secondary endpoints were not met in both studies. Although the musculoskeletal and mucocutaneous domains on SLEDAI and BILAG improved significantly with baricitinib 4mg in BRAVE-1, result was not reproduced in BRAVE-2. The inconsistent results of these two RCTs render the efficacy of baricitinib in SLE inconclusive.228 A long-term extension study of baricitinib in SLE is in progress (SLE-BRAVE-X).
Upadacitinib
Upadacitinib is a selective JAK1 inhibitor that has been approved for rheumatoid arthritis, psoriatic arthritis, spondyloarthritis, atopic dermatitis and inflammatory bowel disease.11 The results of a PBO-controlled phase II RCT comparing the efficacy of two doses of upadacitinib, alone or in combination with elsubrutinib, a BTK inhibitor, was recently published.229 The low-dose upadacitinib/elsubrutinib and elsubrutinib arms were discontinued after an interim analysis showing lack of efficacy. More patients achieved the primary end point of SRI-4 and GC dose ≤10mg/day at week 24 with upadacitinib (30mg/day) and high-dose upadacitinib/elsubrutinib versus PBO. Secondary end points such as the rates of SRI-4, BICLA, LLDAS at week 24 and 48, and reduction of SLE flares were also in favor of these two treatment arms. No new safety signals were observed for upadacitinib or elsubrutinib.
Deucravacitinib
Deucravacitinib is a selective Tyk-2 inhibitor that blocks the downstream signalling of IL-12, IL-23, IL-10, and the type I IFNs.230 The phase II PBO-controlled RCT (PAISLEY) of deucravacitinib in 363 patients with active SLE (skin or joint) achieved the end point of SRI-4 at week 32.231 Secondary endpoints such as SRI-4, BICLA and LLDAS at week 48 were also in favor of the deucravacitinib (3mg twice-daily) group. Moreover, significantly more patients in this group achieved a ≥50% reduction in the CLASI score and combined swollen/tender joint counts. Greater improvement in anti-dsDNA and complement levels was observed in the deucravacitinib groups across all dosages, but not PBO, which were associated with an improvement in IFN signature through 44 weeks. Deucravacitinib was well-tolerated, with no increase in infective complications, including herpes zoster, reported. Two phase III RCTs (POETYK SLE-1/2) are in progress (NCT05617677 and NCT05620407).
Targeting Other Cellular Mechanisms
Targeting the Complements
Complements play an essential role in clearance of immune complexes and apoptotic bodies in SLE. They also regulate the functions of T and B cells.232 Activation of terminal complement is associated with disease activity and organ damage in SLE, particularly LN. Eculizumab is a monoclonal antibody targeting C5 complement and blocks its cleavage. A phase I study demonstrated safety in SLE patients.233 Case series have reported efficacy of eculizumab in SLE/LN with thrombotic microangiopathy (TMA) features or part of the atypical hemolytic uremic syndrome.234,235 Avacopan is a complement 5a receptor (C5aR) antagonist approved as an adjunctive treatment for ANCA-related vasculitis. It is also being investigated for the treatment of C3 glomerulopathy, hidradenitis suppurativa and IgA nephropathy. However, no clinical trial of avacopan in SLE/LN has been registered yet. Iptacopan is an oral inhibitor against complement factor B in the alternative complement pathway. It regulates the cleavage of C3, generation of downstream effectors and the amplification of the terminal complement pathway. The drug is approved for paroxysmal nocturnal haemoglobinuria (PNH) and is being investigated in IgA nephropathy and C3 glomerulopathy.236 A phase II RCT in active LN has been registered (NCT05268289).
Targeting the S1PR1
Sphingosine 1-phosphate (S1P) is a bioactive metabolite of ceramide that binds to five G protein-coupled S1P receptors (S1PR1-S1PR5) and influences cell proliferation, survival, and migration.237 Among the S1PR isoforms, S1PR1 is expressed on leukocytes and endothelial cells and mediates lymphocyte trafficking, Treg/Th17 cell homeostasis, and vascular permeability.238 Four S1PR1 modulators (fingolimod, siponimod, ozanimod, and ponesimod) are approved for multiple sclerosis and two SIPR1 modulators (ozanimod and etrasimod) are approved inflammatory bowel disease.239 The S1PR1 modulators could be effective in reducing SLE activity by inhibiting the trafficking of autoreactive lymphocytes and Th17 cell differentiation, augmenting the number and function of the Tregs, and reducing production of autoantibodies.240 Moreover, these modulators could enhance the endothelial cell and blood-brain barrier functions, diminish adhesion molecule expression for leukocyte transmigration, and type I IFN production by pDCs in response to viral or oligonucleotides, which are potentially beneficial for renal, neuropsychiatric disease and atherosclerotic injury in SLE.241
An open phase Ib safety trial of amiselimod in 17 SLE patients with mild/moderate activity reported lymphopenia after treatment in all patients but no serious infections, cardiac toxicity or SAEs were observed.242 A proof-of-concept PBO-controlled RCT of oral cenerimod in 49 SLE patients with active mucocutaneous or musculoskeletal manifestations demonstrated efficacy in reducing serological and clinical activity at week 12 without increasing TEAEs.243 Reduction in lymphocyte count occurred in a dose-dependent manner and a small but non-clinically relevant drop of heart rate was observed in the first 6 hours of drug administration. A more recent phase II PBO-controlled dose-escalating RCT of cenerimod in 427 SLE patients with moderate/severe activity did not meet the primary end point of mSLEDAI-2K score improvement at month 6.244 However, cenerimod 4 mg reduced IFN-associated protein and gene signature biomarkers after treatment.245 Two phase III RCTs (OPUS-1/2) of cenerimod in SLE are in progress (NCT05648500, NCT05672576).
Synthetic Peptides and Immune Tolerizers
Abetimus sodium, consisting of four dsDNA epitopes conjugated to a non-immunogenic polyethylene glycol platform, crosslinks anti-dsDNA immunoglobulin receptors on B cell surface, triggering signal transduction pathways that result in apoptosis or anergy.246 However, two large phase III RCTs of SLE failed to show efficacy of this agent in reducing disease flares, although it was well tolerated and reduced anti-dsDNA levels.247,248
Edratide is a peptide based on the sequence of the first complementarity-determining (CDR1) region of a pathogenic human anti-DNA monoclonal antibody (16/6 idiotype) that downregulates pathogenic cytokines, apoptosis, IFNα gene expression, but upregulates Tregs in PBMCs of SLE patients.249,250 A 26-week phase II dose-escalating study of SLE patients with mild/moderate activity was terminated because of lack of efficacy.251
Rigerimod (Lupuzor) is a 21-mer linear peptide issued from the small nuclear ribonucleoprotein U1-70K and phosphorylated at the Ser140 position.252 It tolerizes CD4+T cells to react to self-antigens by binding to the MHC class II molecule on cell surface and through other unknown mechanisms.253 A phase IIa study demonstrated safety of rigerimod and improved clinical and serological SLE activity [122]. Another phase IIb RCT confirmed higher SRI response rate than PBO at week 12.254 However, the phase III 52-week RCT of rigerimod in SLE (NCT02504645) did not meet the primary end point.255
Laquinimod is a modulator of antigen presenting cells that directs T cells toward an anti-inflammatory phenotype through downregulation of the pro-inflammatory cytokines (IL-6/12/17/23/TNFα) but upregulation of IL-10. A phase IIa PBO-controlled RCT showed efficacy of laquinimod in improving renal function and proteinuria on a background of MMF and GCs.256 No increase in AEs and SAEs were observed in the laquinimod groups. Another study of laquinimod in lupus arthritis has been completed (NCT01085084).
Arsenic Trioxide
Arsenic trioxide (ATO) is now part of the standard treatment regimen for acute promyelocytic leukemia (PML). ATO binds to the PML-retinoic acid receptor (RAR)α fusion oncoprotein and enhances its proteasomal degradation, leading to senescence of the leukemic cells and restoration of terminal differentiation of the myeloid progenitors.257 The availability of an oral form of ATO has greatly reduced the incidence of cardiotoxicity compared to the intravenous preparation. Increasing evidence shows that ATO has anti-inflammatory properties that include modulation of Treg activation, Th1/Th2 and Th17/Treg balance, depletion of activated T cells and pDCs, and reduction of differentiation, autoantibody and cytokine production by B cells.257 A phase IIa open single-arm dose-escalating study of 11 SLE patients with refractory disease showed that 50% of patients could achieve a SRI-4 response after intravenous ATO treatment for 24 weeks.258 Neutropenia occurred in 20% of the patients. As oral ATO is more convenient and has a reduced toxicity profile, it has a great potential for the treatment of SLE.
Conclusion
Novel therapeutic agents that target different cell types, cytokines, receptors and intracellular pathways of the innate and adaptive immune systems are being developed and tested in patients with SLE. As B cells are pivotal in the production of autoantibodies, targeting B cells is one of the main strategies of SLE therapies in the past two decades. However, the clinical efficacy of B cell modulation in SLE has not been too impressive until the recent data from the newer generation anti-CD20, obinutuzumab, showing benefit in LN when combined with the SOC.35 Previous RCTs of rituximab in non-renal SLE and LN26,27 did not show efficacy although this biologic has been widely used to treat refractory SLE. Moreover, sequential or combination therapy of the anti-CD20 and anti-BAFF has not been shown to be effective in increasing the response rate in SLE and LN but carries a risk of increased infection.259 Clinical trials of anti-CD22, anti-CD19 in SLE are futile, and neither are other anti-BAFF agents such as blisibimod and tabalumab.260
The cytokine inhibitors show promise in SLE. Despite the futility of anti-IL6 and IL-12/23 trials,208,215 belimumab and anifrolumab are anti-cytokine biologics approved for SLE at this juncture. Targeted small molecules have the advantage of lower production cost, convenience of oral administration and lack of immunogenicity.21 The oral jakinibs, proteasome inhibitors and cereblon modulators have shown preliminary success in SLE. The results of phase II studies of deucravacitinib and zetomipzomib in human SLE are encouraging. Iberdomide shows favorable phase II results in SLE, in particularly CLE lesions.
The clinical and serological heterogeneity of SLE is a major factor contributing to the failure of new drug trials in SLE. With the improvement in patient stratification by multi-omic approach, adjustment of background immunosuppressive regimens and study end points, the response rate and the effect size of SLE trials could be improved.260 A new era of SLE therapies is expected in the near future and the treat-to-target approach261 is increasingly feasible in SLE. With the availability of these novel therapeutics and the improved patient selection, it is hoped that the effectiveness of SLE treatment could be improved so that patients can survive longer with improved quality of life.
Disclosure
The author reports no conflicts of interest in this work.
References
1. Tektonidou MG, Lewandowski LB, Hu J, Dasgupta A, Ward MM. Survival in adults and children with systemic lupus erythematosus: a systematic review and Bayesian meta-analysis of studies from 1950 to 2016. Ann Rheum Dis. 2017;76(12):2009–2016. doi:10.1136/annrheumdis-2017-211663
2. Mok CC, Tse SM, Chan KL, Ho LY. Effect of immunosuppressive therapies on survival of systemic lupus erythematosus: a propensity score analysis of a longitudinal cohort. Lupus. 2018;27(5):722–727. doi:10.1177/0961203317739129
3. Mok CC, Ho LY, Chan KL, Tse SM, To CH. Trend of survival of a cohort of Chinese patients with systemic lupus erythematosus over 25 years. Front Med. 2020;7:552. doi:10.3389/fmed.2020.00552
4. Mok CC. Emerging biological therapies for systemic lupus erythematosus. Expert Opin Emerg Drugs. 2014;19(2):303–322. doi:10.1517/14728214.2014.894018
5. Hoi A, Igel T, Mok CC, Arnaud L. Therapies for systemic lupus erythematosus - Authors’ reply. Lancet Lond Engl. 2024;404(10468):2163–2164. doi:10.1016/S0140-6736(24)02381-X
6. Mok CC, Lau CS. Pathogenesis of systemic lupus erythematosus. J Clin Pathol. 2003;56(7):481–490. doi:10.1136/jcp.56.7.481
7. Mok CC. Polygenic risk score: the potential role in the management of systemic lupus erythematosus. RMD Open. 2024;10(2):e004156. doi:10.1136/rmdopen-2024-004156
8. Arnaud L, Chasset F, Martin T. Immunopathogenesis of systemic lupus erythematosus: an update. Autoimmun Rev. 2024;23(10):103648. doi:10.1016/j.autrev.2024.103648
9. Zhou X, Zhou S, Li Y. An updated review on abnormal epigenetic modifications in the pathogenesis of systemic lupus erythematosus. Front Immunol. 2024;15:1501783. doi:10.3389/fimmu.2024.1501783
10. Zharkova O, Celhar T, Cravens PD, Satterthwaite AB, Fairhurst AM, Davis LS. Pathways leading to an immunological disease: systemic lupus erythematosus. Rheumatol Oxf Engl. 2017;56(suppl_1):i55–66. doi:10.1093/rheumatology/kew427
11. Mok CC. The Jakinibs in systemic lupus erythematosus: progress and prospects. Expert Opin Investig Drugs. 2019;28(1):85–92. doi:10.1080/13543784.2019.1551358
12. Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365(22):2110–2121. doi:10.1056/NEJMra1100359
13. Mahajan A, Herrmann M, Muñoz LE. Clearance deficiency and cell death pathways: a model for the pathogenesis of SLE. Front Immunol. 2016;7:35. doi:10.3389/fimmu.2016.00035
14. Fresneda Alarcon M, McLaren Z, Wright HL. Neutrophils in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus: same foe different M.O. Front Immunol. 2021;12:649693. doi:10.3389/fimmu.2021.649693
15. Leffler J, Martin M, Gullstrand B, et al. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol Baltim Md 1950. 2012;188(7):3522–3531.
16. Hakkim A, Fürnrohr BG, Amann K, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A. 2010;107(21):9813–9818. doi:10.1073/pnas.0909927107
17. Fransen JH, van der Vlag J, Ruben J, Adema GJ, Berden JH, Hilbrands LB. The role of dendritic cells in the pathogenesis of systemic lupus erythematosus. Arthritis Res Ther. 2010;12(2):207. doi:10.1186/ar2966
18. Scheinecker C, Bonelli M, Smolen JS. Pathogenetic aspects of systemic lupus erythematosus with an emphasis on regulatory T cells. J Autoimmun. 2010;35(3):269–275. doi:10.1016/j.jaut.2010.06.018
19. Fujio K, Okamura T, Sumitomo S, Yamamoto K. Regulatory cell subsets in the control of autoantibody production related to systemic autoimmunity. Ann Rheum Dis. 2013;72 Suppl 2:ii85–89.
20. Menon M, Blair PA, Isenberg DA, Mauri C. A regulatory feedback between plasmacytoid dendritic cells and regulatory B cells is aberrant in systemic lupus erythematosus. Immunity. 2016;44(3):683–697. doi:10.1016/j.immuni.2016.02.012
21. Mok CC. Targeted small molecules for systemic lupus erythematosus: drugs in the pipeline. Drugs. 2023;83(6):479–496. doi:10.1007/s40265-023-01856-x
22. Mok CC. Current role of rituximab in systemic lupus erythematosus. Int J Rheum Dis. 2015;18(2):154–163. doi:10.1111/1756-185X.12463
23. Atisha-Fregoso Y, Toz B, Diamond B. Meant to B: b cells as a therapeutic target in systemic lupus erythematosus. J Clin Invest. 2021;131(12):e149095. doi:10.1172/JCI149095
24. Gao N, Dresel J, Eckstein V, et al. Impaired suppressive capacity of activation-induced regulatory B cells in systemic lupus erythematosus. Arthritis Rheumatol Hoboken NJ. 2014;66(10):2849–2861. doi:10.1002/art.38742
25. Abeles I, Palma C, Meednu N, Payne AS, Looney RJ, Anolik JH. B cell-directed therapy in autoimmunity. Annu Rev Immunol. 2024;42(1):103–126. doi:10.1146/annurev-immunol-083122-044829
26. 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. doi:10.1002/art.27233
27. 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. doi:10.1002/art.34359
28. Mok CC. Biological and targeted therapies of systemic lupus erythematosus: evidence and the state of the art. Expert Rev Clin Immunol. 2017;13(7):677–692. doi:10.1080/1744666X.2017.1323635
29. Reddy V, Jayne D, Close D, Isenberg D. B-cell depletion in SLE: clinical and trial experience with rituximab and ocrelizumab and implications for study design. Arthritis Res Ther. 2013;15(Suppl 1):S2. doi:10.1186/ar3910
30. Mysler EF, Spindler AJ, Guzman R, et al. Efficacy and safety of ocrelizumab in active proliferative lupus nephritis: results from a randomized, double-blind, phase III study. Arthritis Rheum. 2013;65(9):2368–2379. doi:10.1002/art.38037
31. 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. doi:10.1136/annrheumdis-2021-220920
32. Mössner E, Brünker P, Moser S, et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity. Blood. 2010;115(22):4393–4402. doi:10.1182/blood-2009-06-225979
33. Marinov AD, Wang H, Bastacky SI, et al. The type II anti-CD20 antibody obinutuzumab (GA101) is more effective than rituximab at depleting b cells and treating disease in a murine lupus model. Arthritis Rheumatol Hoboken NJ. 2021;73(5):826–836. doi:10.1002/art.41608
34. Rovin BH, Furie RA, Ross Terres JA, et al. Kidney outcomes and preservation of kidney function with obinutuzumab in patients with lupus nephritis: a post hoc analysis of the NOBILITY Trial. Arthritis Rheumatol Hoboken NJ. 2023;76(2):247–54.
35. Furie RA, Rovin BH, Garg JP, et al. Efficacy and safety of obinutuzumab in active lupus nephritis. N Engl J Med. 2025;392(15):1471–1483. doi:10.1056/NEJMoa2410965
36. Condon MB, Ashby D, Pepper RJ, et al. Prospective observational single-centre cohort study to evaluate the effectiveness of treating lupus nephritis with rituximab and mycophenolate mofetil but no oral steroids. Ann Rheum Dis. 2013;72(8):1280–1286. doi:10.1136/annrheumdis-2012-202844
37. Rossi EA, Goldenberg DM, Michel R, Rossi DL, Wallace DJ, Chang CH. Trogocytosis of multiple B-cell surface markers by CD22 targeting with epratuzumab. Blood. 2013;122(17):3020–3029. doi:10.1182/blood-2012-12-473744
38. Dörner T, Shock A, Goldenberg DM, Lipsky PE. The mechanistic impact of CD22 engagement with epratuzumab on B cell function: implications for the treatment of systemic lupus erythematosus. Autoimmun Rev. 2015;14(12):1079–1086. doi:10.1016/j.autrev.2015.07.013
39. Dörner T, Kaufmann J, Wegener WA, Teoh N, Goldenberg DM, Burmester GR. Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus. Arthritis Res Ther. 2006;8(3):R74. doi:10.1186/ar1942
40. Jacobi AM, Goldenberg DM, Hiepe F, Radbruch A, Burmester GR, Dörner T. Differential effects of epratuzumab on peripheral blood B cells of patients with systemic lupus erythematosus versus normal controls. Ann Rheum Dis. 2008;67(4):450–457. doi:10.1136/ard.2007.075762
41. Wallace DJ, Kalunian K, Petri MA, et al. Efficacy and safety of epratuzumab in patients with moderate/severe active systemic lupus erythematosus: results from EMBLEM, a phase IIb, randomised, double-blind, placebo-controlled, multicentre study. Ann Rheum Dis. 2014;73(1):183–190. doi:10.1136/annrheumdis-2012-202760
42. Wallace DJ, Hobbs K, Clowse MEB, et al. Long-term safety and efficacy of epratuzumab in the treatment of moderate-to- severe systemic lupus erythematosus: results from an open-label extension study. Arthritis Care Res. 2016;68(4):534–543. doi:10.1002/acr.22694
43. Clowse MEB, Wallace DJ, Furie RA, et al. Efficacy and safety of epratuzumab in moderately to severely active systemic lupus erythematosus: results from two phase III randomized, double-blind, placebo-controlled trials. Arthritis Rheumatol Hoboken NJ. 2017;69(2):362–375. doi:10.1002/art.39856
44. Horton HM, Chu SY, Ortiz EC, et al. Antibody-mediated coengagement of FcγRIIb and B cell receptor complex suppresses humoral immunity in systemic lupus erythematosus. J Immunol Baltim Md 1950. 2011;186(7):4223–4233.
45. Merrill JT, Guthridge J, Smith M, et al. Obexelimab in systemic lupus erythematosus with exploration of response based on gene pathway co-expression patterns: a double-blind, randomized, placebo-controlled, phase 2 trial. Arthritis Rheumatol Hoboken NJ. 2023;75(12):2185–2194. doi:10.1002/art.42652
46. Viardot A, Locatelli F, Stieglmaier J, Zaman F, Jabbour E. Concepts in immuno-oncology: tackling B cell malignancies with CD19-directed bispecific T cell engager therapies. Ann Hematol. 2020;99(10):2215–2229. doi:10.1007/s00277-020-04221-0
47. Bucci L, Hagen M, Rothe T, et al. Bispecific T cell engager therapy for refractory rheumatoid arthritis. Nat Med. 2024;30(6):1593–1601. doi:10.1038/s41591-024-02964-1
48. Subklewe M, Magno G, Gebhardt C, et al. Application of blinatumomab, a bispecific anti-CD3/CD19 T-cell engager, in treating severe systemic sclerosis: a case study. Eur J Cancer Oxf Engl. 2024;204:114071. doi:10.1136/annrheumdis-2020-eular.2700
49. Shah K, Leandro M, Cragg M, et al. Disrupting B and T-cell collaboration in autoimmune disease: t-cell engagers versus CAR T-cell therapy? Clin Exp Immunol. 2024;217(1):15–30. doi:10.1093/cei/uxae031
50. Moreau P, Garfall AL, van de Donk NWCJ, et al. Teclistamab in relapsed or refractory multiple myeloma. N Engl J Med. 2022;387(6):495–505. doi:10.1056/NEJMoa2203478
51. Alexander T, Krönke J, Cheng Q, Keller U, Krönke G. Teclistamab-induced remission in refractory systemic lupus erythematosus. N Engl J Med. 2024;391(9):864–866. doi:10.1056/NEJMc2407150
52. Mishra AK, Gupta A, Dagar G, et al. CAR-T-cell therapy in multiple myeloma: b-cell maturation antigen (BCMA) and beyond. Vaccines. 2023;11(11):1721. doi:10.3390/vaccines11111721
53. Müller F, Taubmann J, Bucci L, et al. CD19 CAR T-cell therapy in autoimmune disease - a case series with follow-up. N Engl J Med. 2024;390(8):687–700. doi:10.1056/NEJMoa2308917
54. Hiepe F, Radbruch A. Plasma cells as an innovative target in autoimmune disease with renal manifestations. Nat Rev Nephrol. 2016;12(4):232–240. doi:10.1038/nrneph.2016.20
55. Merino-Vico A, Frazzei G, Van Hamburg JP, Tas SW. Targeting B cells and plasma cells in autoimmune diseases: from established treatments to novel therapeutic approaches. Eur J Immunol. 2023;53(1):e2149675. doi:10.1002/eji.202149675
56. Xi J, Zhuang R, Kong L, He R, Zhu H, Zhang J. Immunoproteasome-selective inhibitors: an overview of recent developments as potential drugs for hematologic malignancies and autoimmune diseases. Eur J Med Chem. 2019;182:111646. doi:10.1016/j.ejmech.2019.111646
57. Fuchs O. Targeting cereblon in hematologic malignancies. Blood Rev. 2022;57:100994.
58. Alexander T, Sarfert R, Klotsche J, et al. The proteasome inhibitior bortezomib depletes plasma cells and ameliorates clinical manifestations of refractory systemic lupus erythematosus. Ann Rheum Dis. 2015;74(7):1474–1478. doi:10.1136/annrheumdis-2014-206016
59. Zhang H, Liu Z, Huang L, et al. The short-term efficacy of bortezomib combined with glucocorticoids for the treatment of refractory lupus nephritis. Lupus. 2017;26(9):952–958. doi:10.1177/0961203316686703
60. Alexander T, Cheng Q, Klotsche J, et al. Proteasome inhibition with bortezomib induces a therapeutically relevant depletion of plasma cells in SLE but does not target their precursors. Eur J Immunol. 2018;48(9):1573–1579. doi:10.1002/eji.201847492
61. Segarra A, Arredondo KV, Jaramillo J, et al. Efficacy and safety of bortezomib in refractory lupus nephritis: a single-center experience. Lupus. 2020;29(2):118–125. doi:10.1177/0961203319896018
62. Walhelm T, Gunnarsson I, Heijke R, et al. Clinical experience of proteasome inhibitor bortezomib regarding efficacy and safety in severe systemic lupus erythematosus: a nationwide study. Front Immunol. 2021;12:756941. doi:10.3389/fimmu.2021.756941
63. Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP. Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Cancer Drug Targets. 2011;11(3):239–253. doi:10.2174/156800911794519752
64. Fang Y, Johnson H, Anderl JL, et al. Role of epoxide hydrolases and cytochrome P450s on metabolism of KZR-616, a First-in-class selective inhibitor of the immunoproteasome. Drug Metab Dispos Biol Fate Chem. 2021;49(9):810–821. doi:10.1124/dmd.120.000307
65. Furie R, Parikh S, Maiquez A, et al. Ab0373 treatment of Sle with the immunoproteasome inhibitor Kzr-616: results from the first 4 cohorts of the mission study, an open-label Phase 1b dose escalation trial. Ann Rheum Dis. 2020;79(Suppl 1):1486–1487.
66. Zetomipzomib (KZR-616), a first-in-class selective immunoproteasome inhibitor, demonstrated improvements in SLE/LN disease measures and biomarkers in patients with highly active SLE or nephrotic range proteinuria in the open-label phase 1b/2 MISSION study [Internet]. ACR Meeting Abstracts. Available from: https://acrabstracts.org/abstract/zetomipzomib-kzr-616-a-first-in-class-selective-immunoproteasome-inhibitor-demonstrated-improvements-in-sle-ln-disease-measures-and-biomarkers-in-patients-with-highly-active-sle-or-nephrotic-range/.
67. Richardson PG, Mateos MV, Vangsted AJ, et al. The role of E3 ubiquitin ligase in multiple myeloma: potential for cereblon E3 ligase modulators in the treatment of relapsed/refractory disease. Expert Rev Proteomics;2022. 1–12. doi:10.1080/14789450.2022.2039631
68. John LB, Ward AC. The Ikaros gene family: transcriptional regulators of hematopoiesis and immunity. Mol Immunol. 2011;48(9–10):1272–1278. doi:10.1016/j.molimm.2011.03.006
69. Gandhi AK, Kang J, Havens CG, et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4(CRBN.). Br J Haematol. 2014;164(6):811–821. doi:10.1111/bjh.12708
70. Yuki EFN, Silva CA, Aikawa NE, et al. Thalidomide and lenalidomide for refractory systemic/cutaneous lupus erythematosus treatment: a narrative review of literature for clinical practice. J Clin Rheumatol Pract Rep Rheum Musculoskelet Dis. 2021;27(6):248–259.
71. Wang X, Li M, Zeng X, et al. Lenalidomide for systemic lupus erythematosus with refractory cutaneous eruptions. Rheumatol Oxf Engl. 2023;62(10):e293–4. doi:10.1093/rheumatology/kead116
72. Malara G, Verduci C, Altomonte M, et al. Thalidomide and discoid lupus erythematosus: case series and review of literature. Drugs Context. 2022;11. doi:10.7573/dic.2021-9-8
73. Cortés-Hernández J, Ávila G, Vilardell-Tarrés M, Ordi-Ros J. Efficacy and safety of lenalidomide for refractory cutaneous lupus erythematosus. Arthritis Res Ther. 2012;14(6):R265. doi:10.1186/ar4111
74. Okon L, Rosenbach M, Krathen M, et al. Lenalidomide in treatment-refractory cutaneous lupus erythematosus: efficacy and safety in a 52-week trial. J Am Acad Dermatol. 2014;70(3):583–584. doi:10.1016/j.jaad.2013.11.007
75. Domingo S, Solé C, Moliné T, Ferrer B, Ordi-Ros J, Cortés-Hernández J. Efficacy of thalidomide in discoid lupus erythematosus: insights into the molecular mechanisms. Dermatol Basel Switz. 2020;236(5):467–476. doi:10.1159/000508672
76. Li W, Garcia D, Cornell RF, et al. Cardiovascular and thrombotic complications of novel multiple myeloma therapies: a review. JAMA Oncol. 2017;3(7):980–988. doi:10.1001/jamaoncol.2016.3350
77. Matyskiela ME, Zhang W, Man HW, et al. A cereblon modulator (CC-220) with improved degradation of ikaros and aiolos. J Med Chem. 2018;61(2):535–542. doi:10.1021/acs.jmedchem.6b01921
78. Lipsky PE, van Vollenhoven R, Dörner T, et al. Biological impact of iberdomide in patients with active systemic lupus erythematosus. Ann Rheum Dis. 2022;81(8):1136–1142. doi:10.1136/annrheumdis-2022-222212
79. Furie RA, Hough DR, Gaudy A, et al. Iberdomide in patients with systemic lupus erythematosus: a randomised, double-blind, placebo-controlled, ascending-dose, phase 2a study. Lupus Sci Med. 2022;9(1):e000581. doi:10.1136/lupus-2021-000581
80. 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. doi:10.1056/NEJMoa2106535
81. Werth VP, Merrill JT, Furie R, et al. Effect of iberdomide on cutaneous manifestations in systemic lupus erythematosus: a randomized phase 2 clinical trial. J Am Acad Dermatol. 2024;S0190-9622(24):3044–3045.
82. Facon T, Kumar SK, Plesner T, et al. Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): overall survival results from a randomised, open-label, Phase 3 trial. Lancet Oncol. 2021;22(11):1582–1596. doi:10.1016/S1470-2045(21)00466-6
83. Ostendorf L, Burns M, Durek P, et al. Targeting CD38 with daratumumab in refractory systemic lupus erythematosus. N Engl J Med. 2020;383(12):1149–1155. doi:10.1056/NEJMoa2023325
84. Roccatello D, Fenoglio R, Caniggia I, et al. Daratumumab monotherapy for refractory lupus nephritis. Nat Med. 2023;29(8):2041–2047. doi:10.1038/s41591-023-02479-1
85. Rovin BH, Ronco PM, Wetzels JFM, et al. Phase 1b/2a study assessing the safety and efficacy of felzartamab in Anti-Phospholipase A2 receptor autoantibody-positive primary membranous nephropathy. Kidney Int Rep. 9(9):2635–2647. doi:10.1016/j.ekir.2024.06.018.
86. Moore PA, Belvedere O, Orr A, et al. BLyS: member of the tumor necrosis factor family and B lymphocyte stimulator. Science. 1999;285(5425):260–263. doi:10.1126/science.285.5425.260
87. Petri M, Stohl W, Chatham W, et al. Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008;58(8):2453–2459. doi:10.1002/art.23678
88. Collins CE, Gavin AL, Migone TS, Hilbert DM, Nemazee D, Stohl W. B lymphocyte stimulator (BLyS) isoforms in systemic lupus erythematosus: disease activity correlates better with blood leukocyte BLyS mRNA levels than with plasma BLyS protein levels. Arthritis Res Ther. 2006;8(1):R6. doi:10.1186/ar1855
89. Fairfax K, Mackay IR, Mackay F. BAFF/BLyS inhibitors: a new prospect for treatment of systemic lupus erythematosus. IUBMB Life. 2012;64(7):595–602. doi:10.1002/iub.1046
90. Burness CB, McCormack PL. Belimumab: in systemic lupus erythematosus. Drugs. 2011;71(18):2435–2444. doi:10.2165/11208440-000000000-00000
91. 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 Lond Engl. 2011;377(9767):721–731. doi:10.1016/S0140-6736(10)61354-2
92. 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. doi:10.1002/art.30613
93. 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. doi:10.1136/annrheumdis-2017-211631
94. Doria A, Stohl W, Schwarting A, et al. Efficacy and safety of subcutaneous belimumab in anti-double-stranded DNA-positive, hypocomplementemic patients with systemic lupus erythematosus. Arthritis Rheumatol Hoboken NJ. 2018;70(8):1256–1264. doi:10.1002/art.40511
95. van Vollenhoven RF, Navarra SV, Levy RA, et al. Long-term safety and limited organ damage in patients with systemic lupus erythematosus treated with belimumab: a Phase III study extension. Rheumatol Oxf Engl. 2020;59(2):281–291. doi:10.1093/rheumatology/kez279
96. Wei LQ, Liang YG, Zhao Y, Liang HT, Qin DC, She MC. Efficacy and safety of belimumab plus standard therapy in patients with systemic lupus erythematosus: a meta-analysis. Clin Ther. 2016;38(5):1134–1140. doi:10.1016/j.clinthera.2016.02.022
97. van Vollenhoven RF, Petri M, Wallace DJ, et al. Cumulative corticosteroid dose over fifty-two weeks in patients with systemic lupus erythematosus: pooled analyses from the phase III belimumab trials. Arthritis Rheumatol Hoboken NJ. 2016;68(9):2184–2192. doi:10.1002/art.39682
98. Gomez A, Jägerback S, Sjöwall C, Parodis I. Belimumab and antimalarials combined against renal flares in patients treated for extra-renal systemic lupus erythematosus: results from 4 phase III clinical trials. Rheumatol Oxf Engl. 2024;63(2):338–348. doi:10.1093/rheumatology/kead253
99. Furie R, Rovin BH, Houssiau F, et al. Two-Year, Randomized, Controlled Trial of Belimumab in Lupus Nephritis. N Engl J Med. 2020;383(12):1117–1128. doi:10.1056/NEJMoa2001180
100. Yu X, Chen N, Xue J, et al. Efficacy and safety of belimumab in patients with lupus nephritis: subgroup analyses of a phase 3 randomized trial in the East Asian population. Am J Kidney Dis off J Natl Kidney Found. 2023;81(3):294–306.e1. doi:10.1053/j.ajkd.2022.06.013
101. Anders HJ, Furie R, Malvar A, et al. Effect of belimumab on kidney-related outcomes in patients with lupus nephritis: post hoc subgroup analyses of the phase 3 BLISS-LN trial. Nephrol Dial Transplant off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2023;38(12):2733–2742.
102. Furie R, Rovin BH, Houssiau F, et al. Safety and efficacy of belimumab in patients with lupus nephritis: open-label extension of BLISS-LN study. Clin J Am Soc Nephrol CJASN. 2022;17(11):1620–1630. doi:10.2215/CJN.02520322
103. Rovin BH, Furie R, Teng YKO, et al. A secondary analysis of the Belimumab international study in lupus nephritis trial examined effects of belimumab on kidney outcomes and preservation of kidney function in patients with lupus nephritis. Kidney Int. 2022;101(2):403–413. doi:10.1016/j.kint.2021.08.027
104. Stohl W, Scholz JL, Cancro MP. Targeting BLyS in rheumatic disease: the sometimes-bumpy road from bench to bedside. Curr Opin Rheumatol. 2011;23(3):305–310. doi:10.1097/BOR.0b013e328344c15e
105. Isenberg DA, Petri M, Kalunian K, et al. Efficacy and safety of subcutaneous tabalumab in patients with systemic lupus erythematosus: results from ILLUMINATE-1, a 52-week, phase III, multicentre, randomised, double-blind, placebo-controlled study. Ann Rheum Dis. 2016;75(2):323–331. doi:10.1136/annrheumdis-2015-207653
106. Merrill JT, van Vollenhoven RF, Buyon JP, et al. Efficacy and safety of subcutaneous tabalumab, a monoclonal antibody to B-cell activating factor, in patients with systemic lupus erythematosus: results from ILLUMINATE-2, a 52-week, phase III, multicentre, randomised, double-blind, placebo-controlled study. Ann Rheum Dis. 2016;75(2):332–340. doi:10.1136/annrheumdis-2015-207654
107. Rovin BH, Dooley MA, Radhakrishnan J, Ginzler EM, Forrester TD, Anderson PW. The impact of tabalumab on the kidney in systemic lupus erythematosus: results from two phase 3 randomized, clinical trials. Lupus. 2016;25(14):1597–1601. doi:10.1177/0961203316650734
108. Stohl W, Merrill JT, Looney RJ, et al. Treatment of systemic lupus erythematosus patients with the BAFF antagonist “peptibody” blisibimod (AMG 623/A-623): results from randomized, double-blind phase 1a and phase 1b trials. Arthritis Res Ther. 2015;17(1):215. doi:10.1186/s13075-015-0741-z
109. Furie RA, Leon G, Thomas M, et al. A phase 2, randomised, placebo-controlled clinical trial of blisibimod, an inhibitor of B cell activating factor, in patients with moderate-to-severe systemic lupus erythematosus, the PEARL-SC study. Ann Rheum Dis. 2015;74(9):1667–1675. doi:10.1136/annrheumdis-2013-205144
110. Merrill JT, Shanahan WR, Scheinberg M, et al. Phase III trial results with blisibimod, a selective inhibitor of B-cell activating factor, in subjects with systemic lupus erythematosus (SLE): results from a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2018;77(6):883–889. doi:10.1136/annrheumdis-2018-213032
111. Stohl W. Systemic lupus erythematosus and its ABCs (April/BLyS complexes). Arthritis Res Ther. 2010;12(2):111. doi:10.1186/ar2976
112. Dall’Era M, Chakravarty E, Wallace D, et al. Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial. Arthritis Rheum. 2007;56(12):4142–4150. doi:10.1002/art.23047
113. 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. doi:10.1186/ar3738
114. 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. doi:10.1136/annrheumdis-2013-205067
115. Gordon C, Wofsy D, Wax S, Li Y, Pena Rossi C, Isenberg D. Post hoc analysis of the phase II/III April-SLE study: association between response to atacicept and serum biomarkers including BLyS and APRIL. Arthritis Rheumatol Hoboken NJ. 2017;69(1):122–130. doi:10.1002/art.39809
116. Merrill JT, Wallace DJ, Wax S, et al. Efficacy and safety of atacicept in patients with systemic lupus erythematosus: results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled, parallel-arm, phase IIb study. Arthritis Rheumatol Hoboken NJ. 2018;70(2):266–276. doi:10.1002/art.40360
117. Yao X, Ren Y, Zhao Q, et al. Pharmacokinetics analysis based on target-mediated drug distribution for RC18, a novel BLyS/April fusion protein to treat systemic lupus erythematosus and rheumatoid arthritis. Eur J Pharm Sci off J Eur Fed Pharm Sci. 2021;159:105704.
118. Wu D, Li J, Xu D, et al. Telitacicept in patients with active systemic lupus erythematosus: results of a phase 2b, randomised, double-blind, placebo-controlled trial. Ann Rheum Dis. 2024;83(4):475–487. doi:10.1136/ard-2023-224854
119. Carter LM, Isenberg DA, Ehrenstein MR. Elevated serum BAFF levels are associated with rising anti-double-stranded DNA antibody levels and disease flare following B cell depletion therapy in systemic lupus erythematosus. Arthritis Rheum. 2013;65(10):2672–2679. doi:10.1002/art.38074
120. Kraaij T, Arends EJ, van Dam LS, et al. Long-term effects of combined B-cell immunomodulation with rituximab and belimumab in severe, refractory systemic lupus erythematosus: 2-year results. Nephrol Dial Transplant off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2021;36(8):1474–1483.
121. Atisha-Fregoso Y, Malkiel S, Harris KM, et al. Phase II randomized trial of rituximab plus cyclophosphamide followed by belimumab for the treatment of lupus nephritis. Arthritis Rheumatol Hoboken NJ. 2021;73(1):121–131. doi:10.1002/art.41466
122. Shipa M, Embleton-Thirsk A, Parvaz M, et al. Effectiveness of belimumab after rituximab in systemic lupus erythematosus: a randomized controlled trial. Ann Intern Med. 2021;174(12):1647–1657. doi:10.7326/M21-2078
123. Aranow C, Allaart CF, Amoura Z, et al. Efficacy and safety of sequential therapy with subcutaneous belimumab and one cycle of rituximab in patients with systemic lupus erythematosus: the phase 3, randomised, placebo-controlled BLISS-BELIEVE study. Ann Rheum Dis. 2024;83(11):1502–1512. doi:10.1136/ard-2024-225686
124. Mavragani CP, Moutsopoulos HM. Sjögren’s syndrome: old and new therapeutic targets. J Autoimmun. 2020;110:102364. doi:10.1016/j.jaut.2019.102364
125. Abuqayyas L, Cheng LE, Teixeira Dos Santos M, et al. Safety and biological activity of rozibafusp alfa, a bispecific inhibitor of inducible costimulator ligand and B cell activating factor, in patients with rheumatoid arthritis: results of a phase 1b, randomized, double-blind, placebo-controlled, multiple ascending dose study. ACR Open Rheumatol. 2022;4(10):903–911. doi:10.1002/acr2.11487
126. Neys SFH, Rip J, Hendriks RW, Corneth OBJ. Bruton’s tyrosine kinase inhibition as an emerging therapy in systemic autoimmune disease. Drugs. 2021;81(14):1605–1626. doi:10.1007/s40265-021-01592-0
127. Zhang D, Gong H, Meng F. Recent advances in BTK inhibitors for the treatment of inflammatory and autoimmune diseases. Mol. 2021;26(16):4907.
128. Rip J, de Bruijn MJW, Appelman MK, Pal Singh S, Hendriks RW, Corneth OBJ. Toll-like receptor signaling drives btk-mediated autoimmune disease. Front Immunol. 2019;10:95. doi:10.3389/fimmu.2019.00095
129. Kong W, Deng W, Sun Y, et al. Increased expression of Bruton’s tyrosine kinase in peripheral blood is associated with lupus nephritis. Clin Rheumatol. 2018;37(1):43–49. doi:10.1007/s10067-017-3717-3
130. Wallace DJ, Dörner T, Pisetsky DS, et al. Efficacy and safety of the bruton’s tyrosine kinase inhibitor evobrutinib in systemic lupus erythematosus: results of a phase ii, randomized, double-blind, placebo-controlled dose-ranging trial. ACR Open Rheumatol. 2023;5(1):38–48. doi:10.1002/acr2.11511
131. Isenberg D, Furie R, Jones NS, et al. Efficacy, safety, and pharmacodynamic effects of the bruton’s tyrosine kinase inhibitor fenebrutinib (GDC-0853) in systemic lupus erythematosus: results of a phase II, randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol Hoboken NJ. 2021;73(10):1835–1846. doi:10.1002/art.41811
132. Li R, Zhu X, Liu S, et al. Lb0005 orelabrutinib, an irreversible inhibitor of bruton’s tyrosine kinase (btk), for the treatment of systemic lupus erythematosus (sle): results of a randomized, double-blind, placebo-controlled, phase Ib/Iia dose-finding study. Ann Rheum Dis. 2022;81(Suppl 1):210. doi:10.1136/annrheumdis-2022-eular.5086a
133. Leu JH, Vermeulen A, Abbes C, Arroyo S, Denney WS, Ling LE. Pharmacokinetics and pharmacodynamics across infusion rates of intravenously administered nipocalimab: results of a phase 1, placebo-controlled study. Front Neurosci. 2024;18:1302714. doi:10.3389/fnins.2024.1302714
134. Gottenberg JE, Sivils K, Campbell K, et al. LBA0010 efficacy and safety of nipocalimab, an anti-fcrn monoclonal antibody. Ann Rheum Dis. 2024;83(Suppl 1):240.
135. Antozzi C, Vu T, Ramchandren S, et al. Safety and efficacy of nipocalimab in adults with generalised myasthenia gravis (Vivacity-MG3): a phase 3, randomised, double-blind, placebo-controlled study. Lancet Neurol. 2025;24(2):105–116. doi:10.1016/S1474-4422(24)00498-8
136. Jacobs JW, Booth GS, Raza S, et al. Current state and potential applications of neonatal Fc receptor (FcRn) inhibitors in hematologic conditions. Am J Hematol. 2024;99(12):2351–2366. doi:10.1002/ajh.27487
137. Mok CC. Calcineurin inhibitors in systemic lupus erythematosus. Best Pract Res Clin Rheumatol. 2017;31(3):429–438. doi:10.1016/j.berh.2017.09.010
138. Mok CCP. The use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transplant off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2016;31(10):1561–1566.
139. Mok CC, Wong RWS, Lai KN. Treatment of severe proliferative lupus nephritis: the current state. Ann Rheum Dis. 2003;62(9):799–804. doi:10.1136/ard.62.9.799
140. Mok CC, Ho LY, Ying SKY, Leung MC, To CH, Ng WL. Long-term outcome of a randomised controlled trial comparing tacrolimus with mycophenolate mofetil as induction therapy for active lupus nephritis. Ann Rheum Dis. 2020;79(8):1070–1076. doi:10.1136/annrheumdis-2020-217178
141. Zheng Z, Zhang H, Peng X, et al. Effect of tacrolimus vs intravenous cyclophosphamide on complete or partial response in patients with lupus nephritis: a randomized clinical trial. JAMA Network Open. 2022;5(3):e224492. doi:10.1001/jamanetworkopen.2022.4492
142. Liu Z, Zhang H, Liu Z, et al. Multitarget therapy for induction treatment of lupus nephritis: a randomized trial. Ann Intern Med. 2015;162(1):18–26. doi:10.7326/M14-1030
143. Zhang H, Liu Z, Zhou M, et al. Multitarget therapy for maintenance treatment of lupus nephritis. J Am Soc Nephrol JASN. 2017;28(12):3671–3678. doi:10.1681/ASN.2017030263
144. Mok CC, To CH, Yu KL, Ho LY. Combined low-dose mycophenolate mofetil and tacrolimus for lupus nephritis with suboptimal response to standard therapy: a 12-month prospective study. Lupus. 2013;22(11):1135–1141. doi:10.1177/0961203313502864
145. Lanata CM, Mahmood T, Fine DM, Petri M. Combination therapy of mycophenolate mofetil and tacrolimus in lupus nephritis. Lupus. 2010;19(8):935–940. doi:10.1177/0961203310365714
146. Webster P, Wardle A, Bramham K, Webster L, Nelson-Piercy C, Lightstone L. Tacrolimus is an effective treatment for lupus nephritis in pregnancy. Lupus. 2014;23(11):1192–1196. doi:10.1177/0961203314540353
147. Mok CC. Towards new avenues in the management of lupus glomerulonephritis. Nat Rev Rheumatol. 2016;12(4):221–234. doi:10.1038/nrrheum.2015.174
148. Takeuchi T, Wakasugi N, Hashida T, Uno S, Makino H. Long-term safety and effectiveness of tacrolimus in patients with lupus nephritis in japan: 10-year analysis of the real-world TRUST study. J Rheumatol. 2024;51(6):613–621. doi:10.3899/jrheum.2023-0210
149. Mok CC, So H, Hamijoyo L, et al. The 2024 APLAR consensus on the management of lupus nephritis. Int J Rheum Dis. 2025;28(1):e70021. doi:10.1111/1756-185X.70021
150. van Gelder T, Lerma E, Engelke K, Huizinga RB. Voclosporin: a novel calcineurin inhibitor for the treatment of lupus nephritis. Expert Rev Clin Pharmacol. 2022;15(5):515–529. doi:10.1080/17512433.2022.2092470
151. 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 Lond Engl. 2021;397(10289):2070–2080. doi:10.1016/S0140-6736(21)00578-X
152. Mok CC. A new old treatment for lupus nephritis. Lancet Lond Engl. 2021;397(10289):2027–2029. doi:10.1016/S0140-6736(21)00663-2
153. Rovin BH, Ayoub IM, Chan TM, et al. Executive summary of the KDIGO 2024 clinical practice guideline for the management of lupus nephritis. Kidney Int. 2024;105(1):31–34. doi:10.1016/j.kint.2023.09.001
154. Fanouriakis A, Kostopoulou M, Cheema K, et al. 2019 update of the joint European league against rheumatism and European renal association-European dialysis and transplant association (EULAR/ERA-EDTA) recommendations for the management of lupus nephritis. Ann Rheum Dis. 2020;79(6):713–723. doi:10.1136/annrheumdis-2020-216924
155. Mao Z, Tan Y, Tao J, et al. Renal mTORC1 activation is associated with disease activity and prognosis in lupus nephritis. Rheumatol Oxf Engl. 2022;61(9):3830–3840. doi:10.1093/rheumatology/keac037
156. Ma MKM, Yung S, Chan TM. mTOR Inhibition and Kidney Diseases. Transplantation. 2018;102(2S Suppl 1):S32–40. doi:10.1097/TP.0000000000001729
157. Lai ZW, Kelly R, Winans T, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet Lond Engl. 2018;391(10126):1186–1196. doi:10.1016/S0140-6736(18)30485-9
158. Eriksson P, Wallin P, Sjöwall C. Clinical experience of sirolimus regarding efficacy and safety in systemic lupus erythematosus. Front Pharmacol. 2019;10:82. doi:10.3389/fphar.2019.00082
159. Peng L, Wu C, Hong R, et al. Clinical efficacy and safety of sirolimus in systemic lupus erythematosus: a real-world study and meta-analysis. Ther Adv Musculoskelet Dis. 2020;12:1759720X20953336. doi:10.1177/1759720X20953336
160. Ji L, Xie W, Zhang Z. Efficacy and safety of sirolimus in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Semin Arthritis Rheum. 2020;50(5):1073–1080. doi:10.1016/j.semarthrit.2020.07.006
161. Mok CC. Abatacept for systemic lupus erythematosus: the outlook. Expert Opin Biol Ther. 2012;12(12):1559–1561. doi:10.1517/14712598.2012.721773
162. 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. doi:10.1002/art.27601
163. 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 Hoboken NJ. 2014;66(2):379–389. doi:10.1002/art.38260
164. ACCESS Trial Group. Treatment of lupus nephritis with abatacept: the abatacept and cyclophosphamide combination efficacy and safety study. Arthritis Rheumatol Hoboken NJ. 2014;66(11):3096–3104. doi:10.1002/art.38790
165. Yazdany J, Davis J. The role of CD40 ligand in systemic lupus erythematosus. Lupus. 2004;13(5):377–380. doi:10.1191/0961203304lu1030oa
166. Manea ME, Mueller RB, Dejica D, et al. Increased expression of CD154 and FAS in SLE patients’ lymphocytes. Rheumatol Int. 2009;30(2):181–185. doi:10.1007/s00296-009-0933-4
167. Ramanujam M, Steffgen J, Visvanathan S, Mohan C, Fine JS, 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(11):102668. doi:10.1016/j.autrev.2020.102668
168. Boumpas DT, Furie R, Manzi S, et al. A short course of BG9588 (anti-CD40 ligand antibody) improves serologic activity and decreases hematuria in patients with proliferative lupus glomerulonephritis. Arthritis Rheum. 2003;48(3):719–727. doi:10.1002/art.10856
169. Tocoian A, Buchan P, Kirby H, et al. First-in-human trial of the safety, pharmacokinetics and immunogenicity of a PEGylated anti-CD40L antibody fragment (CDP7657) in healthy individuals and patients with systemic lupus erythematosus. Lupus. 2015;24(10):1045–1056. doi:10.1177/0961203315574558
170. Furie RA, Bruce IN, Dörner T, et al. Phase 2, randomized, placebo-controlled trial of dapirolizumab pegol in patients with moderate-to-severe active systemic lupus erythematosus. Rheumatol Oxf Engl. 2021;60(11):5397–5407. doi:10.1093/rheumatology/keab381
171. Tomasini D, Mentzel T, Hantschke M, et al. Plasmacytoid dendritic cells: an overview of their presence and distribution in different inflammatory skin diseases, with special emphasis on Jessner’s lymphocytic infiltrate of the skin and cutaneous lupus erythematosus. J Cutan Pathol. 2010;37(11):1132–1139. doi:10.1111/j.1600-0560.2010.01587.x
172. Tucci M, Quatraro C, Lombardi L, Pellegrino C, Dammacco F, Silvestris F. Glomerular accumulation of plasmacytoid dendritic cells in active lupus nephritis: role of interleukin-18. Arthritis Rheum. 2008;58(1):251–262. doi:10.1002/art.23186
173. Cho YM, Furie R. The development of litifilimab (BIIB 059) for cutaneous and systemic lupus erythematosus. Immunotherapy. 2024;16(1):15–20. doi:10.2217/imt-2023-0086
174. Liu J, Zhang X, Cao X. Dendritic cells in systemic lupus erythematosus: from pathogenesis to therapeutic applications. J Autoimmun. 2022;132:102856. doi:10.1016/j.jaut.2022.102856
175. 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. doi:10.1056/NEJMoa2118025
176. Werth VP, Furie RA, Romero-Diaz J, et al. Trial of Anti-BDCA2 antibody litifilimab for cutaneous lupus erythematosus. N Engl J Med. 2022;387(4):321–331. doi:10.1056/NEJMoa2118024
177. Karnell JL, Wu Y, Mittereder N, et al. Depleting plasmacytoid dendritic cells reduces local type I interferon responses and disease activity in patients with cutaneous lupus. Sci Transl Med. 2021;13(595):eabf8442. doi:10.1126/scitranslmed.abf8442
178. Baribaud F, Saini J, Ignatenko S, et al. POS0551 pharmacodynamic effects and exploratory efficacy of afimetoran, A TLR7/8 inhibitor, in patients with cutaneous lupus erythematosus. Ann Rheum Dis. 2024;83:1110–1111. doi:10.1136/annrheumdis-2024-eular.1540
179. Apostolidis SA, Lieberman LA, Kis-Toth K, Crispín JC, Tsokos GC. The dysregulation of cytokine networks in systemic lupus erythematosus. J Interferon Cytokine Res off J Int Soc Interferon Cytokine Res. 2011;31(10):769–779. doi:10.1089/jir.2011.0029
180. Jacob N, Stohl W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res Ther. 2011;13(4):228. doi:10.1186/ar3349
181. Su DL, Lu ZM, Shen MN, Li X, Sun LY. Roles of pro- and anti-inflammatory cytokines in the pathogenesis of SLE. J Biomed Biotechnol. 2012;2012:347141. doi:10.1155/2012/347141
182. Rönnblom L, Alm GV. Systemic lupus erythematosus and the type I interferon system. Arthritis Res Ther. 2003;5(2):68–75. doi:10.1186/ar625
183. Dall’era MC, Cardarelli PM, Preston BT, Witte A, Davis JC. Type I interferon correlates with serological and clinical manifestations of SLE. Ann Rheum Dis. 2005;64(12):1692–1697. doi:10.1136/ard.2004.033753
184. Nikpour M, Dempsey AA, Urowitz MB, Gladman DD, Barnes DA. Association of a gene expression profile from whole blood with disease activity in systemic lupus erythaematosus. Ann Rheum Dis. 2008;67(8):1069–1075. doi:10.1136/ard.2007.074765
185. Feng X, Wu H, Grossman JM, et al. Association of increased interferon-inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum. 2006;54(9):2951–2962. doi:10.1002/art.22044
186. Postal M, Vivaldo JF, Fernandez-Ruiz R, Paredes JL, Appenzeller S, Niewold TB. Type I interferon in the pathogenesis of systemic lupus erythematosus. Curr Opin Immunol. 2020;67:87–94. doi:10.1016/j.coi.2020.10.014
187. McBride JM, Jiang J, Abbas AR, et al. Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a Phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis Rheum. 2012;64(11):3666–3676. doi:10.1002/art.34632
188. Kalunian KC, Merrill JT, Maciuca R, et al. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-α) in patients with systemic lupus erythematosus (ROSE). Ann Rheum Dis. 2016;75(1):196–202. doi:10.1136/annrheumdis-2014-206090
189. Khamashta M, Merrill JT, Werth VP, et al. Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis. 2016;75(11):1909–1916. doi:10.1136/annrheumdis-2015-208562
190. Kalunian KC. Interferon-targeted therapy in systemic lupus erythematosus: is this an alternative to targeting B and T cells? Lupus. 2016;25(10):1097–1101. doi:10.1177/0961203316652495
191. 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 Hoboken NJ. 2017;69(2):376–386. doi:10.1002/art.39962
192. 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):e208–19. doi:10.1016/S2665-9913(19)30076-1
193. 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. doi:10.1056/NEJMoa1912196
194. Morand EF, Abreu G, Furie RA, Golder V, Tummala R. Lupus low disease activity state attainment in the phase 3 TULIP trials of anifrolumab in active systemic lupus erythematosus. Ann Rheum Dis. 2023;82(5):639–645. doi:10.1136/ard-2022-222748
195. Bruce IN, van Vollenhoven RF, Morand EF, et al. Sustained glucocorticoid tapering in the phase 3 trials of anifrolumab: a post hoc analysis of the TULIP-1 and TULIP-2 trials. Rheumatol Oxf Engl. 2023;62(4):1526–1534. doi:10.1093/rheumatology/keac491
196. Vital EM, Merrill JT, Morand EF, et al. Anifrolumab efficacy and safety by type I interferon gene signature and clinical subgroups in patients with SLE: post hoc analysis of pooled data from two phase III trials. Ann Rheum Dis. 2022;81(7):951–961. doi:10.1136/annrheumdis-2021-221425
197. 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 Hoboken NJ. 2023;75(2):253–265. doi:10.1002/art.42392
198. Tummala R, Abreu G, Pineda L, et al. Safety profile of anifrolumab in patients with active SLE: an integrated analysis of phase II and III trials. Lupus Sci Med. 2021;8(1):e000464. doi:10.1136/lupus-2020-000464
199. 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. doi:10.1136/annrheumdis-2021-221478
200. Jayne D, Rovin B, Mysler E, et al. Anifrolumab in lupus nephritis: results from second-year extension of a randomised phase II trial. Lupus Sci Med. 2023;10(2):e000910. doi:10.1136/lupus-2023-000910
201. Mathian A, Amoura Z, Adam E, et al. Active immunisation of human interferon α transgenic mice with a human interferon α Kinoid induces antibodies that neutralise interferon α in sera from patients with systemic lupus erythematosus. Ann Rheum Dis. 2011;70(6):1138–1143. doi:10.1136/ard.2010.141101
202. Lauwerys BR, Hachulla E, Spertini F, et al. Down-regulation of interferon signature in systemic lupus erythematosus patients by active immunization with interferon α-kinoid. Arthritis Rheum. 2013;65(2):447–456. doi:10.1002/art.37785
203. Ducreux J, Houssiau FA, Vandepapelière P, et al. Interferon α kinoid induces neutralizing anti-interferon α antibodies that decrease the expression of interferon-induced and B cell activation associated transcripts: analysis of extended follow-up data from the interferon α kinoid phase I/II study. Rheumatol Oxf Engl. 2016;55(10):1901–1905. doi:10.1093/rheumatology/kew262
204. Chun HY, Chung JW, Kim HA, et al. Cytokine IL-6 and IL-10 as biomarkers in systemic lupus erythematosus. J Clin Immunol. 2007;27(5):461–466. doi:10.1007/s10875-007-9104-0
205. Illei GG, Shirota Y, Yarboro CH, et al. 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(2):542–552. doi:10.1002/art.27221
206. Shirota Y, Yarboro C, Fischer R, Pham TH, Lipsky P, Illei GG. Impact of anti-interleukin-6 receptor blockade on circulating T and B cell subsets in patients with systemic lupus erythematosus. Ann Rheum Dis. 2013;72(1):118–128. doi:10.1136/annrheumdis-2012-201310
207. Szepietowski JC, Nilganuwong S, Wozniacka A, 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(10):2661–2671. doi:10.1002/art.38091
208. Rovin BH, van Vollenhoven RF, Aranow C, et al. A multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of treatment with sirukumab (CNTO 136) in patients with active lupus nephritis. Arthritis Rheumatol Hoboken NJ. 2016;68(9):2174–2183. doi:10.1002/art.39722
209. Kreymborg K, Böhlmann U, Becher B. IL-23: changing the verdict on IL-12 function in inflammation and autoimmunity. Expert Opin Ther Targets. 2005;9(6):1123–1136. doi:10.1517/14728222.9.6.1123
210. Dai H, He F, Tsokos GC, Kyttaris VC. IL-23 limits the production of IL-2 and promotes autoimmunity in lupus. J Immunol Baltim Md 1950. 2017;199(3):903–910.
211. Qiu F, Song L, Yang N, Li X. Glucocorticoid downregulates expression of IL-12 family cytokines in systemic lupus erythematosus patients. Lupus. 2013;22(10):1011–1016. doi:10.1177/0961203313498799
212. Huang X, Hua J, Shen N, Chen S. Dysregulated expression of interleukin-23 and interleukin-12 subunits in systemic lupus erythematosus patients. Mod Rheumatol. 2007;17(3):220–223. doi:10.3109/s10165-007-0568-9
213. van Vollenhoven RF, Hahn BH, Tsokos GC, et al. Maintenance of efficacy and safety of ustekinumab through one year in a phase II multicenter, prospective, randomized, double-blind, placebo-controlled crossover trial of patients with active systemic lupus erythematosus. Arthritis Rheumatol Hoboken NJ. 2020;72(5):761–768. doi:10.1002/art.41179
214. 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 Lond Engl. 2018;392(10155):1330–1339. doi:10.1016/S0140-6736(18)32167-6
215. van Vollenhoven 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. doi:10.1136/ard-2022-222858
216. Anders HJ, Chan TM, Sanchez-Guerrero J, et al. Efficacy and safety of guselkumab in patients with active lupus nephritis: results from a phase 2, randomized, placebo-controlled study. Rheumatol Oxf Engl. 2025;64(5):2731–2740. doi:10.1093/rheumatology/keae647
217. Zhang R, Zhao Y, Chen X, Zhuang Z, Li X, Shen E. Low-dose IL-2 therapy in autoimmune diseases: an update review. Int Rev Immunol. 2024;43(3):113–137. doi:10.1080/08830185.2023.2274574
218. Su QY, Luo J, Wang XM, Di JK, Cao YX, Zhang SX. Efficacy, safety and the lymphocyte subsets changes of low-dose IL-2 in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Immun Inflamm Dis. 2024;12(1):e1165. doi:10.1002/iid3.1165
219. Farooq A, Trehan S, Singh G, et al. A comprehensive review of low-dose interleukin-2 (IL-2) therapy for systemic lupus erythematosus: mechanisms, efficacy, and clinical applications. Cureus. 2024;16(9):e68748. doi:10.7759/cureus.68748
220. Santana S, Papillion A, Foote JB, et al. Cutting edge: low-dose recombinant IL-2 treatment prevents autoantibody responses in systemic lupus erythematosus via regulatory T cell-independent depletion of T follicular helper cells. J Immunol Baltim Md 1950. 2024;213(8):1053–1060.
221. Humrich JY, Cacoub P, Rosenzwajg M, 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(12):1685–1694. doi:10.1136/ard-2022-222501
222. He J, Zhang R, Shao M, 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(1):141–149. doi:10.1136/annrheumdis-2019-215396
223. Wallace DJ, Furie RA, Tanaka Y, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Lond Engl. 2018;392(10143):222–231. doi:10.1016/S0140-6736(18)31363-1
224. Dörner T, van Vollenhoven RF, Doria A, et al. Baricitinib decreases anti-dsDNA in patients with systemic lupus erythematosus: results from a phase II double-blind, randomized, placebo-controlled trial. Arthritis Res Ther. 2022;24(1):112. doi:10.1186/s13075-022-02794-x
225. Dörner T, Tanaka Y, Petri MA, et al. Baricitinib-associated changes in global gene expression during a 24-week phase II clinical systemic lupus erythematosus trial implicates a mechanism of action through multiple immune-related pathways. Lupus Sci Med. 2020;7(1):e000424. doi:10.1136/lupus-2020-000424
226. Morand EF, Vital EM, Petri M, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-I). Lancet Lond Engl. 2023;401(10381):1001–1010. doi:10.1016/S0140-6736(22)02607-1
227. Petri M, Bruce IN, Dörner T, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-II). Lancet Lond Engl. 2023;401(10381):1011–1019. doi:10.1016/S0140-6736(22)02546-6
228. Mok CC. Outlook of the jakinibs in systemic lupus erythematous after baricitinib failed. Int J Rheum Dis. 2024;27(2):e15082. doi:10.1111/1756-185X.15082
229. Merrill JT, Tanaka Y, D’Cruz D, et al. Efficacy and safety of upadacitinib or elsubrutinib alone or in combination for patients with systemic lupus erythematosus: a phase 2 randomized controlled trial. Arthritis Rheumatol Hoboken NJ. 2024;76(10):1518–1529.
230. Ramakrishna C, Mason A, Edwards CJ. Tyrosine kinase 2 inhibitors in autoimmune diseases. Autoimmun Rev. 2024;23(11):103649. doi:10.1016/j.autrev.2024.103649
231. 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 Hoboken NJ. 2023;75(2):242–252. doi:10.1002/art.42391
232. Barilla-Labarca ML, Toder K, Furie R. Targeting the complement system in systemic lupus erythematosus and other diseases. Clin Immunol Orlando Fla. 2013;148(3):313–321. doi:10.1016/j.clim.2013.02.014
233. Rother RP, Mojcik CF, McCroskery EW. Inhibition of terminal complement: a novel therapeutic approach for the treatment of systemic lupus erythematosus. Lupus. 2004;13(5):328–334. doi:10.1191/0961203303lu1021oa
234. Wright RD, Bannerman F, Beresford MW, Oni L. A systematic review of the role of eculizumab in systemic lupus erythematosus-associated thrombotic microangiopathy. BMC Nephrol. 2020;21(1):245. doi:10.1186/s12882-020-01888-5
235. El-Husseini A, Hannan S, Awad A, Jennings S, Cornea V, Sawaya BP. Thrombotic microangiopathy in systemic lupus erythematosus: efficacy of eculizumab. Am J Kidney Dis off J Natl Kidney Found. 2015;65(1):127–130. doi:10.1053/j.ajkd.2014.07.031
236. Syed YY. Iptacopan: first Approval. Drugs. 2024;84(5):599–606. doi:10.1007/s40265-024-02009-4
237. Kitsou K, Kokkotis G, Rivera-Nieves J, Bamias G. Targeting the sphingosine-1-phosphate pathway: new opportunities in inflammatory bowel disease management. Drugs. 2024;84(10):1179–1197. doi:10.1007/s40265-024-02094-5
238. Pérez-Jeldres T, Alvarez-Lobos M, Rivera-Nieves J. Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis. Drugs. 2021;81(9):985–1002. doi:10.1007/s40265-021-01528-8
239. Tsai HC, Han MH. Sphingosine-1-phosphate (S1P) and S1P signaling pathway: therapeutic targets in autoimmunity and inflammation. Drugs. 2016;76(11):1067–1079. doi:10.1007/s40265-016-0603-2
240. Chen H, Wang J, Zhang C, et al. Sphingosine 1-phosphate receptor, a new therapeutic direction in different diseases. Biomed Pharmacother Biomedecine Pharmacother. 2022;153:113341. doi:10.1016/j.biopha.2022.113341
241. Burg N, Salmon JE, Hla T. Sphingosine 1-phosphate receptor-targeted therapeutics in rheumatic diseases. Nat Rev Rheumatol. 2022;18(6):335–351. doi:10.1038/s41584-022-00784-6
242. Tanaka Y, Kondo K, Ichibori A, et al. Amiselimod, a sphingosine 1-phosphate receptor-1 modulator, for systemic lupus erythematosus: a multicenter, open-label exploratory study. Lupus. 2020;29(14):1902–1913. doi:10.1177/0961203320966385
243. Hermann V, Batalov A, Smakotina S, Juif PE, Cornelisse P. First use of cenerimod, a selective S1P1 receptor modulator, for the treatment of SLE: a double-blind, randomised, placebo-controlled, proof-of-concept study. Lupus Sci Med. 2019;6(1):e000354. doi:10.1136/lupus-2019-000354
244. Askanase AD, D’Cruz D, Kalunian K, et al. Cenerimod, a sphingosine-1-phosphate receptor modulator, versus placebo in patients with moderate-to-severe systemic lupus erythematosus (CARE): an international, double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Rheumatol. 2025;7(1):e21–32. doi:10.1016/S2665-9913(24)00246-7
245. Suffiotti M, Brazauskas P, Keller MP, et al. Pharmacodynamics of the S1P1 receptor modulator cenerimod in a phase 2b randomised clinical trial in patients with moderate to severe SLE. Ann Rheum Dis. 2025;84(2):284–293. doi:10.1136/ard-2024-226547
246. Furie RA, Cash JM, Cronin ME, et al. Treatment of systemic lupus erythematosus with LJP 394. J Rheumatol. 2001;28(2):257–265.
247. Cardiel MH, Tumlin JA, Furie RA, et al. Abetimus sodium for renal flare in systemic lupus erythematosus: results of a randomized, controlled phase III trial. Arthritis Rheum. 2008;58(8):2470–2480. doi:10.1002/art.23673
248. Horowitz DM, Furie RA. Abetimus sodium: a medication for the prevention of lupus nephritis flares. Expert Opin Pharmacother. 2009;10(9):1501–1507. doi:10.1517/14656560902946419
249. Sthoeger ZM, Sharabi A, Dayan M, et al. The tolerogenic peptide hCDR1 downregulates pathogenic cytokines and apoptosis and upregulates immunosuppressive molecules and regulatory T cells in peripheral blood mononuclear cells of lupus patients. Hum Immunol. 2009;70(3):139–145. doi:10.1016/j.humimm.2009.01.018
250. Sthoeger Z, Zinger H, Sharabi A, Asher I, Mozes E. The tolerogenic peptide, hCDR1, down-regulates the expression of interferon-α in murine and human systemic lupus erythematosus. PLoS One. 2013;8(3):e60394. doi:10.1371/journal.pone.0060394
251. Urowitz MB, Isenberg DA, Wallace DJ. Safety and efficacy of hCDR1 (Edratide) in patients with active systemic lupus erythematosus: results of phase II study. Lupus Sci Med. 2015;2(1):e000104. doi:10.1136/lupus-2015-000104
252. Schall N, Page N, Macri C, Chaloin O, Briand JP, Muller S. Peptide-based approaches to treat lupus and other autoimmune diseases. J Autoimmun. 2012;39(3):143–153. doi:10.1016/j.jaut.2012.05.016
253. Monneaux F, Parietti V, Briand JP, Muller S. Importance of spliceosomal RNP1 motif for intermolecular T-B cell spreading and tolerance restoration in lupus. Arthritis Res Ther. 2007;9(5):R111. doi:10.1186/ar2317
254. Zimmer R, Scherbarth HR, Rillo OL, Gomez-Reino JJ, Muller S. Lupuzor/P140 peptide in patients with systemic lupus erythematosus: a randomised, double-blind, placebo-controlled phase IIb clinical trial. Ann Rheum Dis. 2013;72(11):1830–1835. doi:10.1136/annrheumdis-2012-202460
255. Lorenzo-Vizcaya A, Isenberg DA. Clinical trials in systemic lupus erythematosus: the dilemma-Why have phase III trials failed to confirm the promising results of phase II trials? Ann Rheum Dis. 2023;82(2):169–174. doi:10.1136/ard-2022-222839
256. Jayne D, Appel G, Chan TM, Barkay H, Weiss R, Wofsy D. LB0003 A randomized controlled study of laquinimod in active lupus nephritis patients in combination with standard of care. Ann Rheum Dis. 2013;72:A164. doi:10.1136/annrheumdis-2013-eular.528
257. Mok TC, Mok CC. The potential use of arsenic trioxide in the treatment of systemic lupus erythematosus. Int J Mol Sci. 2024;25(17):9577. doi:10.3390/ijms25179577
258. Hamidou M, Néel A, Poupon J, et al. Safety and efficacy of low-dose intravenous arsenic trioxide in systemic lupus erythematosus: an open-label phase IIa trial (Lupsenic). Arthritis Res Ther. 2021;23(1):70. doi:10.1186/s13075-021-02454-6
259. Mok CC. Combination strategies for lupus nephritis: facts and controversies. Expert Rev Clin Immunol. 2023;19(5):527–536. doi:10.1080/1744666X.2023.2192927
260. Mok CC. The dawn of a new era of therapies in systemic lupus erythematosus. Rheumatol Immunol Res. 2020;1(1):31–37. doi:10.2478/rir-2020-0005
261. Mok CC. Is treat-to-target in lupus nephritis realistic in clinical practice? Curr Rheumatol Rev. 2019;15(1):2–6. doi:10.2174/1573397114666180406100857
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles
Spotlight on New Therapeutic Opportunities for MYC-Driven Cancers
D'Avola A, Kluckova K, Finch AJ, Riches JC
OncoTargets and Therapy 2023, 16:371-383
Published Date: 7 June 2023

Emerging Therapies for Ulcerative Colitis: Updates from Recent Clinical Trials
AlAmeel T, AlMutairdi A, Al-Bawardy B
Clinical and Experimental Gastroenterology 2023, 16:147-167
Published Date: 17 August 2023

Management Strategies for Pediatric Moderate-to-Severe Plaque Psoriasis: Spotlight on Biologics
Ruggiero A, Portarapillo A, Megna M, Patruno C, Napolitano M
Pediatric Health, Medicine and Therapeutics 2023, 14:435-451
Published Date: 14 November 2023