Skip to main content
PLOS One logoLink to PLOS One
. 2025 Apr 30;20(4):e0320179. doi: 10.1371/journal.pone.0320179

Clinical outcomes of baricitinib in patients with systemic lupus erythematosus: Pooled analysis of SLE-BRAVE-I and SLE-BRAVE-II trials

Juntao Yin 1,#, Yantao Hou 2,#, Chaoyang Wang 3,*,#, Changjiang Qin 3,*,#
Editor: Vipula Rasanga Bataduwaarachchi4
PMCID: PMC12043178  PMID: 40305472

Abstract

Background

Baricitinib is an oral selective inhibitor of Janus kinase 1/2 that has achieved clinically meaningful outcomes for systemic lupus erythematosus (SLE). Only two phase 3 randomized clinical trials (SLE-BRAVE-I and SLE-BRAVE-II) have been completed, but their conclusions were inconsistent. We aimed to determine the efficacy and safety of once-daily oral baricitinib 4 mg or 2 mg treatment versus placebo in participants with active SLE.

Methods

SLE-BRAVE-I and SLE-BRAVE-II are two multicenter, placebo-controlled, double-blind, phase 3 randomized clinical trials with follow-up to 52 weeks. At baseline, 1535 patients (aged ≥18 years) with active SLE (excluding those with central nervous system and severe active renal disease) receiving stable background therapy were randomly assigned 1:1:1 to baricitinib 4 mg, baricitinib 2 mg, or placebo once daily for 52 weeks. The primary endpoint was the proportion of patients with an SLE Responder Index (SRI)-4 response at week 52 in the baricitinib 4 mg treatment group compared with placebo.

Results

There was no difference in the primary efficacy outcome of the proportion of SRI-4 responders at week 52 between participants who received baricitinib 4mg (263 [52%]; odds ratio (OR) 1.27 [95% CI 0.99, 1.63]), 2 mg (246 [48%]; 1.09 [0.85, 1.39]) and placebo (232 [46%]). However, based on SLE-BRAVE-I and SLE-BRAVE-II studies, baricitinib 4 mg and baricitinib 2 mg were more effective in reducing the disease activity in participants who treated with a glucocorticoid dose at baseline of 10 mg per day or higher of prednisone or equivalent, and baricitinib 4 mg was more effective in reducing the disease activity in participants with highly active disease (SLEDAI-2K score at baseline >10). None of the major secondary endpoints, including time to first severe flare and glucocorticoid tapering, were met. Compared with placebo, baricitinib 4 mg and baricitinib 2 mg in the treatment of SLE did not increase the incidence of treatment-emergent adverse events (TEAEs).

Conclusions

Pooled analysis showed that once-daily oral baricitinib 4 mg or baricitinib 2 mg in addition to SOC did not reduce the overall disease activity compared with placebo although they were safe for SLE. However, baricitinib might be a potential treatment option for SLE in certain subpopulation, such as in participants who received a glucocorticoid dose at baseline of 10 mg per day or higher of prednisone or equivalent (baricitinib 4 mg and baricitinib 2 mg) and in participants with highly active disease (SLEDAI-2K score at baseline >10) (baricitinib 4 mg).

Trial registration number

ClinicalTrials. gov Registry (NCT03616912 and NCT03616964).

Introduction

Systematic lupus erythematosus (SLE) is a multisystem chronic autoimmune disease with extremely various clinical manifestations, which is characterized by systemic inflammation, excessive production of autoantibodies and aberrant activities of the immune system [1,2].

Different from rheumatoid arthritis (RA) and other diseases [3], the main outcomes of SLE, including mortality, did not improve in the first twenty years of this century [4]. One quarter to one third of patients with SLE show poor disease control [5]. With significant morbidity and mortality, SLE is still one of the main causes of death among young women aged 15–45 [4,6]. The positive results of the phase 3 trials of belimumab and anivolumab [7] have promoted regulatory approval for these drugs and supported the involvement of humoral and innate immunity in the pathogenesis of SLE. Many other studies, however, have yielded negative results, and high medical demand for treatment of SLE still cannot be met [8]. Routine treatment of SLE includes antimalarials (hydroxychloroquine), glucocorticoids (prednisone, methylprednisolone) and immunosuppressants (azathioprine, cyclophosphamide, mycophenolate mofetil, methotrexate, rituximab) [9]. However, the control of disease activity in many patients remains unimproved may result in end-organ damage [5]. In order to meet the medical needs, SLE clinical drug development has never stopped, however, in the past 60 years, only two kinds of new treatment have been approved [10].

Many cytokines, such as interferons (IFNs), B-cell activating factor, interleukin (IL)-6, IL-12, IL-17, IL-23, and tumor necrosis factor (TNF), are associated with the etiology of SLE [1113]. Moreover, these cytokines activate intracellular signaling pathways by activating Janus kinases (JAKs) [1,14,15]. JAK inhibitors, a new category of drugs, can effectively treat a variety of autoimmune diseases [16]. Baricitinib, a selective JAK1/2 inhibitor, is approved for active RA, alopecia areata (AA), and atopic dermatitis (AD) [17].

A phase Ⅱ study showed that at week 24, once-daily oral baricitinib 4 mg plus standard of care (SOC) was superior to SOC in improving the disease activity of SLE [18]. In addition, baricitinib significantly downregulated expression of anti-dsDNA antibodies and key cytokines among seropositive patients [13,19,20].

The baricitinib phase Ⅲ program in severe SLE included two trials, SLE-BRAVE-I [21] and II [22], in which active SLE participants (aged ≥18 years) were enrolled. However, the conclusions of SLE-BRAVE-I and II were inconsistent in the primary outcome. The primary outcome of SLE-BRAVE-I demonstrated that a significantly higher proportion of participants who received baricitinib 4 mg (142 [57%]; odds ratio (OR) 1.57 [95% CI 1.09, 2.27]), but not baricitinib 2 mg (126 [50%]; OR 1.14; 95% CI 0.79, 1.65), reached SLE Responder Index (SRI)-4 response compared with placebo; while the primary outcome of SLE-BRAVE-Ⅱ demonstrated that no significant difference was observed in the proportion of SRI-4 responders at week 52 between participants who received baricitinib 4mg (121 [47%]; OR 1.07; 95% CI 0.75, 1.53]), 2 mg (120 [46%]; OR 1.05; 95% CI 0.73, 1.50) and placebo (116 [46%]). SRI-4 is a composite responder index based on improvement in disease activity (at least 4 point improvement in SLEDAI-2K score) and without the development of substantial disease activity in new organ systems (no new British Isles Lupus Assessment Group (BILAG) A or > 1 new BILAG B. BILAG A means that the disease is active and requiring treatment, and BILAG B means that the activity has improved and there may be some residual or mild symptoms) or worsening of the overall condition (no worsening in Physician Global Assessment, defined as an increase of ≥ 0.3 points [10 mm] from baseline). Therefore, a pooled analysis of the two available phase Ⅲ randomized controlled trials (RCTs) is needed to investigate the efficacy and safety of baricitinib 4 mg or 2 mg compared with placebo in participants with active SLE.

Materials and methods

Pooled data are included from the available phase 3 cohorts of SLE-BRAVE-I [21] and II [22], two double-blind RCTs followed up for 52 weeks (data cutoffs of November 1, 2020, and August 26, 2020, respectively). Overall, 1535 patients with SLE (met at least 4 of eleven American College of Rheumatology (ACR) SLE classification criteria, excluding those with central nervous system and severe active renal disease) [23,24] were enrolled in SLE-BRAVE-I and II. At baseline participants with SLE were randomly assigned to receive baricitinib 4 mg, baricitinib 2 mg, or placebo in a 1:1:1 ratio.

Both the SLE-BRAVE-I and II studies were performed based on ethical principles of the Declaration of Helsinki and Good Clinical Practice guidelines, and approval was obtained from the authorized institutional review boards at all study sites. All patients provided written informed consent. Each participating center has obtained ethical approval.

The same study design (S1 Fig in Supporting information file) and data collection between the 2 studies allowed us to pool data from all studies to increase statistical power. CONSORT (Consolidated Standards of Reporting Trials) reporting guidelines was followed in this study.

The primary endpoint was the proportion of participants reaching a SRI-4 response at week 52 in those receiving baricitinib 4 mg plus SOC compared with placebo plus SOC. Major secondary endpoints included the proportion of participants reaching a SRI-4 response at week 24 in those who received baricitinib 4 mg or 2 mg compared with SOC, proportion of patients who reached a lupus low disease activity state (LLDAS) [25] response at week 52, proportion of patients who received ≥7.5 mg prednisone (or equivalent) at baseline able to decrease dose by ≥ 25% to a prednisone equivalent dose of ≤ 7.5 mg per day maintained between week 40 and week 52, time to first severe fl....are over 52 weeks, change from baseline in Worst Pain Numeric Rating Scale (NRS) at week 52, change from baseline in Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue total score at week 52, and proportion of patients in the baricitinib 2 mg group with an SRI-4 response at week 52. At each study visit, adverse events (AEs) reports were collected. Events were coded according to the Medical Dictionary for Regulatory Activities (version 24.0). AEs of special interest included infections (including tuberculosis, herpes zoster, or opportunistic infections), major adverse cardiovascular events (MACEs), malignancies, hepatic events, and venous thromboembolic events (VTEs). The efficacy measures are detailed in S1 Table.

Descriptive statistics are summarized using nonresponder imputation for missing data. Secondary censoring rule rules out data collected after permanent withdrawal of study drug. Statistical analyses were conducted with SAS (version 9.4 or higher, SAS Institute).

Results

Of the 1535 patients enrolled in SLE-BRAVE-I and II, 510, 516, and 509 patients were randomized to once-daily baricitinib 4 mg plus SOC, baricitinib 2 mg plus SOC, or placebo plus SOC at baseline, respectively. 1190 participants completed the 52-week study (Fig 1) and 345 dropped out of the study during this period. Twelve (0.8%) participants received no dose of any study treatment, so they were excluded from the analyses of efficacy and safety. Missing data (98 (19%) of participants from the baricitinib 4 mg group, 79 (15%) participants from the baricitinib 2 mg group, and 83 (16%) participants from the placebo group) were excluded.

Fig 1. Patient inclusion flow chart.

Fig 1

Baseline demographics of each group are shown in Table 1. 1444 (94%) participants were female. At baseline, the mean age was 42.8 (standard deviation (SD) 12.7) years. The median duration was 6.80 (IQR 2.75–12.64) years and mean duration of SLE was 8.9 (7.9) years at baseline. Mean SLEDAI-2K score of participants was 10.1 (SD 3.1) at baseline. At baseline, the majority (1200 [78%]) of participants were receiving glucocorticoids with a mean prednisone dose (or equivalent) of 9.7 (SD 5.8) mg per day, 1243 (81%) were receiving anti-malarials, and 849 (55%) were receiving immuno-suppressants.

Table 1. Baseline demographics and clinical characteristics.

Placebo (n = 509) Baricitinib 2 mg (n = 516) Baricitinib 4 mg (n = 510)
Mean age, years 42.7 (12.8) 42.8 (12.7) 41.9 (12.5)
Mean time since onset of SLE, years 9.2 (7.9) 8.9 (7.7) 8.6 (8.0)
Sex
 Female 478 (94%) 484 (94%) 482 (95%)
 Male 31 (6%) 32 (6%) 28 (5%)
Race 502 508 502
 America Indian or Alaska Native 26 (5%) 27 (5%) 20 (4%)
 Asian 104 (20%) 105 (20%) 104 (20%)
 Black or African American 53 (10%) 46 (9%) 56 (11%)
 Native Hawaiian or other Pacific Islander 0 0 1 (0.2%)
 White 313 (61%) 324 (63%) 317 (62%)
 Multiple 6 (1%) 6 (1%) 4 (1%)
Ethnicity (USA only) 97 99 99
 Hispanic or Latino 13 (13%) 24 (24%) 19 (19%)
 Not Hispanic or Latino 82 (85%) 73 (74%) 78 (79%)
 Not reported 2 (2%) 2 (2%) 2 (2%)
Region 509 516 510
 North America 97 (19%) 99 (19%) 99 (19%)
 Asia 62 (12%) 65 (13%) 62 (12%)
 Europe 119 (23%) 121 (23%) 120 (24%)
 Central America, South America, and Mexico 133 (26%) 131 (25%) 129 (25%)
 Rest of world 98 (19%) 100 (19%) 100 (20%)
Disease characteristics
 Antinuclear antibodies titer ≥1:80 454 (93%) of 487 456 (91%) of 500 465 (94%) of 496
 Mean anti-dsDNA, IU/mL 82.73 (324.62) 106.4 (509.17) 122.14 (578.4)
 Anti-dsDNA ≥ 15 IU/mL 195 (38%) 220 (43%) 197 (39%)
 Mean C3, g/L 1.06 (0.33) 1.06 (0.31) 1.07 (0.33)
 C3 < 90.0 mg/dL 169 (33%) 164 (32%) 159 (31%)
 Mean C4, g/L 0.19 (0.10) 0.18 (0.10) 0.19 (0.10)
 C4 < 10.0 mg/dL 103 (20) 108 (21%) 110 (22%)
 Mean urine protein:creatinine ratio, mg/mmol 23.46 (35.76) 23.71 (27.41) 22.57 (32.79)
 Urine protein:creatinine ratio <50 mg/mmol 467 (92%) 462 (90%) 469 (92%)
 Urine protein:creatinine ratio ≥50 mg/mmol 40 (8%) 54 (10%) 41 (8%)
 Mean eGFR, mL per min per 1.73m² 94.7 (27.19) 94.02 (25.2) 98.15 (26.19)
Concomitant medications
 Glucocorticoids 402 (79%) 404 (78%) 394 (77%)
 Mean prednisone dose (or equivalent), mg/day 9.3 (5.0) 10.0 (6.5) 9.9 (5.8)
 Prednisone dose (or equivalent) ≥10 mg/day 213 (42%) 215 (42%) 209 (41%)
 Antimalarials 422 (83%) 402 (78%) 419 (82%)
 Immunosuppressants 290 (57%) 285 (55%) 274 (54%)
 Methotrexate 109 (21%) 103 (20%) 112 (22%)
 Azathioprine 85 (17%) 102 (20%) 75 (15%)
 Mycophenolate mofetil 68 (13%) 64 (12%) 59 (12%)
 Non-steroidal anti-inflammatory drug 114 (22%) 131 (25%) 138 (27%)
Mean SLEDAI-2K score 10.1 (3.1) 10.2 (3.2) 10 (3.0)
SLEDAI-2K score ≥10 291 (57%) 301 (58%) 290 (57%)
SLEDAI-2K organ system involvement
 CNS 0 0 0
 Vascular 27 (5%) 26 (5%) 24 (5%)
 Musculoskeleta 499 (98%) 503 (97%) 498 (98%)
 Rena 33 (6%) 46 (9%) 39 (8%)
 Mucocutaneous 487 (96%) 498 (97%) 492 (96%)
 Cardiovascular and respiratory 14 (3%) 12 (2%) 17 (3%)
 Immunological 272 (53%) 274 (53%) 269 (53%)
 Constitutional 12 (2%) 13 (3%) 13 (3%)
 Haematological 33 (6%) 32 (6%) 38 (7%)
≥1 BILAG A scores 326 (64%) 361 (70%) 326 (64%)
≥2 BILAG B scores 154 (30%) 133 (26%) 161 (32%)
Mean Physician’s Global Assessment score
Mean CLASI activity score 6.6 (6.1) 6.6 (6.8) 6.4 (5.7)
Mean tender joint count 10.3 (7.1) 10.3 (6.7) 10.7 (7.1)
Mean swollen joint count 6.75 (5.5) 6.77 (5.1) 6.8 (5.2)
Mean SLICC/ACR Damage Index score 0.63 (1.1) 0.64 (1.1) 0.6 (1.0)

Data are mean (SD) or n (%); Asia; included China and Taiwan; Central America, South America, and Mexico included Brazil and Mexico; Europe included Austria, Belgium, Croatia, Czech Republic, Germany, Greece, Hungary, Switzerland, the Netherlands, and the UK; North America uncluded the USA; and rest of world included Australia, Israel, and Russia; BILAG, British Isles Lupus Assessment Group; CLASI, Cutaneous Lupus Erythematosus Disease Area and Severity Index; eGFR, estimated glomerular filtration rate; SLE, systemic lupus erythematosus; SLEDAI-2K, Systemic Lupus Erythematosus Disease Activity Index 2000; SLICC/ACR, Systemic International Collaborating Clinics/American College of Lupus Rheumatology.

At week 52, there was no significant difference between participants given baricitinib 4 mg (263 [52%]; 1.27; 0.99, 1.63) or baricitinib 2 mg (246 [48%]; 1.09; 0.85, 1.39), and placebo (232 [46%]) in SRI-4 response (Table 2). SRI-4 responses at all assessed timepoints are shown in Fig 2. However, in the subgroup of participants who had glucocorticoid ≥ 10mg/day at baseline, significant difference was found in the SRI-4 response between participants receiving baricitinib 4 mg (154 [74%]; 2.32; 1.50, 3.61) or baricitinib 2 mg (152 [71%]; 1.90; 1.23, 2.95) and placebo, at week 52 (Fig 3). There was significant difference between participants

Table 2. Primary and key secondary outcomes in the intention-to-treat population at week 52.

Placebo (n = 509) Baricitinib 2 mg (n = 516) Baricitinib 2 mg odds ratio (95% CI); difference with placebo (95% CI); p value Baricitinib 4 mg (n = 510) Baricitinib 4 mg odds ratio (95% CI); difference with placebo (95% CI); p value
Primary outcome
SRI-4*,, 232 (46%) 246 (48%) 1.09 (0.85, 1.39); 0.50 263 (52%) 1.27 (0.99, 1.63); 0.56
Reduction of ≥ 4 points from baseline in SLEDAI-2K score*,, 233 (46%) 252 (49%) 1.13 (0.88, 1.45); 0.33 269 (53%) 1.32 (1.03, 1.69); 0.26
No new BILAG A and no more than one new BILAG B disease activity score*,, 378 (74%) 391 (76%) 1.08 (0.82, 1.44); 0.58 388 (76%) 1.10 (0.83, 1.46); 0.50
No worsening (defined as an increase of ≥ 0.3 points [10 mm] from baseline) in the PGA*, 381 (75%) 394 (76%) 1.08 (0.82, 1.44); 0.58 388 (76%) 1.07 (0.80, 1.42); 0.65
Major secondary outcomes n (%)
SRI-4 (week 24)*,, 197 (39%) 218 (42%) 1.16 (0.90, 1.49); 0.25 225 (44%) 1.25 (0.97, 1.61); 0.79
Participants with ≥1 severe flare 64 (13%) 63 (12%) 0.97 (0.67, 1.40); 0.86 55 (11%) 0.84 (0.57, 1.23); 0.37
Glucocorticoid sparing*,,§ 69/221 (31%) 65/220 (30%) 0.87 (0.55, 1.36); 0.70 72/211 (34%) 1.29 (0.82, 2.05); 0.52
LLDAS*,, 125 (25%) 127 (25%) 1.00 (0.75, 1.33); 0.98 139 (27%) 1.15 (0.87, 1.52); 0.33
Other secondary outcomes
BICLA 216 (42%) 228 (44%) 1.07 (0.84, 1.37); 0.57 240 (47%) 1.21 (0.94, 1.54); 0.14
SLEDAI-2K remission of arthritis or rash 251 (49%) 257 (50%) 1.02 (0.80, 1.30); 0.87 280 (55%) 1.25 (0.98, 1.60); 0.74
≥50% reduction in CLASI activity score*,,** 63/108 (58%) 54/97 (56%) 0.96 (0.75, 1.22); 0.70 53/93 (57%) 0.95 (0.54, 1.66); 0.85

Data are n (%), n/N (%), or least squares mean (SE), unless stated otherwise; Nominal, non-multiplicity-controlled p values are shown for all outcomes; BILAG: British Isles Lupus Assessment Group; CLASI: Cutaneous Lupus Erythematosus Disease Area and Severity Index; FACIT-Fatigue: Functional Assessment of Chronic Illness Therapy-Fatigue Scale; LLDAS, lupus low disease activity state; NA, not available; NRS: Numeric Rating Scale; PGA, Physician Global Assessment; SLE, Systemic Lupus Erythematosus; SLEDAI-2K: Systemic Lupus Erythematosus Disease Activity Index 2000; SRI: Systemic Lupus Erythematosus Responder Index;

*The number of patients is not the observed value but is estimated from the responder proportion of the multiple imputation results multiplied by the number of patients in the modified intention-to-treat population;

†Comparison is odds ratio;

‡Comparison is hazard ratio;

§Proportion of patients receiving >7·5 mg prednisone (or equivalent) at baseline able, decrease dose by ≥ 25%, a prednisone equivalent dose of ≤ 7·5 mg/day maintained between week 40 and week 52;

¶Comparison is least squares mean difference; ||Patients with CLASI,tal activity score ≥10 at baseline who achieved ≥50% reduction; **Comparison is difference vs placebo.

Fig 2. SRI-4 response through 52 weeks in overall study population.

Fig 2

Proportion of patients reaching an SRI-4 response over the 52-week study period; SRI-4: Systemic Lupus Erythematosus Responder Index-4; *P ≤ 0.05; †P ≤ 0.01.

Fig 3. Proportion of patients achieving an SRI-4 response who had glucocorticoid .

Fig 3

 10mg/day at baseline. I- SRI-4: Systemic Lupus Erythematosus Responder Index-4; *P ≤ 0.05; †P ≤ 0.01.

treated with baricitinib 4 mg and placebo at week 52 (1.83; 1.27, 2.64; P < 0.01) in the subgroup of participants with a SLEDAI-2K score of 10 or more at baseline (Fig 4). For this subpopulation, at multiple timepoints besides week 52, the proportion of participants receiving baricitinib 4 mg achieved SRI-4 response was higher than that treated with placebo.

Fig 4. Proportion of patients achieving an SRI-4 response who had SLEDAI-2K .

Fig 4

 10 at baseline. SLEDAI-2K: Systemic Lupus Erythematosus Disease Activity Index-2000; SRI-4: Systemic Lupus Erythematosus Responder Index-4; *P ≤ 0.05; †P ≤ 0.01.

Given the fact that the primary endpoint has not been reached, major secondary outcomes were not considered to be of multiplicity-controlled significance, whatever their nominal p value were. No significant difference was found between baricitinib 4 mg or 2 mg and placebo in any non-multiplicity-controlled secondary outcomes or exploratory outcomes. For the subpopulation who were able to reduce the dose of prednisone or equivalent to 7.5 mg or less per day before week 52, the proportion of participants in the baricitinib 4 mg group was 34% (n = 72) (1.06; 0.71, 1.59) and in the baricitinib 2 mg group was 30% (n = 65) (0.92; 0.62, 1.39) compared with 31% (n = 69) in the placebo group. Significant difference was observed in median time to first severe flare between participants treated with baricitinib 4 mg group and placebo (2.25; 1.38, 3.64), but no significant difference between participants treated with baricitinib 2 mg and placebo (1.11; 0.64, 1.91). At week 52, the proportions of participants with LLDAS responses were 27% (n = 139) in the baricitinib 4 mg group (1.15; 0.87, 1.52) and 25% (n = 127) in the baricitinib 2 mg group (1.00; 0.75, 1.33), compared with 25% (n = 125) in the placebo group. Compared with placebo, baricitinib 4 mg or baricitinib 2 mg showed significant difference in changes from baseline in Worst Pain NRS and FACIT-Fatigue total score at week 52 (Table 2). Other secondary outcomes are shown in Table 2.

At week 52, in non-multiplicity-controlled analyses of secondary outcomes, no improvements in any organ domains of interest were observed. However, improvements in the musculoskeletal SLEDAI-2K domain were seen in both the baricitinib 4 mg and baricitinib 2 mg groups at multiple timepoints (Fig 5A). No significant difference in the mucocutaneous SLEDAI-2K domain were seen in baricitinib 4 mg or baricitinib 2 mg group at any timepoint (Fig 5B). As a post-hoc analysis, the improvements in the musculoskeletal and mucocutaneous domains were also evaluated for the BILAG organ system. Improvements in the musculoskeletal domains were observed in the baricitinib 4 mg treatment group at multiple timepoints; and improvements in the mucocutaneous domains were also observed both in the baricitinib 4 mg and baricitinib 2 mg groups at multiple timepoints (Fig 5C and 5D).

Fig 5. Improvement from baseline in SLEDAI-2K and BILAG organ systems through 52 weeks.

Fig 5

(A) Im(A) Improvement from baseline in the SLEDAI-2K organ system for the musculoskeletal domain. (B) Improvement from baseline in the SLEDAI-2K organ system for the mucocutaneous domain. (C) Improvement from baseline in the BILAG organ system for the musculoskeletal domain. (D) Improvement from baseline in the BILAG organ system for the mucocutaneous domain.

The analysis population included participants who had a positive organ domain score in each assessment at baseline; BILAG: British Isles Lupus Assessment Group; SLEDAI-2K: Systemic Lupus Erythematosus Disease Activity Index-2000; *P ≤ 0.05; †Nominal P ≤ 0.01.

At least one treatment-emergent adverse event (AE) was observed for 408 (80%) participants in the baricitinib 4 mg group, 409 (79%) in the baricitinib 2 mg group, and 408 (80%) in the placebo group (Table 3). In most AEs, the severity is mild or moderate. Serious adverse events (SAEs) were reported in 55 (11%) participants in the baricitinib 4 mg group, 59 (11%) in the baricitinib 2 mg group, and 40 (8%) in the placebo group (Table 3). Four (1%) deaths were reported in participants randomized to baricitinib 4 mg group. Of these four deaths, 2 were due to myocardial infarctions, 1 was sepsis, and 1 was confirmed COVID-19-related acute respiratory failure (this one was randomly assigned to baricitinib 4 mg but received actually baricitinib 2 mg). One (0.2%) death was reported in participants assigned to baricitinib 2 mg. Four (1%) deaths were reported in the placebo group, all of whom were due to respiratory failure (three was confirmed COVID-19-related respiratory failure) (Table 3). Table 3 summarizes the withdrawal of treatment due to AEs. Compared with placebo (11 [2%] participants), more serious infections were reported in participants treated with baricitinib 4 mg (22 [4%] participants) and baricitinib 2 mg (20 [4%] participants) (Table 3). One (0.2%) adjudicated venous thrombotic event was reported in participants treated with baricitinib 4 mg, 3 (1%) in participants treated with baricitinib 2 mg, and 6 (1%) in the placebo group. Three (1%) adjudicated major cardiovascular events were reported in participants treated with baricitinib 4 mg, 1 (0.2%) in participants treated with baricitinib 2 mg, and none in participants treated with placebo (Table 3). There were 4 (1%) malignancies reported in participants treated with placebo, 3 (1%) in participants treated with baricitinib 2 mg, and 2 (0.4%) in participants treated with baricitinib 4 mg (Table 3). Laboratory results from patients would indicate no potential cases of Hy’s Law, which is defined as having a maximum post-baseline total bilirubin of twice or more upper limit of normal and a maximum post-baseline alanine aminotransferase or aspartate aminotransferase of concentrations triple or more upper limit of normal. Safety data are further described in Table 3 and S2-4 Tables.

Table 3. Overview of adverse events.

Placebo (n = 509) Baricitinib 2 mg (n = 516) Baricitinib 4 mg (n = 510)
Treatment-emergent AEs 408 (80%) 409 (79%) 408 (80%)
Mild 174 (34%) 170 (33%) 171 (34%)
Moderate 199 (39%) 192 (37%) 189 (37%)
Severe 35 (7%) 47 (9%) 48 (9%)
 SAEs 40 (8%) 59 (11%) 55 (11%)
 Death 4 (1%) 1 (0.2%) 4 (1%)
 Discontinuation from study because of an AE 44 (8.6%) 48 (9%) 46 (9%)
Infections 260 (51%) 266 (52%) 264 (52%)
 Serious infections 11 (2%) 20 (4%) 22 (4%)
 Herpes simplex 16 (3%) 19 (4%) 11 (2%)
 Herpes zoster virus infection 17 (3%) 17 (3%) 28 (5%)
 Opportunistic infections 23 (5%) 30 (6%) 38 (7%)
Major adverse cardiovascular events* 0 1 (0.2%) 3 (1%)
Cardiovascular death 0 0 2 (0.4%)
Myocardial infarction 0 1 (0.2%) 1 (0.2%)
Stroke 0 0 1 (0.2%)
 Arterial thromboembolic events* 0 0 0
 Venous thromboembolic events* 6 (1%) 3 (1%) 1 (0.2%)
 Deep-vein thrombosis 1 (0.2%) 3 (1%) 0
 Pulmonary embolism 1 (0.2%) 2 (0.4%) 0
 Other 4 (1%) 0 1 (0.2%)
Malignancies 4 (1%) 3 (1%) 2 (0.4%)
 Non-melanoma skin cancer 2 (0.4%) 0 0
 Malignancies other than non-melanoma skin cancer 2 (0.4%) 3 (1%) 2 (0.4%)
Gastrointestinal perforations 0 1 (0.2%) 0
Hepatic disorders 21 (4%) 38 (8%) 22 (4%)

Data are n (%); AE, adverse event; SAE, Serious adverse events; AEs that occurred between baseline and week 52 and up to 28 days after treatment are shown;

*Positively adjudicated by an independent external masked clinical event committee;

†Other events were all classified as non-superficial below-knee thrombosis.

Discussion

With various and selective inhibitory profiles, JAK inhibitors have enabled a shift in the treatment of multiple autoimmune diseases [2629]. The etiology of SLE, RA, AA, and other inflammatory diseases involves the intracellular signaling of multiple cytokines, JAK inhibitors can block these signal pathways [30,31].

Baricitinib is an oral small-molecule inhibitor of JAK1/2 signaling which is approved for the management of RA, juvenile idiopathic arthritis (JIA), atopic dermatitis (AD), AA, and coronavirus disease 2019 (COVID-19). Substantial evidence backs the role of pathways blocked by baricitinib in the pathogenesis of systemic lupus erythematosus [32], including evidence showed effects on expression of serum cytokines, IFNs responsive genes, and JAK-STAT pathway genes [13,1820], and positive results of a phase Ⅲ study (SLE-BRAVE-I), which showed that baricitinib 4 mg plus SOC was superior to placebo in conjunction with SOC in reaching SRI-4 response at week 52 [21]. However, the result was not replicated in another phase Ⅲ study (SLE-BRAVE-II) [22], necessitating a pooled analysis of the only two available phase Ⅲ randomized clinical trials to investigate the efficacy and safety of baricitinib 4 mg and baricitinib 2 mg versus placebo in active SLE participants treated with SOC.

In this study, the proportion of participants reaching an SRI-4 response was not achieved with once-daily oral baricitinib 4 mg or baricitinib 2 mg plus SOC compared with placebo. The reasons for not achieving SRI-4 response at week 52 are detailed in S5 Table. In any non-multiplicity-controlled secondary outcomes or exploratory outcomes, no difference was found between baricitinib 4 mg or 2 mg and placebo. However, in some clinically important subpopulations, SRI-4 response was achieved, such as in participants treated with a glucocorticoid dose at baseline of 10 mg per day or higher of prednisone or equivalent (baricitinib 4 mg or baricitinib 2 mg) and in participants with highly active disease (SLEDAI-2K score at baseline >10) (baricitinib 4 mg). These findings confirm the efficacy of baricitinib for treating SLE in these subgroups. Patient selection might not be a major factor affecting the efficacy [8], because the patients profile in this study, including concomitant medication and organ involvement, was similar to those of other recently published SLE clinical trials [21,22,8]. Patients with baseline glucocorticoid use of ≥ 10 mg/day often exhibit more severe disease activity or flares that necessitate aggressive treatment. Baricitinib functions by inhibiting JAK enzymes, which can help reduce inflammation and immune system activity in patients with SLE. Clinical trials suggest that baricitinib may improve disease activity in SLE patients, particularly those who have inadequate responses to conventional therapies, including glucocorticoids [15]. Identifying biomarkers that predict response to treatment in SLE is essential for personalizing therapy and improving patient outcomes. Key biomarkers include anti-dsDNA antibodies, complement levels (C3 and C4), and SLEDAI scores, among others.

In general, baricitinib 4 mg or baricitinib 2 mg for treating SLE did not increase the incidence of treatment-emergent adverse events (TEAEs). There were no new safety signals observed, and safety profile of baricitinib was consistent with what is known. Patients receiving baricitinib 4 mg had higher rates of herpes zoster infections, which is consistent with other treatment groups and is a typical complication of JAK1 inhibitors [15]. In both the baricitinib 4 mg and baricitinib 2 mg groups, the serious infection was higher, but within the expected range. Other TEAEs and SAEs, including MACE, infections, malignancy, and hepatic disorders, were all within the expected range. It is important to note that in the population at high risk of VTEs, there was no gastrointestinal perforations, or VTE excess [33].

This study also has several limitations. Firstly, similar to other trials in SLE, this study did not include patients with central nervous system (CNS) and severe active renal disease. Secondly, the overall public health context during COVID-19 created an acute focus on health issues, potentially fostering a placebo-enhancing environment in which patients were more inclined to attribute improvements or changes in symptoms to their treatment, even when they received a placebo. Part of the two trials took place during the COVID-19 pandemic, and the placebo rates were higher compared with other recent SLE studies [34,35], which may cover up the efficacy of baricitinib for treating SLE to some extent. Thirdly, steroid tapering was not carried out as rigorously as it might have been in other recent trials, which could lead to an increase in placebo response rate. Lastly, analysis should be interpreted with caution due to missing data and inadequate analytical power in the subgroups of participants who received glucocorticoid dose at baseline of 10 mg daily or higher of prednisone or equivalent or SLEDAI-2K score of 10 or higher.

Conclusions

In conclusion, this pooled analysis of the SLE-BRAVE-I and Ⅱ trials demonstrated that once-daily oral baricitinib 4 mg or baricitinib 2 mg plus SOC did not reduce the overall disease activity. However, baricitinib is likely to be a good treatment option for SLE in certain subpopulation, such as in participants treated with a glucocorticoid dose at baseline of 10 mg daily or higher of prednisone or equivalent (baricitinib 4 mg and baricitinib 2 mg) and in participants with highly active disease (SLEDAI-2K score at baseline >10) (baricitinib 4 mg). Furthermore, baricitinib 4 mg and baricitinib 2 mg for treating SLE did not increase the incidence of TEAEs. These data are invaluable for healthcare professionals to provide consultation on treatment expectations and potential benefits to patients.

Supporting information

S1 Fig. Study Design.

(TIF)

pone.0320179.s001.tif (308.5KB, tif)
S1 Table. Description of Efficacy Measures.

(DOCX)

pone.0320179.s002.docx (19.8KB, docx)
S2 Table. Treatment Emergent Adverse Events Occurring in ≥ 5% of Patients in Either Baricitinib Dose Group, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s003.docx (17.5KB, docx)
S3 Table. Serious Adverse Events by System Organ Class, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s004.docx (17.9KB, docx)
S4 Table. Serious Infections and Infestations, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s005.docx (17.3KB, docx)
S5 Table. Reasons for not Achieving SLE Responder Index-4 at Week 52.

(DOCX)

pone.0320179.s006.docx (17.5KB, docx)

Acknowledgments

Our gratitude goes to the authors who conducted the two primary investigations included in this study.

Abbreviations

AE

Adverse event

AA

Alopecia areata

AD

Atopic dermatitis

CNS

Central nervous system

CONSORT

Consolidated Standards of Reporting Trial

COVID-19

Coronavirus disease 2019

FACIT

Functional Assessment of Chronic Illness Therapy

IFN

Interferons

IL

Interleukin

JAK

Janus kinases

JIA

Juvenile idiopathic arthritis

LLDAS

Lupus low disease activity state

MACE

Major adverse cardiovascular events

NRS

Numeric Rating Scale

OR

Odds ratio

RA

Rheumatoid arthritis

SAE

Serious adverse events

SRI

SLE Responder Index

SLE

Systemic lupus erythematosus

SOC

Standard of care

TEAE

Treatment-emergent adverse event

TNF

Tumor necrosis factor

VTE

Venous thromboembolic event

Data Availability

The data underlying the results presented in this study are owned by non-authors and are available from the following sources: https://clinicaltrials.gov/study/NCT03616912 https://clinicaltrials.gov/study/NCT03616964

Funding Statement

This research was funded by grant from Science and Technology Department of Henan Province (Award Number 112102310306). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12(12):716–30. doi: 10.1038/nrrheum.2016.186 [DOI] [PubMed] [Google Scholar]
  • 2.Shiozawa S, Tsumiyama K, Miyazaki Y, Uto K, Sakurai K, Nakashima T, et al. DOCK8-expressing T follicular helper cells newly generated beyond self-organized criticality cause systemic lupus erythematosus. iScience. 2022;25(1):103537. doi: 10.1016/j.isci.2021.103537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Aletaha D, Smolen JS. Diagnosis and management of rheumatoid arthritis: a review. JAMA. 2018;320(13):1360–72. doi: 10.1001/jama.2018.13103 [DOI] [PubMed] [Google Scholar]
  • 4.Jorge AM, Lu N, Zhang Y, Rai SK, Choi HK. Unchanging premature mortality trends in systemic lupus erythematosus: a general population-based study (1999-2014). Rheumatology (Oxford). 2018;57(2):337–44. doi: 10.1093/rheumatology/kex412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kandane-Rathnayake R, Louthrenoo W, Hoi A, Luo S-F, Wu Y-JJ, Chen Y-H, et al. “Not at target”: prevalence and consequences of inadequate disease control in systemic lupus erythematosus-a multinational observational cohort study. Arthritis Res Ther. 2022;24(1):70. doi: 10.1186/s13075-022-02756-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yen EY, Singh RR. Brief report: lupus-an unrecognized leading cause of death in young females: a population-based study using nationwide death certificates, 2000-2015. Arthritis Rheumatol. 2018;70(8):1251–5. doi: 10.1002/art.40512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Navarra SV, Guzmán RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9767):721–31. doi: 10.1016/s0140-6736(10)61354-2 [DOI] [PubMed] [Google Scholar]
  • 8.Connelly K, Golder V, Kandane-Rathnayake R, Morand EF. Clinician-reported outcome measures in lupus trials: a problem worth solving. Lancet Rheumatol. 2021;3(8):e595–603. doi: 10.1016/S2665-9913(21)00119-3 [DOI] [PubMed] [Google Scholar]
  • 9.Fanouriakis A, Kostopoulou M, Alunno A, Aringer M, Bajema I, Boletis JN, et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann Rheum Dis. 2019;78(6):736–45. doi: 10.1136/annrheumdis-2019-215089 [DOI] [PubMed] [Google Scholar]
  • 10.Singh JA, Shah NP, Mudano AS. Belimumab for systemic lupus erythematosus. Cochrane Database Syst Rev. 2021;2(2):CD010668. doi: 10.1002/14651858.CD010668.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wahren-Herlenius M, Dörner T. Immunopathogenic mechanisms of systemic autoimmune disease. Lancet. 2013;382(9894):819–31. doi: 10.1016/s0140-6736(13)60954-x [DOI] [PubMed] [Google Scholar]
  • 12.Larosa M, Zen M, Gatto M, Jesus D, Zanatta E, Iaccarino L, et al. IL-12 and IL-23/Th17 axis in systemic lupus erythematosus. Exp Biol Med (Maywood). 2019;244(1):42–51. doi: 10.1177/1535370218824547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dörner T, van Vollenhoven RF, Doria A, Jia B, Ross Terres JA, Silk ME, 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hoffman RW, Merrill JT, Alarcón-Riquelme MM, Petri M, Dow ER, Nantz E, et al. Gene Expression and pharmacodynamic changes in 1,760 systemic lupus erythematosus patients from two phase III trials of BAFF blockade with tabalumab. Arthritis Rheumatol. 2017;69(3):643–54. doi: 10.1002/art.39950 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O’Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843–62. doi: 10.1038/nrd.2017.201 [DOI] [PubMed] [Google Scholar]
  • 16.O’Shea JJ, Kontzias A, Yamaoka K, Tanaka Y, Laurence A. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis. 2013;72 Suppl 2(0 2):ii111–5. doi: 10.1136/annrheumdis-2012-202576 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.King B, Ohyama M, Kwon O, Zlotogorski A, Ko J, Mesinkovska NA, et al. Two phase 3 trials of baricitinib for alopecia areata. N Engl J Med. 2022;386(18):1687–99. doi: 10.1056/NEJMoa2110343 [DOI] [PubMed] [Google Scholar]
  • 18.Wallace DJ, Furie RA, Tanaka Y, Kalunian KC, Mosca M, Petri MA, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 2018;392(10143):222–31. doi: 10.1016/S0140-6736(18)31363-1 [DOI] [PubMed] [Google Scholar]
  • 19.Dörner T, Tanaka Y, Petri MA, Smolen JS, Wallace DJ, Dow ER, 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dörner T, Tanaka Y, Dow ER, Koch AE, Silk M, Ross Terres JA, et al. Mechanism of action of baricitinib and identification of biomarkers and key immune pathways in patients with active systemic lupus erythematosus. Ann Rheum Dis. 2022;81(9):1267–72. doi: 10.1136/annrheumdis-2022-222335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Morand EF, Vital EM, Petri M, van Vollenhoven R, Wallace DJ, Mosca M, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-I). Lancet. 2023;401(10381):1001–10. doi: 10.1016/s0140-6736(22)02607-1 [DOI] [PubMed] [Google Scholar]
  • 22.Petri M, Bruce IN, Dörner T, Tanaka Y, Morand EF, Kalunian KC, et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 3 trial (SLE-BRAVE-II). Lancet. 2023;401(10381):1011–9. doi: 10.1016/s0140-6736(22)02546-6 [DOI] [PubMed] [Google Scholar]
  • 23.Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725. doi: 10.1002/art.1780400928 [DOI] [PubMed] [Google Scholar]
  • 24.Tan EM, Cohen AS, Fries JF, Masi AT, Mcshane DJ, Rothfield NF, et al. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982;25(11):1271–7. doi: 10.1002/art.1780400928 [DOI] [PubMed] [Google Scholar]
  • 25.Golder V, Kandane-Rathnayake R, Huq M, Nim HT, Louthrenoo W, Luo SF, et al. Lupus low disease activity state as a treatment endpoint for systemic lupus erythematosus: a prospective validation study. Lancet Rheumatol. 2019;1(2):e95–102. doi: 10.1016/S2665-9913(19)30037-2 [DOI] [PubMed] [Google Scholar]
  • 26.Waibel M, Wentworth JM, So M, Couper JJ, Cameron FJ, MacIsaac RJ, et al. Baricitinib and β-cell function in patients with new-onset type 1 diabetes. N Engl J Med. 2023;389(23):2140–50. doi: 10.1056/NEJMoa2306691 [DOI] [PubMed] [Google Scholar]
  • 27.Ramanan AV, Quartier P, Okamoto N, Foeldvari I, Spindler A, Fingerhutová Š, et al. Baricitinib in juvenile idiopathic arthritis: an international, phase 3, randomised, double-blind, placebo-controlled, withdrawal, efficacy, and safety trial. Lancet. 2023;402(10401):555–70. doi: 10.1016/s0140-6736(23)00921-2 [DOI] [PubMed] [Google Scholar]
  • 28.Szekanecz Z, Buch MH, Charles-Schoeman C, Galloway J, Karpouzas GA, Kristensen LE, et al. Efficacy and safety of JAK inhibitors in rheumatoid arthritis: update for the practising clinician. Nat Rev Rheumatol. 2024;20(2):101–15. doi: 10.1038/s41584-023-01062-9 [DOI] [PubMed] [Google Scholar]
  • 29.Verstovsek S, Gerds AT, Vannucchi AM, Al-Ali HK, Lavie D, Kuykendall AT, et al. Momelotinib versus danazol in symptomatic patients with anaemia and myelofibrosis (MOMENTUM): results from an international, double-blind, randomised, controlled, phase 3 study. Lancet. 2023;401(10373):269–80. doi: 10.1016/s0140-6736(22)02036-0 [DOI] [PubMed] [Google Scholar]
  • 30.Tanaka Y, Luo Y, O’Shea JJ, Nakayamada S. Janus kinase-targeting therapies in rheumatology: a mechanisms-based approach. Nat Rev Rheumatol. 2022;18(3):133–45. doi: 10.1038/s41584-021-00726-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Loftus EV Jr, Panés J, Lacerda AP, Peyrin-Biroulet L, D’Haens G, Panaccione R, et al. Upadacitinib induction and maintenance therapy for crohn’s disease. N Engl J Med. 2023;388(21):1966–80. doi: 10.1056/NEJMoa2212728 [DOI] [PubMed] [Google Scholar]
  • 32.Kubo S, Nakayamada S, Tanaka Y. Baricitinib for the treatment of rheumatoid arthritis and systemic lupus erythematosus: a 2019 update. Expert Rev Clin Immunol. 2019;15(7):693–700. doi: 10.1080/1744666x.2019.1608821 [DOI] [PubMed] [Google Scholar]
  • 33.Hoppe B, Dörner T. Coagulation and the fibrin network in rheumatic disease: a role beyond haemostasis. Nat Rev Rheumatol. 2012;8(12):738–46. doi: 10.1038/nrrheum.2012.184 [DOI] [PubMed] [Google Scholar]
  • 34.Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, et al. Anifrolumab, an anti-interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol. 2017;69(2):376–86. doi: 10.1002/art.39962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med. 2020;382(3):211–21. doi: 10.1056/NEJMoa1912196 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

S1 Fig. Study Design.

(TIF)

pone.0320179.s001.tif (308.5KB, tif)
S1 Table. Description of Efficacy Measures.

(DOCX)

pone.0320179.s002.docx (19.8KB, docx)
S2 Table. Treatment Emergent Adverse Events Occurring in ≥ 5% of Patients in Either Baricitinib Dose Group, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s003.docx (17.5KB, docx)
S3 Table. Serious Adverse Events by System Organ Class, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s004.docx (17.9KB, docx)
S4 Table. Serious Infections and Infestations, Weeks 0–52 and up to 28 Days Post-Treatment.

(DOCX)

pone.0320179.s005.docx (17.3KB, docx)
S5 Table. Reasons for not Achieving SLE Responder Index-4 at Week 52.

(DOCX)

pone.0320179.s006.docx (17.5KB, docx)

Data Availability Statement

The data underlying the results presented in this study are owned by non-authors and are available from the following sources: https://clinicaltrials.gov/study/NCT03616912 https://clinicaltrials.gov/study/NCT03616964


Articles from PLOS One are provided here courtesy of PLOS

RESOURCES