Baricitinib

Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial

Daniel J Wallace, Richard A Furie, Yoshiya Tanaka, Kenneth C Kalunian, Marta Mosca, Michelle A Petri, Thomas Dörner, Mario H Cardiel, Ian N Bruce, Elisa Gomez, Tara Carmack, Amy M DeLozier, Jonathan M Janes, Matthew D Linnik, Stephanie de Bono, Maria E Silk, Robert W Hoffman

Summary
Background Patients with systemic lupus erythematosus have substantial unmet medical need. Baricitinib is an oral selective Janus kinase (JAK)1 and JAK2 inhibitor that we hypothesised might have therapeutic benefit in patients with systemic lupus erythematosus.

Methods In this double-blind, multicentre, randomised, placebo-controlled, 24-week phase 2 study, patients were recruited from 78 centres in 11 countries. Eligible patients were aged 18 years or older, had a diagnosis of systemic lupus erythematosus, and had active disease involving skin or joints. We randomly assigned patients (1:1:1) to receive once-daily baricitinib 2 mg, baricitinib 4 mg, or placebo for 24 weeks. The primary endpoint was the proportion of patients achieving resolution of arthritis or rash at week 24, as defined by Systemic Lupus Erythematosus Disease Activity Index-2000 (SLEDAI-2K). Efficacy and safety analyses included all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT02708095.

Findings Between March 24, 2016, and April 27, 2017, 314 patients were randomly assigned to receive placebo (n=105), baricitinib 2 mg (n=105), or baricitinib 4 mg (n=104). At week 24, resolution of SLEDAI-2K arthritis or rash was achieved by 70 (67%) of 104 patients receiving baricitinib 4 mg (odds ratio [OR] vs placebo 1·8, 95% CI 1·0–3·3; p=0·0414) and 61 (58%) of 105 patients receiving baricitinib 2 mg (OR 1·3, 0·7–2·3; p=0·39). Adverse events were reported in 68 (65%) patients in the placebo group, 75 (71%) patients in the baricitinib 2 mg group, and 76 (73%) patients in the baricitinib 4 mg group. Serious adverse events were reported in ten (10%) patients receiving baricitinib 4 mg, 11 (10%) receiving baricitinib 2 mg, and five (5%) receiving placebo; no deaths were reported. Serious infections were reported in six (6%) patients with baricitinib 4 mg, two (2%) with baricitinib 2 mg, and one (1%) with placebo.

Interpretation The baricitinib 4 mg dose, but not the 2 mg dose, significantly improved the signs and symptoms of active systemic lupus erythematosus in patients who were not adequately controlled despite standard of care therapy, with a safety profile consistent with previous studies of baricitinib. This study provides the foundation for future phase 3 trials of JAK1/2 inhibition with baricitinib as a new potential oral therapy for systemic lupus erythematosus.

Funding Eli Lilly and Company.

Copyright © 2018 Elsevier Ltd. All rights reserved.

Biology, Medicine and Health, The University of Manchester
and NIHR Manchester Biomedical Research Centre,
Manchester University Hospitals NHS Foundation
Trust, Manchester Academic Health Science Centre,
Manchester, UK (Prof I N Bruce MD); Lilly
Biotechnology Center,
San Diego, CA, USA (M D Linnik PhD); and Eli Lilly
and Company, Indianapolis, IN,
USA (E Gomez MS, T Carmack MS,
A M DeLozier MPH, J M Janes MD, S de Bono MD,
M E Silk PharmD, Prof R W Hoffman, DO)
Introduction
Systemic lupus erythematosus is a multisystem, chronic autoimmune disease characterised by the presence of antibodies directed at self-antigens, and broad immune
1 It is believed that abnormalities of both the innate and adaptive arms of the immune system, interconnected by a positive feedback loop, contribute to
2
Many key cytokines implicated in the pathogenesis of systemic lupus erythematosus are dependent on activation of Janus kinases (JAKs) for intracellular
1,3,4 The JAK family of cytoplasmic protein tyrosine kinases mediates the signalling of several pro-inflammatory cytokines, such as type l inter- ferons (JAK1/tyrosine kinase [TYK] 2), interleukin 6 (JAK1/JAK2/TYK2), and interleukin 12 and interleukin 23
3 Baricitinib is an orally administered
5
which has been approved for the treatment of moderately to severely active rheumatoid arthritis in adults in over 40 countries, including European countries, the USA, and Japan. This molecular mechanism of action suggests that baricitinib might inhibit cytokines central to the dysregulated innate and adaptive immune function observed in systemic lupus erythematosus.
We designed this international, double-blind, random- ised, placebo-controlled, phase 2 trial to assess the efficacy, safety, and tolerability of oral baricitinib (2 mg or 4 mg) once-daily in patients with active systemic lupus erythematosus, who were receiving standard background therapy.

Methods
Study design and participants
In this double-blind, randomised, placebo-controlled trial, 79 investigators assessed patients at 78 centres in

Correspondence to:

Research in context Evidence before this study
We searched PubMed using the terms “systemic lupus erythematosus”, “treatment”, and “JAK inhibitor” for articles in English published up to March 1, 2016, regardless of article type. We considered previous clinical trials of investigational medication in systemic lupus erythematosus, as well as previous clinical trials of the Janus kinase (JAK)1/2 inhibitor, baricitinib, before doing this study. Current treatment regimens for the management of systemic lupus erythematosus have resulted in a reduction of morbidity and mortality over previous decades; however, many patients still have incompletely controlled disease and can progress to
end-stage organ involvement. There are a small number of medications approved for the treatment of systemic lupus erythematosus and other medications are used off-label; collectively, these include antimalarials, non-steroidal
anti-inflammatory drugs, aspirin, corticosteroids, azathioprine, mycophenolate, cyclophosphamide, methotrexate,
belimumab, and rituximab. Because of their limited efficacy, these drugs are often used in multiple combinations, rather than as single agents, resulting in added risk of toxicity. New treatment options with an acceptable safety profile that reduce disease activity, reduce flares, delay organ damage, and reduce the requirement for corticosteroids and cytotoxic drugs are urgently needed for patients with systemic lupus erythematosus. Additionally, the need for treatments that improve quality of life, including control of pain and fatigue, has been articulated by patients and physicians. Clinical and laboratory studies have showed a central role for JAK and

11 countries in Asia, Europe, North America, and South America. The study protocol is available from the sponsor.
Eligible participants were 18 years or older and had been diagnosed with systemic lupus erythematosus at least 24 weeks before screening by fulfilling four or more of the revised American College of Rheumatology (ACR) criteria for classification of systemic lupus erythe- matosus, or the 2012 Systemic Lupus Erythematosus International Collaborating Clinics (SLICC) classification
6–8 At baseline, patients were required to have a positive antinuclear antibody (HEp-2 titre ≥1:80), a positive anti-double-stranded DNA (anti-dsDNA; ≥30 IU/mL), IgG INOVA QUANTA Lite SC ELISA (INOVA Diagnostics, San Diego, CA, USA), a Systemic Lupus Erythematosus Disease Activity Index-2000 (SLEDAI-2K) score of 4 or greater based on clinical manifestations, or active arthritis or rash as defined by the SLEDAI-2K. Study drug was added to existing stable background standard of care therapy, which could be non-steroidal anti-inflammatory drugs, corticosteroids, a single antimalarial (such as chloroquine or hydroxy- chloroquine), or a single immunosuppressant (such as azathioprine, methotrexate, or mycophenolate). The

signal transducer and activator of transcription (STAT) in cytokine-mediated immune signalling and inflammation. This pathway includes JAK/STAT-mediated signalling through
type 1 interferons, which have been shown to have an important role in the pathogenesis of systemic lupus erythematosus.
Added value of this study
In this phase 2 trial, baricitinib improved the signs and symptoms of active disease in patients with systemic lupus erythematosus who were receiving standard background therapy. Baricitinib 4 mg treatment resulted in a greater proportion of patients achieving resolution of arthritis, as defined by Systemic Lupus Erythematosus Disease Activity Index-2000, than with placebo. The safety profile of baricitinib was consistent with other drugs used to treat active systemic lupus erythematosus.
Implications of all the available evidence
These findings support the evidence that JAK/STAT signalling could have a central role in the pathogenesis of systemic lupus erythematosus. To our knowledge, this is the first study to show clinical benefit of JAK inhibition in the treatment of systemic lupus erythematosus. This work provides the foundation for additional study of JAK1/2 inhibition with baricitinib as a potentially effective oral treatment option for active systemic lupus erythematosus in patients who have
not achieved adequate disease control with available standard of care therapies.

corticosteroid dose was limited to 20 mg or less of prednisone per day (or equivalent), and was required to be stable for 2 weeks before randomisation; increases in dose were not permitted after randomisation. Decreases in corticosteroid dose were permitted from baseline to week 16. No changes in corticosteroid dose were permitted between week 16 and week 24. No increases in antimalarials or immunosuppressants were allowed at any time. Key exclusion criteria included active severe lupus nephritis, active severe CNS lupus, recent clinically serious infection, and selected laboratory abnormalities. A full list of exclusion criteria are shown in the appendix.
The study was done in accordance with the ethical principles of the Declaration of Helsinki and Good Clinical Practice guidelines. All investigation sites received approval from the appropriate authorised institutional review board or ethics committee. All patients provided written informed consent before the study-related procedures were done.

Randomisation and masking
We used a computer-generated random sequence to allocate patients (1:1:1) to placebo, baricitinib 2 mg, or
Prof Daniel J Wallace, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA [email protected]

See Online for appendix

baricitinib 4 mg. Patients were stratified according to disease severity (SLEDAI-2K score <10 or ≥10), anti- dsDNA status (positive or negative), and region (USA, Europe, Asia, or rest of world). Patients and investigators were masked to allocation. Daily doses were provided at each visit in two bottles containing identical investigational product tablets supplied by Eli Lilly and Company. All study investigators remained masked until the week 24 analysis.

Procedures
All interventions were given orally. Patients were instructed to take one tablet from each bottle each day (one tablet of placebo, one tablet of study drug). Safety outcomes (clinical laboratory tests, vital signs, and other safety assessments) and efficacy outcomes were assessed on scheduled study visits (baseline and weeks 2, 4, 8, 12, 16, 20, and 24) during the double-blind treatment period, as indicated in the protocol schedule of events. Patients were also assessed approximately 28 days after receiving their last dose of study drug.

Outcomes
The primary outcome was the proportion of patients achieving resolution of arthritis or rash, as defined by the SLEDAI-2K, at week 24.
The proportion of patients achieving a Systemic Lupus Erythematosus Responder Index-4 (SRI-4) response at week 24 was a secondary objective. SRI-4 response is defined as a reduction of at least 4 points from baseline in SLEDAI-2K score, no worsening in British Isles Lupus Assessment Group (BILAG) A or B

disease activity scores (no new BILAG A score or no more than one new BILAG B score), and no worsening (defined as an increase of ≥0·3 points [10 mm] from baseline) in the Physician’s Global Assessment of Disease Activity (PGA). Other secondary endpoints were the proportion of patients achieving a reduction of 4 points or more from baseline in SLEDAI-2K score, and change from baseline in SLEDAI-2K total score and PGA at week 24. We analysed plasma baricitinib concentrations using a population pharmacokinetic approach, to characterise the exposure response for SLEDAI-2K and SRI-4 across 24 weeks (data not shown). Exploratory assessments were change from baseline in 28-tender joint count and 28-swollen joint count, Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) activity score, and SLICC/ACR Damage index; risk of flares of any severity, and severe flares on the Safety of Estrogens in Lupus Erythematosus National Assessment (SELENA)- SLEDAI Flare Index (SSFI); and proportion of patients achieving Lupus Low Disease Activity Score (LLDAS). We also recorded corticosteroid doses and measured changes in serological markers, including anti-dsDNA and complement component (C) 3 and C4. We assessed change in the patient-reported outcomes Worst Joint Pain numeric rating scale (NRS), Worst Pain NRS, and Worst Fatigue NRS (appendix pp 20–23).
Incidence and severity of all adverse events were recorded between baseline and week 24, and up to 30 days after treatment had ended, and the Quick Inventory of Depressive Symptomatology Self-Rated-16 was assessed. We used the National Institutes of Health Common Terminology Criteria for Adverse Events (CTCAE) to describe laboratory abnormalities

414 patients screened

81 screening failure
4 physician’s decision

100 excluded
11 withdrew 4 other
314 randomly assigned

105 assigned to placebo 105 assigned to baricitinib 2 mg 104 assigned to baricitinib 4 mg

22 discontinued
4 adverse event 5 withdrew
2 physician’s decision 9 lack of efficacy
2 other
19 discontinued
10 adverse event 3 withdrew
3 physician’s decision
3 lack of efficacy
18 discontinued
11 adverse event
1 protocol violation 4 withdrew
2 physician’s decision

83 completed double-blind treatment period 86 completed double-blind treatment period 86 completed double-blind treatment period
(version 4.03).

Statistical analysis
With approximately 100 patients per treatment group, this study had approximately 81% power to detect a difference of 20% between baricitinib 2 mg or 4 mg and placebo for the primary endpoint. The expected treatment difference was based on an expected overall placebo response rate of 40% and an expected response rate of 60% with baricitinib 2 mg or 4 mg. We calculated the power for planned sample size with a two-group χ² test of equal proportions, using nQuery Advisor (version 7.0).
We analysed the primary, secondary, and exploratory endpoints according to the prespecified statistical analysis plan.
We made no multiplicity adjustments. We analysed efficacy and patient-reported outcomes (PGA, Worst Pain NRS, Worst Joint Pain NRS, Worst Fatigue NRS) in the modified intention-to-treat population, defined as all randomly assigned patients treated with at least one

dose of study drug. We made treatment comparisons of
Figure 1: Trial profile categorical efficacy endpoints using logistic regression,

with treatment, region, baseline disease severity (SLEDAI-2K score <10 vs ≥10), and baseline anti-dsDNA status (positive or negative) in the model. We made

Placebo (n=105)

Baricitinib 2 mg (n=105)

Baricitinib 4 mg (n=104)

treatment comparisons of continuous efficacy endpoints using pairwise comparisons in mixed models repeated measures for outcomes with multiple post-baseline values, or ANCOVA for outcomes with a single post- baseline value. The mixed models repeated measures model included treatment, baseline score, baseline disease severity (SLEDAI-2K score <10 vs ≥10), baseline anti-dsDNA status (positive or negative), region, visit, and the interaction of treatment-by-visit as fixed factors. The ANCOVA model included treatment, baseline score, baseline disease severity (SLEDAI-2K score <10 vs ≥10), baseline anti-dsDNA status (positive or negative), and region as explanatory variables. We made treatment comparisons for the time to first systemic lupus erythematosus flare (SSFI) using a Cox proportional hazards model with treatment group, baseline disease severity (SLEDAI-2K score <10 vs ≥10), baseline anti-dsDNA status (positive or negative), and region fitted as explanatory variables. We assessed safety in patients who received at least one dose of study drug and who were not lost to follow-up before the first post-baseline visit. Adverse events were inclusive of the treatment period and up to 30 days after treat- ment, and laboratory abnormality summaries included the treatment period and up 60 days after treatment. We used the Fisher exact test for all adverse events, discontinuation, and other categorical safety data. We analysed continuous safety data using ANCOVA, adjusting for baseline value and treatment. All analyses were assessed at a two-sided α of 0·05.
For categorical efficacy analyses, patients were con- sidered non-responders at each visit if they were not responders at that visit; permanently discontinued study treatment at any time before that visit for any reason; required initiation or increase (from baseline) in dose of non-steroidal anti-inflammatory drugs (for 30 or more consecutive days), corticosteroids, antimalarials, or immunosuppressants, after randomisation; or had missing data at that visit. No imputation was needed with mixed models repeated measures for analysing the continuous efficacy endpoints. This study is registered with ClincialTrials.gov, number NCT02708095.

Role of the funding source
The funder of the study had a role in study design, data analysis, data collection, data interpretation, or writing of the report. The corresponding author had full access to all the data in the study and had final responsibility for

the decision to submit for publication.

Results
Between March 24, 2016, and April 27, 2017, 314 patients were randomly assigned to receive once-daily placebo (n=105), baricitinib 2 mg (n=105), or baricitinib 4 mg
Table 1: Baseline characteristics and disease activity

(n=104); 255 (81%) completed the 24-week double-blind treatment period (figure 1). No patients were excluded from the efficacy and safety analyses.

A Resolution of arthritis or rash (SLEDAI-2K) B SRI-4 response

100

80

60

40

20

0
Placebo Baricitinib 2 mg Baricitinib 4 mg
100

80

60

40

20

0

0 2 4 8 12 16 20 24 0 2 4 8 12 16 20 24
p value

Baricitinib 2 mg Baricitinib 4 mg
0·97
0·56
0·57
0·18
0·54
0·56
0·91
0·26
0·15
0·06
0·38 0·0293
0·39 0·0414
0·72
0·55
0·34
0·12
0·42
0·48
0·67
0·33
0·25
0·34
0·48
0·09
0·44 0·0151

C SLEDAI-2K score D Physician’s Global Assessment

0

–1·0

–2·0
0

–10

–20

–3·0

–4·0

–5·0
–30

–40

0 2 4 p value
8
12
Time (weeks)
16
20
24
0 2 4
8
12
Time (weeks)
16
20
24

Baricitinib 2 mg Baricitinib 4 mg
0·77
0·31
0·72
0·66
0·68
0·98
0·98
0·88
0·28
0·30
0·97
0·42
0·60
0·24
0·38
0·41
0·98
0·34
0·60
0·26
0·31
0·08
0·11
0·41
0·37
0·08
0·87 0·0218

Figure 2: Primary and secondary efficacy analyses
(A) Proportion of patients achieving resolution of arthritis or rash, as determined by SLEDAI-2K. (B) Proportion of patients achieving an SRI-4 response. (C) Least squares mean change from baseline in the SLEDAI-2K score (scores ranging from 0 to 105, higher scores indicate more severe disease). (D) Least
squares mean change from baseline in the Physician’s Global Assessment of Disease Activity, with scores ranging from 0 (0 mm) to 3 (100 mm; visual analogue scale, higher scores indicate more severe disease). The figure includes all patients in the modified intention-to-treat analysis (n=314). p values are for comparisons of baricitinib 2 mg and 4 mg with placebo. SLEDAI-2K=Systemic Lupus Erythematosus Disease Activity Index-2000. SRI-4=Systemic Lupus Erythematosus Responder Index-4.

Baseline demographics and disease activity were similar among groups (table 1; appendix). 294 (94%) patients were female and the mean age at baseline was 44 years (SD 12). At baseline, the mean duration of systemic lupus erythematosus was 11 years (SD 9), with a mean SLICC/ACR index of 0·5 (SD 0·9). Patients had a baseline mean SLEDAI-2K score of 8·9 (SD 3), mean tender joint count of 8·3 (SD 6), and mean swollen joint count of 5·3 (SD 5). Patients had a mean baseline CLASI index score of 4·2 (SD 5). 187 (60%) patients had worsening of disease, as indicated by BILAG score criteria. At baseline, 230 (73%) patients were receiving corticosteroids, 222 (71%) were receiving antimalarials, and 142 (45%) were receiving immuno- suppressants.
The proportion of patients who achieved resolution of arthritis or rash (SLEDAI-2K), the primary outcome, was significantly higher in the baricitinib 4 mg group than in the placebo group (70 [67%] of 104 patients; odds ratio [OR] vs placebo 1·8, 95% CI 1·0–3·3; p=0·0414; figure 2A, table 2). There was also
improvement in the primary outcome measure for the baricitinib 2 mg group (61 [58%] of 105 patients; OR 1·3, 95% CI 0·7–2·3; p=0·39), but this was not statistically significant. The proportion of patients achieving an SRI-4 response at week 24 was higher in the baricitinib 4 mg group than in the placebo group (67 [64%] patients; OR 2·0, 95% CI 1·2–3·6; p=0·0151); the result did not reach significance for baricitinib 2 mg (54 [51%] patients; OR 1·3, 95% CI 0·7–2·2; p=0·44; figure 2, table 2). The achievement of SRI-4 at week 24 was primarily driven by the SLEDAI-2K component (67 [64%] patients in the baricitinib 4 mg group, OR vs placebo 2·0, 95% CI 1·1–3·5, p=0·0220; 55 [52%] patients in the baricitinib 2 mg group, 1·2, 0·7–2·2, p=0·45; table 2). For the other secondary endpoints, there was a significant decrease in PGA at week 24 for patients who received baricitinib 4 mg versus placebo; results were not significant for the baricitinib 2 mg group (table 2, figure 2). Pharma- cokinetic and pharmacodynamic models showed that the majority of patients in the baricitinib 4 mg

Placebo (n=105)

Baricitinib 2 mg (n=105)

Baricitinib 4 mg (n=104)

Week 24
Week 24 Comparison with placebo (95% CI); p value
Week 24
Comparison with placebo (95% CI); p value

Primary outcome
Resolution of arthritis/rash (SLEDAI-2K)* 56 (53%) 61 (58%) 1·3 (0·7 to 2·3); p=0·39 70 (67%) 1·8 (1·0 to 3·3); p=0·0414
Secondary and exploratory outcomes
SRI-4* 50 (48%) 54 (51%) 1·3 (0·7 to 2·2); p=0·44 67 (64%) 2·0 (1·2 to 3·6); p=0·0151
≥4 point improvement in SLEDAI-2K* 51 (49%) 55 (52%) 1·2 (0·7 to 2·2); p=0·45 67 (64%) 2·0 (1·1 to 3·5); p=0·0220
No worsening (≥1A/2B) by BILAG* 80 (76%) 82 (78%) 1·2 (0·6 to 2·2); p=0·67 85 (82%) 1·4 (0·7 to 2·8); p=0·31
No worsening by PGA* 78 (74%) 82 (78%) 1·3 (0·7 to 2·5); p=0·45 84 (81%) 1·5 (0·8 to 2·9); p=0·26
LLDAS* 27 (26%) 35 (33%) 1·4 (0·8 to 2·7); p=0·25 40 (38%) 1·9 (1·0 to 3·5); p=0·0391

Flares (any severity) on the SELENA-SLEDAI Flare Index†
54 (51%)
45 (43%)
1·0 (0·6 to 1·5); p=0·88
34 (33%) 0·6 (0·4 to 0·9); p=0·0193

Severe flares on the SELENA-SLEDAI Flare Index†
12 (11%)
10 (10%)
1·0 (0·4 to 2·3); p=0·98
6 (6%)
0·5 (0·2 to 1·3); p=0·17

Least squares mean change from baseline‡
SLEDAI-2K –3·8 (0·4) –4·1 (0·4) –0·3 (–1·2 to 0·7); p=0·60 –4·4 (0·4) –0·6 (–1·6 to 0·4); p=0·24
PGA –26·3 (1·8) –25·9 (1·8) 0·4 (–4·6 to 5·4); p=0·87 –32·2 (1·8) –5·9 (–10·9 to –0·9); p=0·0218
CLASI activity score –2·8 (0·4) –1·7 (0·4) 1·1 (0·1 to 2·2); p=0·0371 –2·3 (0·4) 0·5 (–0·5 to 1·6); p=0·33
28-tender joint count –5·6 (0·4) –6·5 (0·4) –0·9 (–2·1 to 0·3); p=0·13 –6·9 (0·4) –1·3 (–2·5 to –0·1); p=0·0377
28-swollen joint count –4·6 (0·2) –4·1 (0·2) 0·5 (–0·2 to 1·1); p=0·14 –4·8 (0·2) –0·2 (–0·8 to 0·5); p=0·60
SLICC/ACR Damage Index score 0·05 (0·03) 0·07 (0·03) 0·03 (–0·05 to 0·10); p=0·53 0·07 (0·03) 0·03 (–0·05 to 0·10); p=0·52
Worst Joint Pain NRS –0·9 (0·3) –1·6 (0·3) –0·6 (1·3 to 0·1); p=0·07 –1·8 (0·3) –0·9 (–1·6 to –0·2); p=0·0157
Worst Pain NRS –0·6 (0·3) –1·2 (0·3) –0·6 (–1·3 to 0·1); p=0·10 –1·3 (0·3) –0·8 (–1·5 to 0); p=0·0403
Worst Fatigue NRS –1·2 (0·2) –1·1 (0·2) 0·1 (–0·6 to 0·7); p=0·89 –1·5 (0·2) –0·3 (–1·0 to 0·3); p=0·32
Anti-dsDNA, IU/mL 55·4 (26·8) 1·0 (27·1) –54·4 (–128·0 to 19·2); p=0·15 48·5 (26·9) –6·8 (–80·6 to 66·9); p=0·86
Complement C3, g/L 0 (0·02) 0 (0·02) –0·01 (–0·06 to 0·04); p=0·75 –0·02 (0·02) –0·02 (–0·07 to 0·02); p=0·31
Complement C4, g/L 0·01 (0·01) –0·01 (0·01) –0·01 (–0·03 to 0); p=0·18 –0·01 (0·01) –0·02 (–0·03 to 0); p=0·0314
Data are n (%) or least squares mean (SE). Data were analysed with a logistic regression model with non-responder imputation for response rates, mixed-models repeated-measure analysis or ANCOVA for least squares mean change from baseline, and Cox proportional hazard model for time to event. SLEDAI-2K=Systemic Lupus Erythematosus Disease Activity Index-2000. SRI-4=Systemic Lupus Erythematosus Responder Index-4. BILAG=British Isles Lupus Assessment Group. PGA=Physician’s Global Assessment of Disease Activity. LLDAS=Lupus Low Disease Activity Score. SELENA=Safety of Estrogens in Lupus Erythematosus National Assessment. CLASI=Cutaneous Lupus Erythematosus Disease Area and Severity Index. SLICC=Systemic Lupus International Collaborating Clinics. ACR=American College of Rheumatology. NRS=Numeric Rating Scale. dsDNA=double-stranded DNA. *Comparisons are odds ratios. †Comparisons are hazard ratios for time to event. ‡Comparisons are least squares mean difference.
Table 2: Clinical and biomarker outcomes at week 24 in the intention-to-treat population

group had exposure at the plateau of the curve (data not shown).
There were some improvements in exploratory outcomes, including patient-reported outcomes, with baricitinib treatment. A greater proportion of patients attained LLDAS with baricitinib 4 mg than with placebo at week 24 (p=0·0391); the result did not reach significance for baricitinib 2 mg (table 2). A decreased risk of any flare, as measured by the SSFI, was observed for patients receiving baricitinib 4 mg compared with placebo at week 24 (p=0·0193); the result did not reach significance for baricitinib 2 mg (table 2, figure 3). There were significant improvements in 28-tender joint count (p=0·0377), Worst Joint Pain NRS (p=0·0157), and Worst Pain NRS (p=0·0403) for patients receiving baricitinib 4 mg compared with those receiving placebo at week 24 (table 2, figure 3; appendix). Treatment with baricitinib 4 mg did not
result in significant improvements at week 24 in other exploratory assessments. Results of the exploratory analyses were not significant for the baricitinib 2 mg group compared with placebo at week 24, for most measures (table 2).
Rates of treatment discontinuation because of adverse events are shown in table 3. Serious adverse events were reported in five (5%) patients receiving placebo, 11 (10%) patients receiving baricitinib 2 mg, and ten (10%) patients receiving baricitinib 4 mg (table 3, appendix). There were no deaths, malignancies, or major adverse cardiovascular events in the study. One serious adverse event of deep-vein thrombosis was reported in the baricitinib 4 mg group, 46 days after the patient’s first dose of baricitinib, in a patient with antiphospholipid antibodies (table 3). There were more serious infections reported in the baricitinib 4 mg group (six [6%] patients) than in the 2 mg group

A Tender joint count, 0–28 B Swollen joint count, 0–28
0
–1
–2
–3
–4
–5
–6
–7
–8 p value
Baricitinib 2 mg Baricitinib 4 mg

0·7
0·6
0·5
0·4
0·3
0·2
0·1
0
04 8 12 16 20 24 28 0 4 8 12 16 20 24 28

Number at risk
Time (weeks) Time (weeks)

Placebo Baricitinib 2 mg Baricitinib 4 mg
105
105
104
78
77
86
70
68
81
65
63
76
60
61
71
52
59
67
39
41
53
0
0
0
105
105
104
96
96
100
92
90
96
90
89
93
84
87
87
81
83
85
64
63
64
0
0
0

Figure 3: Improvements in systemic lupus erythematosus disease activity, weeks 0–24
The least squares mean change from baseline in tender joint count (A) and swollen joint count (B). Time to first flare of any severity (C) and time to first severe flare (D), as defined by the SSFI. p values are for comparisons of baricitinib 2 mg and 4 mg with placebo. HR=hazard ratio. SSFI=Safety of Estrogens in Lupus Erythematosus National Assessment-Systemic Lupus Erythematosus Disease Activity Index Flare Index.

(two [2%] patients) or placebo group (one [1%] patient; table 3, appendix). There were no cases of serious or multidermatomal herpes zoster virus infection or opportunistic infection, and no reports of tuberculosis. There was one occurrence of non-serious herpes zoster virus infection in the placebo group, none in the baricitinib 2 mg group, and one in the baricitinib 4 mg group.
Mean changes from baseline and CTCAE grade increases for selected laboratory analytes from baseline to week 24 are shown in the appendix. There were modest dose-associated decreases in haemoglobin and neutrophil concentrations. There were early increases in lymphocyte concentration with baricitinib treatment, but lymphocytes returned to baseline concentrations by week 24. At week 12, there were statistically significant dose-associated increases in platelet counts, creatine phosphokinase, HDL cholesterol, and total cholesterol (appendix). There were modest dose-associated inc- reases in triglyceride concentrations in the baricitinib groups. There were no clinically meaningful differences in laboratory abnormalities. Laboratory changes were generally of low grade and consistent with changes
9–14 and four led to discontinuation of study drug (n=2 in baricitinib 2 mg and n=2 in baricitinib 4 mg).

Discussion
Use of 4 mg daily oral baricitinib treatment, in addition to standard of care therapy, was superior to placebo plus standard of care in improving signs and symptoms of active systemic lupus erythematosus.
We chose the primary objective, resolution of arthritis or rash by SLEDAI-2K, for this phase 2 trial because arthritis and rash are among the most common manifestations of systemic lupus erythematosus and are the two most frequent clinical manifestations at entry in recent clinical trials of extra-renal systemic lupus erythematosus. The SLEDAI-2K organ domain scoring is a validated, widely used outcome measure for the
15,16 Arthritis, as well as musculoskeletal pain, is a prominent symptom in patients with systemic lupus erythematosus, and improvement in musculoskeletal symptoms, assessed with several measures, is associated with improvement in
17–20 Treatment with 4 mg baricitinib showed,

by physician assessment, improvement in the proportion of patients with arthritis, as measured using the

Placebo (n=105)

Baricitinib
2 mg (n=105)

Baricitinib
4 mg (n=104)

SLEDAI-2K arthritis organ domain score (data not shown), and improvement in the proportion with joint tenderness, as measured by 28-joint examination. Treatment with 4 mg baricitinib also showed a significant reduction in the proportion of patients with Worst Joint Pain and Worst Pain when compared with placebo. In this phase 2 study, it was also important to benchmark improvement with an established composite endpoint, SRI-4, which assesses overall disease activity improvement with a global disease index. SRI-4 is a validated, widely accepted clinical trial and global regulatory endpoint for
21 Additional results supporting the findings for the primary endpoint

were the significant improvements in SRI-4 for patients in the baricitinib 4 mg group, as well as other important general measures of disease activity, such as PGA, LLDAS, SSFI flares, and patient-reported outcomes. Baricitinib 2 mg did not show any significant differences from placebo across the primary and secondary efficacy objectives studied at week 24, although there were improvements compared with placebo.
There were no significant improvements in the signs or symptoms of skin disease, whether measured using proportions of patients meeting the SLEDAI-2K muco- cutaneous organ domain score (data not shown) or using the CLASI activity score for severity, when comparing placebo with baricitinib 2 mg or 4 mg. Although the percentage of patients at baseline with mucocutaneous disease activity, measured by SLEDAI-2K, was high (84%), the overall activity score for severity, as measured by CLASI, was low. Currently, there are limited data about the use of CLASI in phase 2 or 3 clinical trials in systemic lupus erythematosus; hence, the use of CLASI remains uncertain. A mean baseline CLASI activity score of 4·2, as seen in this study, might be inadequate to discern a difference between placebo and treatment with baricitinib 2 mg or 4 mg, and the study was not powered to observe small changes in CLASI. Notably, however, JAK inhibitors have been reported to have positive effects on skin in several dermatological conditions including atopic dermatitis, psoriasis, graft-versus-host disease, and alopecia areata, and there remains reason to anticipate JAK inhibitors could have potential efficacy in treating the mucocutaneous features of systemic lupus
3,13,14,22
The safety profile of baricitinib in patients with active systemic lupus erythematosus receiving standard of care treatment was consistent with published findings in patients receiving baricitinib in other studies. No notable safety observations emerged, compared with results from studies of baricitinib for other
9–14 Compared with placebo, baricitinib treatment was associated with a higher proportion of patients who discontinued study treatment because of adverse events, and serious adverse events were more
Table 3: Adverse events

frequent with baricitinib treatment. However in this study, serious infection rates were more frequent with baricitinib 4 mg (6%) than with baricitinib 2 mg (2%) or placebo (1%). The serious infection rate with baricitinib 4 mg was similar to that reported in other trials, such as those for belimumab, which reported 5% for the placebo plus standard of care arm, and 6% for
23 The rate of serious infections might be affected by the frequent use of potent immune- modifying standard of care medication as background therapy. There were no reports of death, malignancies, major adverse cardiovascular events, tuberculosis, or serious herpes zoster.
The occurrence of thrombosis with JAK inhibition is an area of increased attention and debate. There was a single episode of deep-vein thrombosis in this study, which occurred in a patient receiving baricitinib 4 mg, who was positive for antiphospholipid antibodies. It is noteworthy that only one event was observed in the trial, despite a study population in which nearly 30% of patients had antiphospholipid antibodies (data not shown). No patients were excluded from the study based on risk factors, including history of thrombosis (unless the occurrence was recent—ie, within the previous 24 weeks).
Although there were no clinically meaningful differences in laboratory abnormalities, baricitinib treatment resulted in modest dose-associated decreases in haemoglobin neutrophil, and triglyceride concen- trations, and statistically significant increases in platelet counts, concentrations of creatine phosphokinase, HDL cholesterol, and total cholesterol.
There are limitations to the conclusions that can be drawn from this study. Notably, this trial is among few other phase 2 studies that have shown a positive
24,25 However, this timeframe limited the ability to assess long-term outcomes and damage. Further improvements in efficacy might be seen in a 52-week study. Patients were allowed to

continue existing stable background standard of care therapy, including corticosteroids, and while baseline standard of care use was balanced across groups, background therapy could potentially confound the results and contribute to the high placebo response rate. This is a recurring issue in trials for systemic lupus
26but despite this confounder, the results were positive.
Despite treatment advances, systemic lupus erythema- tosus remains a disease with unacceptable morbidity
27Existing treatments can be associated
28Since 5 it could
inhibit pathways that have been implicated as central to the pathogenesis of systemic lupus erythematosus, and we hypothesised that baricitinib could have clinical benefit in active systemic lupus erythematosus. We found that once-daily baricitinib 4 mg was associated with significant clinical improvements compared with placebo. Baricitinib treatment was not associated with any notable safety findings compared with results
9-14 These findings support further study of baricitinib as a potential therapy for patients with systemic lupus erythematosus.
Contributors
All authors contributed to the concept and design of the study, data analysis and interpretation, critical revision of the publication, and final approval to submit, and were accountable for the accuracy and integrity of the publication.
Declaration of interests
DJW has received consulting support from Amgen, Eli Lilly and Company, EMD Merck Serono, and Pfizer. RAF has received consulting support from Eli Lilly and Company. YT has received grant support, research support, or speaker bureau fees from AbbVie, Astellas, BMS, Chugai, Daiichi-Sankyo, Eisai, Eli Lilly and Company, Janssen, Kyowa-Kirin, Mitsubishi-Tanabe, MSD, Ono, Pfizer, Takeda, Sanofi, UCB, and YL Biologics. KCK has received consulting support
from Eli Lilly and Company. MM has received consulting support from Eli Lilly and Company. MAP has received consulting support from
Eli Lilly and Company. TD has received grant support, research support, consulting support, or speaker bureau fees from AbbVie, Biogen, Celgene, Chugai, Eli Lilly and Company, Janssen, MSD, Novartis, Pfizer, Roche, Sanofi, and UCB. MHC has received grant support, research support, consulting support, or speaker bureau fees from AbbVie, Eli Lilly and Company, Gilead, Janssen, Pfizer, and Roche. INB has received grant support, research support, consulting support, or speaker bureau fees from AstraZeneca, BMS, Eli Lilly and Company, Genzyme, GlaxoSmithKline, Merck Serono, and UCB. EG, AMD, JMJ, MDL, SdB, MES, and RWH are employees and stockholders of Eli Lilly and Company. TC is an employee of Eli Lilly and Company.
Acknowledgments
We would like to thank the patients who participated in the study, Nicole Byers (Eli Lilly and Company) for assisting with manuscript preparation and process support, and Julie Sherman (Eli Lilly and Company) for figure assistance. Eli Lilly or its representatives provided data, laboratory, and site monitoring services. INB is a National Institute for Health Research (NIHR) Senior Investigator and is supported by the NIHR Manchester Biomedical Research Centre. The views expressed in this publication are those of the author(s) and not necessarily those of the National Health Service, the NIHR, or the Department of Health.

References
1Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus.
Nat Rev Rheumatol 2016; 12: 716–30.
2Wahren-Herlenius M, Dorner T. Immunopathogenic mechanisms of systemic autoimmune disease. Lancet 2013; 382: 819–31.
3Schwartz 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: 843–62.
4Hoffman RW, Merrill JT, Alarcon-Riquelme MM, 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: 643–54.
5Fridman JS, Scherle PA, Collins R, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol 2010; 184: 5298–307.
6Tan EM, Cohen AS, Fries JF, et al. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1982; 25: 1271–77.
7Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1997; 40: 1725.
8Petri M, Orbai AM, Alarcon GS, et al. Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus.
Arthritis Rheum 2012; 64: 2677–86.
9Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med 2016;
374: 1243–52.
10Dougados M, van der Heijde D, Chen YC, et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study.
Ann Rheum Dis 2017; 76: 88–95.
11Fleischmann R, Schiff M, van der Heijde D, et al. Baricitinib, methotrexate, or combination in patients with rheumatoid arthritis and no or limited prior disease-modifying antirheumatic drug treatment. Arthritis Rheumatol 2017; 69: 506–17.
12Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med 2017; 376: 652–62.
13Guttman-Yassky E, Silverberg JI, Nemoto O, et al. Baricitinib in adult patients with moderate-to-severe atopic dermatitis: a phase 2 parallel, double-blinded, randomized placebo-controlled
multiple-dose study. J Am Acad Dermatol 2018; published online Feb 1. DOI:10.1016/j.jaad.2018.01.018.
14Papp KA, Menter MA, Raman M, et al. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor,
in patients with moderate-to-severe psoriasis. Br J Dermatol 2016; 174: 1266–76.
15Manzi S, Sanchez-Guerrero J, Merrill JT, et al. Effects of
belimumab, a B lymphocyte stimulator-specific inhibitor, on disease activity across multiple organ domains in patients with systemic lupus erythematosus: combined results from two phase III trials. Ann Rheum Dis 2012; 71: 1833–38.
16Kalunian KC, Urowitz MB, Isenberg D, et al. Clinical trial parameters that influence outcomes in lupus trials that use the systemic lupus erythematosus responder index.
Rheumatology (Oxford) 2018; 57: 125–33.
17Urowitz M, Gladman DD, Ibanez D, et al. Changes in quality of life in the first 5 years of disease in a multicenter cohort of patients
with systemic lupus erythematosus. Arthritis Care Res (Hoboken) 2014; 66: 1374–79.
18Urowitz MB, Gladman DD, Ibanez D, et al. Evolution of disease burden over five years in a multicenter inception systemic lupus erythematosus cohort. Arthritis Care Res (Hoboken) 2012; 64: 132–37.
19Jolly M, Pickard AS, Block JA, et al. Disease-specific patient reported outcome tools for systemic lupus erythematosus.
Semin Arthritis Rheum 2012; 42: 56–65.
20Moorthy LN, Baldino ME, Kurra V, et al. Relationship between health-related quality of life, disease activity and disease damage in a prospective international multicenter cohort of childhood onset systemic lupus erythematosus patients. Lupus 2017; 26: 255–65.

21Furie RA, Petri MA, Wallace DJ, et al. Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum 2009; 61: 1143–51.
22MacDonald KPA, Betts BC, Couriel D. Emerging therapeutics for the control of chronic graft-versus-host disease.
Biol Blood Marrow Transplant 2018; 24: 19–26.
23GlaxoSmithKline. Belimumab prescribing information 2017. https://www.gsksource.com/pharma/content/dam/
GlaxoSmithKline/US/en/Prescribing_Information/Benlysta/pdf/
BENLYSTA-PI-MG-IFU-COMBINED.PDF (accessed May 31, 2018).
24Wallace 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: 183–90.

Baricitinib
25Furie R, Khamashta M, Merrill JT, et al. Anifrolumab,
an anti-interferon-alpha receptor monoclonal antibody, in
moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol 2017; 69: 376–86.
26Merrill JT, Manzi S, Aranow C, et al. Lupus community panel proposals for optimising clinical trials: 2018. Lupus Sci Med 2018; 5: e000258.
27Yurkovich M, Vostretsova K, Chen W, Avina-Zubieta JA. Overall and cause-specific mortality in patients with systemic lupus erythematosus: a meta-analysis of observational studies.
Arthritis Care Res (Hoboken) 2014; 66: 608–16.
28Thong B, Olsen NJ. Systemic lupus erythematosus diagnosis and management. Rheumatology (Oxford) 2017; 56 (suppl 1): i3–13.