You are on page 1of 10

Articles

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
Lancet 2018; 392: 222–31 Background Patients with systemic lupus erythematosus have substantial unmet medical need. Baricitinib is an oral
See Comment page 190 selective Janus kinase (JAK)1 and JAK2 inhibitor that we hypothesised might have therapeutic benefit in patients with
Division of Rheumatology, systemic lupus erythematosus.
Cedars-Sinai Medical Center,
University of California at Methods In this double-blind, multicentre, randomised, placebo-controlled, 24-week phase 2 study, patients were
Los Angeles, Los Angeles, CA,
USA (Prof D J Wallace MD);
recruited from 78 centres in 11 countries. Eligible patients were aged 18 years or older, had a diagnosis of systemic
Division of Rheumatology, lupus erythematosus, and had active disease involving skin or joints. We randomly assigned patients (1:1:1) to receive
Zucker School of Medicine at once-daily baricitinib 2 mg, baricitinib 4 mg, or placebo for 24 weeks. The primary endpoint was the proportion of
Hofstra, Northwell, New York,
patients achieving resolution of arthritis or rash at week 24, as defined by Systemic Lupus Erythematosus Disease
NY, USA (Prof R A Furie MD);
The First Department of Activity Index-2000 (SLEDAI-2K). Efficacy and safety analyses included all patients who received at least one dose of
Internal Medicine, School study drug. This study is registered with ClinicalTrials.gov, number NCT02708095.
of Medicine, University of
Occupational and
Findings Between March 24, 2016, and April 27, 2017, 314 patients were randomly assigned to receive placebo
Environmental Health,
Kitakyushu, Japan (n=105), baricitinib 2 mg (n=105), or baricitinib 4 mg (n=104). At week 24, resolution of SLEDAI-2K arthritis or rash
(Prof Y Tanaka MD); Division of 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;
Rheumatology, University 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
of California at San Diego
reported in 68 (65%) patients in the placebo group, 75 (71%) patients in the baricitinib 2 mg group, and 76 (73%)
School of Medicine, La Jolla,
CA, USA (Prof K C Kalunian MD); patients in the baricitinib 4 mg group. Serious adverse events were reported in ten (10%) patients receiving
Division of Rheumatology, baricitinib 4 mg, 11 (10%) receiving baricitinib 2 mg, and five (5%) receiving placebo; no deaths were reported.
University of Pisa, Pisa, Italy Serious infections were reported in six (6%) patients with baricitinib 4 mg, two (2%) with baricitinib 2 mg, and
(Prof M Mosca MD); Division of
one (1%) with placebo.
Rheumatology, Johns Hopkins
University School of Medicine,
Baltimore, MD, USA Interpretation The baricitinib 4 mg dose, but not the 2 mg dose, significantly improved the signs and symptoms of
(Prof M A Petri MD); Division of active systemic lupus erythematosus in patients who were not adequately controlled despite standard of care therapy,
Rheumatology, Charite
Universitätsmedizin Berlin,
with a safety profile consistent with previous studies of baricitinib. This study provides the foundation for future
Berlin, Germany phase 3 trials of JAK1/2 inhibition with baricitinib as a new potential oral therapy for systemic lupus erythematosus.
(Prof T Dörner MD); Centro de
Investigación Clínica de Funding Eli Lilly and Company.
Morelia SC, Morelia, México
(Prof M H Cardiel MD); Arthritis
Research UK Centre for Copyright © 2018 Elsevier Ltd. All rights reserved.
Epidemiology, Faculty of
Biology, Medicine and Health, Introduction which has been approved for the treatment of moderately
The University of Manchester
and NIHR Manchester
Systemic lupus erythematosus is a multisystem, chronic to severely active rheumatoid arthritis in adults in over
Biomedical Research Centre, autoimmune disease characterised by the presence of 40 countries, including European countries, the USA,
Manchester University antibodies directed at self-antigens, and broad immune and Japan. This molecular mechanism of action suggests
Hospitals NHS Foundation dysregulation.1 It is believed that abnormalities of both that baricitinib might inhibit cytokines central to the
Trust, Manchester Academic
Health Science Centre,
the innate and adaptive arms of the immune system, dysregulated innate and adaptive immune function
Manchester, UK interconnected by a positive feedback loop, contribute to observed in systemic lupus erythematosus.
(Prof I N Bruce MD); Lilly disease pathogenesis.2 We designed this international, double-blind, random­
Biotechnology Center, ised, placebo-controlled, phase 2 trial to assess the
Many key cytokines implicated in the pathogenesis
San Diego, CA, USA
(M D Linnik PhD); and Eli Lilly of systemic lupus erythematosus are dependent on efficacy, safety, and tolerability of oral baricitinib (2 mg or
and Company, Indianapolis, IN, activation of Janus kinases (JAKs) for intracellular 4 mg) once-daily in patients with active systemic lupus
USA (E Gomez MS, signalling.1,3,4 The JAK family of cytoplasmic protein erythematosus, who were receiving standard background
T Carmack MS,
tyrosine kinases mediates the signalling of several therapy.
A M DeLozier MPH,
J M Janes MD, S de Bono MD, pro-inflammatory cytokines, such as type l inter­
M E Silk PharmD, ferons (JAK1/tyrosine kinase [TYK] 2), interleukin 6 Methods
Prof R W Hoffman, DO) (JAK1/JAK2/TYK2), and interleukin 12 and interleukin 23 Study design and participants
(JAK2/TYK2).3 Baricitinib is an orally administered In this double-blind, randomised, placebo-controlled
selective and reversible inhibitor of JAK1 and JAK2,5 trial, 79 investigators assessed patients at 78 centres in

222 www.thelancet.com Vol 392 July 21, 2018


Articles

Correspondence to:
Research in context Prof Daniel J Wallace, Division of
Rheumatology, Cedars-Sinai
Evidence before this study signal transducer and activator of transcription (STAT) in Medical Center, Los Angeles, CA
We searched PubMed using the terms “systemic lupus cytokine-mediated immune signalling and inflammation. This 90048, USA
erythematosus”, “treatment”, and “JAK inhibitor” for articles in pathway includes JAK/STAT-mediated signalling through danielwallac@gmail.com

English published up to March 1, 2016, regardless of article type 1 interferons, which have been shown to have an
type. We considered previous clinical trials of investigational important role in the pathogenesis of systemic lupus
medication in systemic lupus erythematosus, as well as erythematosus.
previous clinical trials of the Janus kinase (JAK)1/2 inhibitor,
Added value of this study
baricitinib, before doing this study. Current treatment
In this phase 2 trial, baricitinib improved the signs and
regimens for the management of systemic lupus
symptoms of active disease in patients with systemic lupus
erythematosus have resulted in a reduction of morbidity and
erythematosus who were receiving standard background
mortality over previous decades; however, many patients still
therapy. Baricitinib 4 mg treatment resulted in a greater
have incompletely controlled disease and can progress to
proportion of patients achieving resolution of arthritis, as
end-stage organ involvement. There are a small number of
defined by Systemic Lupus Erythematosus Disease Activity
medications approved for the treatment of systemic lupus
Index-2000, than with placebo. The safety profile of baricitinib
erythematosus and other medications are used off-label;
was consistent with other drugs used to treat active systemic
collectively, these include antimalarials, non-steroidal
lupus erythematosus.
anti-inflammatory drugs, aspirin, corticosteroids, azathioprine,
mycophenolate, cyclophosphamide, methotrexate, Implications of all the available evidence
belimumab, and rituximab. Because of their limited efficacy, These findings support the evidence that JAK/STAT signalling
these drugs are often used in multiple combinations, rather could have a central role in the pathogenesis of systemic
than as single agents, resulting in added risk of toxicity. New lupus erythematosus. To our knowledge, this is the first study
treatment options with an acceptable safety profile that reduce to show clinical benefit of JAK inhibition in the treatment of
disease activity, reduce flares, delay organ damage, and reduce systemic lupus erythematosus. This work provides the
the requirement for corticosteroids and cytotoxic drugs are foundation for additional study of JAK1/2 inhibition with
urgently needed for patients with systemic lupus baricitinib as a potentially effective oral treatment option for
erythematosus. Additionally, the need for treatments that active systemic lupus erythematosus in patients who have
improve quality of life, including control of pain and fatigue, not achieved adequate disease control with available
has been articulated by patients and physicians. Clinical and standard of care therapies.
laboratory studies have showed a central role for JAK and

11 countries in Asia, Europe, North America, and South corticosteroid dose was limited to 20 mg or less of
America. The study protocol is available from the prednisone per day (or equivalent), and was required to
sponsor. be stable for 2 weeks before randomisation; increases in
Eligible participants were 18 years or older and had dose were not permitted after randomisation. Decreases
been diagnosed with systemic lupus erythematosus in cor­ticosteroid dose were permitted from baseline to
at least 24 weeks before screening by fulfilling four week 16. No changes in corticosteroid dose were
or more of the revised American College of Rheumatology permitted between week 16 and week 24. No increases
(ACR) criteria for classification of sys­temic lupus erythe­ in antimalarials or immunosuppressants were allowed
matosus, or the 2012 Systemic Lupus Erythematosus at any time. Key exclusion criteria included active
International Collaborating Clinics (SLICC) classification severe lupus nephritis, active severe CNS lupus, recent
criteria.6–8 At baseline, patients were required to have clinically serious infection, and selected laboratory
a positive antinuclear antibody (HEp-2 titre ≥1:80), a abnormalities. A full list of exclusion criteria are shown
positive anti-double-stranded DNA (anti-dsDNA; in the appendix. See Online for appendix
≥30 IU/mL), IgG INOVA QUANTA Lite SC ELISA The study was done in accordance with the ethical
(INOVA Diagnostics, San Diego, CA, USA), a Systemic principles of the Declaration of Helsinki and Good
Lupus Erythematosus Disease Activity Index-2000 Clinical Practice guidelines. All investigation sites
(SLEDAI-2K) score of 4 or greater based on clinical received approval from the appropriate authorised
manifestations, or active arthritis or rash as defined by institutional review board or ethics committee. All
the SLEDAI-2K. Study drug was added to existing patients provided written informed consent before the
stable background standard of care ther­apy, which could study-related procedures were done.
be non-steroidal anti-inflam­matory drugs, corticosteroids,
a single antimalarial (such as chloroquine or hydroxy­ Randomisation and masking
chloroquine), or a single immuno­ suppressant (such We used a computer-generated random sequence to
as azathioprine, metho­trexate, or mycophenolate). The allocate patients (1:1:1) to placebo, baricitinib 2 mg, or

www.thelancet.com Vol 392 July 21, 2018 223


Articles

baricitinib 4 mg. Patients were stratified according to disease activity scores (no new BILAG A score or no
disease severity (SLEDAI-2K score <10 or ≥10), anti- more than one new BILAG B score), and no worsening
dsDNA status (positive or negative), and region (USA, (defined as an increase of ≥0·3 points [10 mm] from
Europe, Asia, or rest of world). Patients and investigators baseline) in the Physician’s Global Assessment of
were masked to allocation. Daily doses were provided Disease Activity (PGA). Other secondary endpoints
at each visit in two bottles containing identical were the proportion of patients achieving a reduction
investigational product tablets supplied by Eli Lilly and of 4 points or more from baseline in SLEDAI-2K score,
Company. All study investigators remained masked until and change from baseline in SLEDAI-2K total score
the week 24 analysis. and PGA at week 24. We analysed plasma baricitinib
concen­trations using a population pharmaco­ kinetic
Procedures approach, to characterise the exposure response for
All interventions were given orally. Patients were SLEDAI-2K and SRI-4 across 24 weeks (data not
instructed to take one tablet from each bottle each day shown). Exploratory assessments were change from
(one tablet of placebo, one tablet of study drug). Safety baseline in 28-tender joint count and 28-swollen joint
outcomes (clinical laboratory tests, vital signs, and other count, Cutaneous Lupus Erythematosus Disease
safety assessments) and efficacy outcomes were assessed Area and Severity Index (CLASI) activity score, and
on scheduled study visits (baseline and weeks 2, 4, 8, 12, SLICC/ACR Damage index; risk of flares of any severity,
16, 20, and 24) during the double-blind treatment period, and severe flares on the Safety of Estrogens in
as indicated in the protocol schedule of events. Patients Lupus Erythematosus National Assessment (SELENA)-
were also assessed approximately 28 days after receiving SLEDAI Flare Index (SSFI); and proportion of patients
their last dose of study drug. achieving Lupus Low Disease Activity Score (LLDAS).
We also recorded corticosteroid doses and measured
Outcomes changes in serological markers, including anti-dsDNA
The primary outcome was the proportion of patients and complement component (C) 3 and C4. We assessed
achieving resolution of arthritis or rash, as defined by the change in the patient-reported outcomes Worst Joint
SLEDAI-2K, at week 24. Pain numeric rating scale (NRS), Worst Pain NRS, and
The proportion of patients achieving a Systemic Worst Fatigue NRS (appendix pp 20–23).
Lupus Erythematosus Responder Index-4 (SRI-4) Incidence and severity of all adverse events were
response at week 24 was a secondary objective. SRI-4 recorded between baseline and week 24, and up to
response is defined as a reduction of at least 4 points 30 days after treatment had ended, and the Quick
from baseline in SLEDAI-2K score, no worsening in Inventory of Depressive Symptomatology Self-Rated-16
British Isles Lupus Assessment Group (BILAG) A or B was assessed. We used the National Institutes of
Health Common Terminology Criteria for Adverse
Events (CTCAE) to describe laboratory abnormalities
414 patients screened (version 4.03).

100 excluded
Statistical analysis
81 screening failure With approximately 100 patients per treatment group,
11 withdrew this study had approximately 81% power to detect a
4 physician’s decision
4 other 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
314 randomly assigned
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
105 assigned to placebo 105 assigned to baricitinib 2 mg 104 assigned to baricitinib 4 mg χ² test of equal proportions, using nQuery Advisor
(version 7.0).
22 discontinued 19 discontinued 18 discontinued
We analysed the primary, secondary, and exploratory
4 adverse event 10 adverse event 11 adverse event endpoints according to the prespecified statistical
5 withdrew 3 withdrew 1 protocol violation analysis plan.
2 physician’s decision 3 physician’s 4 withdrew
9 lack of efficacy decision 2 physician’s We made no multiplicity adjustments. We analysed
2 other 3 lack of efficacy decision efficacy and patient-reported outcomes (PGA, Worst
Pain NRS, Worst Joint Pain NRS, Worst Fatigue NRS) in
83 completed double-blind 86 completed double-blind 86 completed double-blind the modified intention-to-treat population, defined as all
treatment period treatment period treatment period 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,

224 www.thelancet.com Vol 392 July 21, 2018


Articles

with treatment, region, baseline disease severity


Placebo Baricitinib Baricitinib
(SLEDAI-2K score <10 vs ≥10), and baseline anti-dsDNA (n=105) 2 mg 4 mg
status (positive or negative) in the model. We made (n=105) (n=104)
treatment comparisons of continuous efficacy endpoints Mean age, years 44·9 (12·8) 43·2 (11·0) 45·0 (12·4)
using pairwise comparisons in mixed models repeated Mean time since onset of 9·7 (7·7) 11·8 (9·1) 11·5 (10·3)
measures for outcomes with multiple post-baseline systemic lupus
values, or ANCOVA for outcomes with a single post- erythematosus, years
baseline value. The mixed models repeated measures Concomitant medications
model included treatment, baseline score, baseline Corticosteroids 77 (73%) 79 (75%) 74 (71%)
disease severity (SLEDAI-2K score <10 vs ≥10), baseline Mean prednisone dose 7·9 (4·6) 8·7 (5·8) 10·5 (17·4)
anti-dsDNA status (positive or negative), region, visit, (or equivalent), mg/day

and the interaction of treatment-by-visit as fixed Prednisone dose 36/77 (47%) 40/79 (51%) 41/74 (55%)
(or equivalent),
factors. The ANCOVA model included treatment, ≥7·5 mg/day
baseline score, baseline disease severity (SLEDAI-2K Antimalarials 75 (71%) 71 (68%) 76 (73%)
score <10 vs ≥10), baseline anti-dsDNA status (positive Immunosuppressants 45 (43%) 47 (45%) 50 (48%)
or negative), and region as explanatory variables. We Methotrexate 13 (12%) 17 (16%) 13 (13%)
made treatment comparisons for the time to first Azathioprine 15 (14%) 10 (10%) 11 (11%)
systemic lupus erythematosus flare (SSFI) using a Cox
Mycophenolate mofetil 11 (10%) 10 (10%) 16 (15%)
proportional hazards model with treatment group,
Non-steroidal 27 (26%) 29 (28%) 32 (31%)
baseline disease severity (SLEDAI-2K score <10 vs ≥10), anti-inflammatory drug
baseline anti-dsDNA status (positive or negative), and Mean SLEDAI-2K score* 8·9 (2·9) 8·8 (3·4) 9·0 (3·3)
region fitted as explanatory variables. We assessed safety SLEDAI-2K score ≥10 43 (41%) 35 (33%) 44 (42%)
in patients who received at least one dose of study SLEDAI-2K organ system involvement
drug and who were not lost to follow-up before the CNS 3 (3%) 1 (1%) 2 (2%)
first post-baseline visit. Adverse events were inclusive Vascular 1 (1%) 4 (4%) 3 (3%)
of the treatment period and up to 30 days after treat­ Musculoskeletal 93 (89%) 93 (89%) 96 (92%)
ment, and laboratory abnormality summaries included Renal 9 (9%) 9 (9%) 7 (7%)
the treatment period and up 60 days after treatment.
Mucocutaneous 90 (86%) 82 (78%) 92 (88%)
We used the Fisher exact test for all adverse events,
Cardiovascular and 2 (2%) 1 (1%) 1 (1%)
discontinuation, and other categorical safety data. We respiratory
analysed continuous safety data using ANCOVA, Immunological 62 (59%) 63 (60%) 64 (62%)
adjusting for baseline value and treatment. All analyses Constitutional 2 (2%) 2 (2%) 0
were assessed at a two-sided α of 0·05. Haematological 13 (12%) 9 (9%) 5 (5%)
For categorical efficacy analyses, patients were con­ ≥1 A or ≥2 B BILAG scores† 62 (59%) 56 (53%) 69 (66%)
sidered non-responders at each visit if they were not Mean Physician’s Global 49·5 (16·9) 48·8 (15·8) 51·7 (16·0)
responders at that visit; permanently discontinued study Assessment score‡
treatment at any time before that visit for any reason; Mean CLASI activity score§ 4·9 (5·7) 3·8 (5·4) 4·0 (3·4)
required initiation or increase (from baseline) in dose Mean tender joint count 7·7 (5·8) 8·7 (6·6) 8·5 (6·2)
of non-steroidal anti-inflamma­ tory drugs (for 30 or Mean swollen joint count 5·3 (4·7) 5·2 (4·7) 5·5 (4·2)
more consecutive days), cortico­steroids, antimalarials, Mean SLICC/ACR Damage 0·59 (0·97) 0·44 (0·68) 0·40 (0·88)
or immunosuppressants, after randomisation; or had Index score¶
missing data at that visit. No imputation was needed Data are mean (SD), n (%), or n/N (%). Additional baseline characteristics and
with mixed models repeated measures for analysing the disease activity are described in the appendix (pp 24–25). SLEDAI-2K=Systemic
continuous efficacy endpoints. This study is registered Lupus Erythematosus Disease Activity Index-2000. BILAG=British Isles Lupus
Assessment Group. CLASI=Cutaneous Lupus Erythematosus Disease Area and
with ClincialTrials.gov, number NCT02708095. Severity Index. SLICC=Systemic Lupus International Collaborating Clinics.
ACR=American College of Rheumatology. *Scores range from 0 to 105, with
Role of the funding source higher values indicating more severe disease. †A score indicates severe disease
and B score indicates moderate disease. ‡Scores range from 0 (0 mm) to
The funder of the study had a role in study design, data
3 (100 mm; visual analogue scale), with higher values indicating more severe
analysis, data collection, data interpretation, or writing of disease. §Scores range from 0 to 70, with higher values indicating greater
the report. The corresponding author had full access to severity. ¶Scores range from 0 to 45, with higher values indicating
all the data in the study and had final responsibility for more damage.

the decision to submit for publication. Table 1: Baseline characteristics and disease activity

Results
Between March 24, 2016, and April 27, 2017, 314 patients (n=104); 255 (81%) completed the 24-week double-blind
were randomly assigned to receive once-daily placebo treatment period (figure 1). No patients were excluded
(n=105), baricitinib 2 mg (n=105), or baricitinib 4 mg from the efficacy and safety analyses.

www.thelancet.com Vol 392 July 21, 2018 225


Articles

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


100 Placebo 100
Baricitinib 2 mg
Baricitinib 4 mg
80 80
Patients (%)

60 60

40 40

20 20

0 0
0 2 4 8 12 16 20 24 0 2 4 8 12 16 20 24
p value
Baricitinib 2 mg 0·97 0·57 0·54 0·91 0·15 0·38 0·39 0·72 0·34 0·42 0·67 0·25 0·48 0·44
Baricitinib 4 mg 0·56 0·18 0·56 0·26 0·06 0·0293 0·0414 0·55 0·12 0·48 0·33 0·34 0·09 0·0151

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


0 0
Least squares mean change from

–1·0
–10

–2·0
baseline

–20
–3·0

–30
–4·0

–5·0 –40
0 2 4 8 12 16 20 24 0 2 4 8 12 16 20 24
Time (weeks) Time (weeks)
p value
Baricitinib 2 mg 0·77 0·72 0·68 0·98 0·28 0·97 0·60 0·38 0·98 0·60 0·31 0·11 0·37 0·87
Baricitinib 4 mg 0·31 0·66 0·98 0·88 0·30 0·42 0·24 0·41 0·34 0·26 0·08 0·41 0·08 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 improvement in the primary outcome measure for the
similar among groups (table 1; appendix). 294 (94%) baricitinib 2 mg group (61 [58%] of 105 patients;
patients were female and the mean age at baseline was OR 1·3, 95% CI 0·7–2·3; p=0·39), but this was not
44 years (SD 12). At baseline, the mean duration of statistically significant. The proportion of patients
systemic lupus erythematosus was 11 years (SD 9), with achieving an SRI-4 response at week 24 was higher in
a mean SLICC/ACR index of 0·5 (SD 0·9). Patients the baricitinib 4 mg group than in the placebo group
had a baseline mean SLEDAI-2K score of 8·9 (SD 3), (67 [64%] patients; OR 2·0, 95% CI 1·2–3·6; p=0·0151);
mean tender joint count of 8·3 (SD 6), and mean the result did not reach significance for baricitinib
swollen joint count of 5·3 (SD 5). Patients had a mean 2 mg (54 [51%] patients; OR 1·3, 95% CI 0·7–2·2;
baseline CLASI index score of 4·2 (SD 5). 187 (60%) p=0·44; figure 2, table 2). The achievement of SRI-4 at
patients had worsening of disease, as indicated by week 24 was primarily driven by the SLEDAI-2K
BILAG score criteria. At baseline, 230 (73%) patients component (67 [64%] patients in the baricitinib 4 mg
were receiving corticosteroids, 222 (71%) were receiving group, OR vs placebo 2·0, 95% CI 1·1–3·5, p=0·0220;
antimalarials, and 142 (45%) were receiving immuno­ 55 [52%] patients in the baricitinib 2 mg group, 1·2,
suppressants. 0·7–2·2, p=0·45; table 2). For the other secondary
The proportion of patients who achieved resolution endpoints, there was a significant decrease in PGA at
of arthritis or rash (SLEDAI-2K), the primary outcome, week 24 for patients who received baricitinib 4 mg
was significantly higher in the baricitinib 4 mg group versus placebo; results were not significant for the
than in the placebo group (70 [67%] of 104 patients; baricitinib 2 mg group (table 2, figure 2). Pharma­
odds ratio [OR] vs placebo 1·8, 95% CI 1·0–3·3; cokinetic and pharma­codynamic models showed that
p=0·0414; figure 2A, table 2). There was also the majority of patients in the baricitinib 4 mg

226 www.thelancet.com Vol 392 July 21, 2018


Articles

Placebo Baricitinib 2 mg (n=105) Baricitinib 4 mg (n=104)


(n=105)
Week 24 Week 24 Comparison with placebo Week 24 Comparison with placebo
(95% CI); p value (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 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
SELENA-SLEDAI Flare Index†
Severe flares on the SELENA-SLEDAI 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
Flare Index†
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 result in significant improve­ments at week 24 in other
not shown). exploratory assessments. Results of the exploratory
There were some improvements in exploratory analyses were not significant for the baricitinib 2 mg
outcomes, including patient-reported outcomes, with group compared with placebo at week 24, for most
baricitinib treatment. A greater proportion of patients measures (table 2).
attained LLDAS with baricitinib 4 mg than with placebo Rates of treatment discontinuation because of adverse
at week 24 (p=0·0391); the result did not reach events are shown in table 3. Serious adverse events
significance for baricitinib 2 mg (table 2). A decreased were reported in five (5%) patients receiving placebo,
risk of any flare, as measured by the SSFI, was observed 11 (10%) patients receiving baricitinib 2 mg, and
for patients receiving baricitinib 4 mg compared ten (10%) patients receiving baricitinib 4 mg (table 3,
with placebo at week 24 (p=0·0193); the result did not appendix). There were no deaths, malignancies, or
reach significance for baricitinib 2 mg (table 2, major adverse cardiovascular events in the study. One
figure 3). There were significant improvements in serious adverse event of deep-vein thrombosis was
28-tender joint count (p=0·0377), Worst Joint Pain NRS reported in the baricitinib 4 mg group, 46 days after the
(p=0·0157), and Worst Pain NRS (p=0·0403) for patient’s first dose of baricitinib, in a patient with
patients receiving baricitinib 4 mg compared with antiphospholipid antibodies (table 3). There were more
those receiving placebo at week 24 (table 2, figure 3; serious infections reported in the baricitinib 4 mg
appendix). Treatment with baricitinib 4 mg did not group (six [6%] patients) than in the 2 mg group

www.thelancet.com Vol 392 July 21, 2018 227


Articles

A Tender joint count, 0–28 B Swollen joint count, 0–28


0 0
Placebo
Least squares change from baseline
–1 Baricitinib 2 mg –1
–2 Baricitinib 4 mg
–2
–3
–4 –3
–5
–4
–6
–5
–7
–8 –6
0 2 4 8 12 16 20 24 0 2 4 8 12 16 20 24
p value
Baricitinib 2 mg 0·36 0·29 0·08 0·66 0·51 0·38 0·13 0·70 0·99 0·10 0·0170 0·0054 0·0201 0·14
Baricitinib 4 mg 0·96 0·15 0·14 0·16 0·22 0·0235 0·0377 0·45 0·83 0·77 0·86 0·73 0·38 0·60

C Time to flare, any severity D Time to severe flare


0·7 Baricitinib 2 mg HR 1·0 (95% CI 0·6–1·5); p=0·88 0·25 Baricitinib 2 mg HR 1·0 (95% CI 0·4–2·3); p=0·97
Baricitinib 4 mg HR 0·6 (95% CI 0·4–0·9); p=0·019 0·612 Baricitinib 4 mg HR 0·5 (95% CI 0·2–1·3); p=0·17 0·221
0·6
0·20
0·5
Cumulative event

0·463
0·4 0·15
0·337 0·120
0·3 0·10
0·2 0·060
Placebo 0·05
0·1 Baricitinib 2 mg
Baricitinib 4 mg
0 0
0 4 8 12 16 20 24 28 0 4 8 12 16 20 24 28
Time (weeks) Time (weeks)
Number at risk
Placebo 105 78 70 65 60 52 39 0 105 96 92 90 84 81 64 0
Baricitinib 2 mg 105 77 68 63 61 59 41 0 105 96 90 89 87 83 63 0
Baricitinib 4 mg 104 86 81 76 71 67 53 0 104 100 96 93 87 85 64 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; observed for other conditions,9–14 and four led to
table 3, appendix). There were no cases of serious discontinuation of study drug (n=2 in baricitinib 2 mg
or multi­dermatomal herpes zoster virus infection or and n=2 in baricitinib 4 mg).
opportunistic infection, and no reports of tuberculosis.
There was one occurrence of non-serious herpes zoster Discussion
virus infection in the placebo group, none in the Use of 4 mg daily oral baricitinib treatment, in addition
baricitinib 2 mg group, and one in the baricitinib to standard of care therapy, was superior to placebo plus
4 mg group. standard of care in improving signs and symptoms of
Mean changes from baseline and CTCAE grade active systemic lupus erythematosus.
increases for selected laboratory analytes from baseline We chose the primary objective, resolution of arthritis
to week 24 are shown in the appendix. There were or rash by SLEDAI-2K, for this phase 2 trial because
modest dose-associated decreases in haemoglobin and arthritis and rash are among the most common
neutrophil concentrations. There were early increases manifestations of systemic lupus erythematosus and are
in lymphocyte concentration with baricitinib treatment, the two most frequent clinical manifestations at entry
but lymphocytes returned to baseline concentrations by in recent clinical trials of extra-renal systemic lupus
week 24. At week 12, there were statistically significant erythematosus. The SLEDAI-2K organ domain scoring
dose-associated increases in platelet counts, creatine is a validated, widely used outcome measure for the
phosphokinase, HDL cholesterol, and total cholesterol study of systemic lupus erythematosus.15,16 Arthritis, as
(appendix). There were modest dose-associated inc­ well as musculoskeletal pain, is a prominent symptom
reases in triglyceride concentrations in the baricitinib in patients with systemic lupus erythematosus, and
groups. There were no clinically meaningful differences improvement in musculoskeletal symptoms, assessed
in laboratory abnormalities. Laboratory changes were with several measures, is associated with improvement in
generally of low grade and consistent with changes quality of life.17–20 Treatment with 4 mg baricitinib showed,

228 www.thelancet.com Vol 392 July 21, 2018


Articles

by physician assessment, improve­ment in the proportion


Placebo Baricitinib Baricitinib
of patients with arthritis, as measured using the (n=105) 2 mg (n=105) 4 mg (n=104)
SLEDAI-2K arthritis organ domain score (data not
Discontinuation from 4 (4%) 10 (10%) 11 (11%)
shown), and improvement in the proportion with joint study treatment because
tenderness, as measured by 28-joint examination. of an adverse event
Treatment with 4 mg baricitinib also showed a significant Any adverse event after 68 (65%) 75 (71%) 76 (73%)
reduction in the proportion of patients with Worst Joint the start of therapy
Pain and Worst Pain when compared with placebo. Infections 41 (39%) 47 (45%) 47 (45%)
In this phase 2 study, it was also important to bench­mark Serious infections 1 (1%) 2 (2%) 6 (6%)
improvement with an established composite endpoint, Herpes zoster virus 1 (1%) 0 1 (1%)
SRI-4, which assesses overall disease activity improvement infection

with a global disease index. SRI-4 is a validated, widely Deep-vein thrombosis 0 0 1 (1%)
accepted clinical trial and global regulatory endpoint for Serious adverse events 5 (5%) 11 (10%) 10 (10%)
studies of systemic lupus erythematosus.21 Additional Data are n (%). Adverse events that occurred between baseline and week 24, and
results supporting the findings for the primary endpoint up to 30 days after treatment, are shown.

were the significant improvements in SRI-4 for patients Table 3: Adverse events
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. frequent with baricitinib treatment. However in this
Baricitinib 2 mg did not show any significant differences study, serious infection rates were more frequent with
from placebo across the primary and secondary efficacy baricitinib 4 mg (6%) than with baricitinib 2 mg (2%) or
objectives studied at week 24, although there were placebo (1%). The serious infection rate with baricitinib
improvements compared with placebo. 4 mg was similar to that reported in other trials, such as
There were no significant improvements in the signs those for belimumab, which reported 5% for the
or symptoms of skin disease, whether measured using placebo plus standard of care arm, and 6% for
proportions of patients meeting the SLEDAI-2K muco­ belimumab.23 The rate of serious infections might be
cutaneous organ domain score (data not shown) or using affected by the frequent use of potent immune-
the CLASI activity score for severity, when comparing modifying standard of care medication as background
placebo with baricitinib 2 mg or 4 mg. Although the therapy. There were no reports of death, malignancies,
percentage of patients at baseline with mucocutaneous major adverse cardiovascular events, tuberculosis, or
disease activity, measured by SLEDAI-2K, was high serious herpes zoster.
(84%), the overall activity score for severity, as measured The occurrence of thrombosis with JAK inhibition is
by CLASI, was low. Currently, there are limited data an area of increased attention and debate. There was a
about the use of CLASI in phase 2 or 3 clinical trials in single episode of deep-vein thrombosis in this study,
systemic lupus erythematosus; hence, the use of CLASI which occurred in a patient receiving baricitinib 4 mg,
remains uncertain. A mean baseline CLASI activity score who was positive for antiphospholipid antibodies. It is
of 4·2, as seen in this study, might be inadequate to noteworthy that only one event was observed in the trial,
discern a difference between placebo and treatment with despite a study population in which nearly 30% of
baricitinib 2 mg or 4 mg, and the study was not powered patients had antiphospholipid antibodies (data not
to observe small changes in CLASI. Notably, however, shown). No patients were excluded from the study based
JAK inhibitors have been reported to have positive effects on risk factors, including history of thrombosis (unless
on skin in several dermatological conditions including the occurrence was recent—ie, within the previous
atopic dermatitis, psoriasis, graft-versus-host disease, 24 weeks).
and alopecia areata, and there remains reason to Although there were no clinically meaningful
anticipate JAK inhibitors could have potential efficacy in differences in laboratory abnormalities, baricitinib
treating the mucocutaneous features of systemic lupus treatment resulted in modest dose-associated decreases
erythematosus.3,13,14,22 in haemoglobin neutrophil, and triglyceride concen­
The safety profile of baricitinib in patients with active trations, and statistically significant increases in platelet
systemic lupus erythematosus receiving standard of counts, concentrations of creatine phosphokinase, HDL
care treatment was consistent with published findings cholesterol, and total cholesterol.
in patients receiving baricitinib in other studies. No There are limitations to the conclusions that can be
notable safety observations emerged, compared with drawn from this study. Notably, this trial is among few
results from studies of baricitinib for other other phase 2 studies that have shown a positive
conditions.9–14 Compared with placebo, baricitinib outcome within 24 weeks.24,25 However, this timeframe
treatment was associated with a higher proportion of limited the ability to assess long-term outcomes and
patients who discontinued study treatment because of damage. Further improvements in efficacy might be
adverse events, and serious adverse events were more seen in a 52-week study. Patients were allowed to

www.thelancet.com Vol 392 July 21, 2018 229


Articles

continue existing stable background standard of care References


therapy, including corticosteroids, and while baseline 1 Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the
immunopathogenesis of systemic lupus erythematosus.
standard of care use was balanced across groups, Nat Rev Rheumatol 2016; 12: 716–30.
background therapy could potentially confound the 2 Wahren-Herlenius M, Dorner T. Immunopathogenic mechanisms of
results and contribute to the high placebo response rate. systemic autoimmune disease. Lancet 2013; 382: 819–31.
3 Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O’Shea JJ.
This is a recurring issue in trials for systemic lupus JAK inhibition as a therapeutic strategy for immune and
erythematosus,26 but despite this confounder, the results inflammatory diseases. Nat Rev Drug Discov 2017; 16: 843–62.
were positive. 4 Hoffman RW, Merrill JT, Alarcon-Riquelme MM, et al.
Gene expression and pharmacodynamic changes in
Despite treatment advances, systemic lupus erythema­ 1,760 systemic lupus erythematosus patients from two phase III trials
tosus remains a disease with unacceptable morbidity of BAFF blockade with tabalumab. Arthritis Rheumatol 2017;
and mortality.27 Existing treatments can be associated 69: 643–54.
5 Fridman JS, Scherle PA, Collins R, et al. Selective inhibition of JAK1
with incomplete efficacy or substantial toxicity.28 Since and JAK2 is efficacious in rodent models of arthritis: preclinical
baricitinib has selectivity for JAK1 and JAK2,5 it could characterization of INCB028050. J Immunol 2010; 184: 5298–307.
inhibit pathways that have been implicated as central to 6 Tan EM, Cohen AS, Fries JF, et al. The 1982 revised criteria for the
the pathogenesis of systemic lupus erythematosus, and classification of systemic lupus erythematosus. Arthritis Rheum
1982; 25: 1271–77.
we hypothesised that baricitinib could have clinical 7 Hochberg MC. Updating the American College of Rheumatology
benefit in active systemic lupus erythematosus. We revised criteria for the classification of systemic lupus
found that once-daily baricitinib 4 mg was associated erythematosus. Arthritis Rheum 1997; 40: 1725.
8 Petri M, Orbai AM, Alarcon GS, et al. Derivation and validation of
with significant clinical improvements compared with the Systemic Lupus International Collaborating Clinics
placebo. Baricitinib treatment was not associated with classification criteria for systemic lupus erythematosus.
any notable safety findings compared with results Arthritis Rheum 2012; 64: 2677–86.
9 Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients
from baricitinib studies for other conditions.9-14 These with refractory rheumatoid arthritis. N Engl J Med 2016;
findings support further study of baricitinib as a 374: 1243–52.
potential therapy for patients with systemic lupus 10 Dougados M, van der Heijde D, Chen YC, et al. Baricitinib in
erythematosus. patients with inadequate response or intolerance to conventional
synthetic DMARDs: results from the RA-BUILD study.
Contributors Ann Rheum Dis 2017; 76: 88–95.
All authors contributed to the concept and design of the study, data 11 Fleischmann R, Schiff M, van der Heijde D, et al. Baricitinib,
analysis and interpretation, critical revision of the publication, and final methotrexate, or combination in patients with rheumatoid arthritis
approval to submit, and were accountable for the accuracy and integrity of and no or limited prior disease-modifying antirheumatic drug
the publication. treatment. Arthritis Rheumatol 2017; 69: 506–17.
12 Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus
Declaration of interests placebo or adalimumab in rheumatoid arthritis. N Engl J Med 2017;
DJW has received consulting support from Amgen, Eli Lilly and 376: 652–62.
Company, EMD Merck Serono, and Pfizer. RAF has received 13 Guttman-Yassky E, Silverberg JI, Nemoto O, et al. Baricitinib in
consulting support from Eli Lilly and Company. YT has received grant adult patients with moderate-to-severe atopic dermatitis: a phase 2
support, research support, or speaker bureau fees from AbbVie, parallel, double-blinded, randomized placebo-controlled
Astellas, BMS, Chugai, Daiichi-Sankyo, Eisai, Eli Lilly and Company, multiple-dose study. J Am Acad Dermatol 2018; published online
Janssen, Kyowa-Kirin, Mitsubishi-Tanabe, MSD, Ono, Pfizer, Takeda, Feb 1. DOI:10.1016/j.jaad.2018.01.018.
Sanofi, UCB, and YL Biologics. KCK has received consulting support 14 Papp KA, Menter MA, Raman M, et al. A randomized phase 2b trial
from Eli Lilly and Company. MM has received consulting support from of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor,
Eli Lilly and Company. MAP has received consulting support from in patients with moderate-to-severe psoriasis. Br J Dermatol 2016;
Eli Lilly and Company. TD has received grant support, research 174: 1266–76.
support, consulting support, or speaker bureau fees from AbbVie, 15 Manzi S, Sanchez-Guerrero J, Merrill JT, et al. Effects of
Biogen, Celgene, Chugai, Eli Lilly and Company, Janssen, MSD, belimumab, a B lymphocyte stimulator-specific inhibitor, on disease
Novartis, Pfizer, Roche, Sanofi, and UCB. MHC has received grant activity across multiple organ domains in patients with systemic
lupus erythematosus: combined results from two phase III trials.
support, research support, consulting support, or speaker bureau fees
Ann Rheum Dis 2012; 71: 1833–38.
from AbbVie, Eli Lilly and Company, Gilead, Janssen, Pfizer, and
16 Kalunian KC, Urowitz MB, Isenberg D, et al. Clinical trial
Roche. INB has received grant support, research support, consulting
parameters that influence outcomes in lupus trials that use the
support, or speaker bureau fees from AstraZeneca, BMS, Eli Lilly and systemic lupus erythematosus responder index.
Company, Genzyme, GlaxoSmithKline, Merck Serono, and UCB. EG, Rheumatology (Oxford) 2018; 57: 125–33.
AMD, JMJ, MDL, SdB, MES, and RWH are employees and 17 Urowitz M, Gladman DD, Ibanez D, et al. Changes in quality of life
stockholders of Eli Lilly and Company. TC is an employee of Eli Lilly in the first 5 years of disease in a multicenter cohort of patients
and Company. with systemic lupus erythematosus. Arthritis Care Res (Hoboken)
Acknowledgments 2014; 66: 1374–79.
We would like to thank the patients who participated in the study, 18 Urowitz MB, Gladman DD, Ibanez D, et al. Evolution of disease
Nicole Byers (Eli Lilly and Company) for assisting with manuscript burden over five years in a multicenter inception systemic lupus
erythematosus cohort. Arthritis Care Res (Hoboken) 2012; 64: 132–37.
preparation and process support, and Julie Sherman (Eli Lilly and
Company) for figure assistance. Eli Lilly or its representatives provided 19 Jolly M, Pickard AS, Block JA, et al. Disease-specific patient reported
outcome tools for systemic lupus erythematosus.
data, laboratory, and site monitoring services. INB is a National
Semin Arthritis Rheum 2012; 42: 56–65.
Institute for Health Research (NIHR) Senior Investigator and is
20 Moorthy LN, Baldino ME, Kurra V, et al. Relationship between
supported by the NIHR Manchester Biomedical Research Centre. The
health-related quality of life, disease activity and disease damage in
views expressed in this publication are those of the author(s) and not a prospective international multicenter cohort of childhood onset
necessarily those of the National Health Service, the NIHR, or the systemic lupus erythematosus patients. Lupus 2017; 26: 255–65.
Department of Health.

230 www.thelancet.com Vol 392 July 21, 2018


Articles

21 Furie RA, Petri MA, Wallace DJ, et al. Novel evidence-based 25 Furie R, Khamashta M, Merrill JT, et al. Anifrolumab,
systemic lupus erythematosus responder index. Arthritis Rheum an anti-interferon-alpha receptor monoclonal antibody, in
2009; 61: 1143–51. moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol
22 MacDonald KPA, Betts BC, Couriel D. Emerging therapeutics for 2017; 69: 376–86.
the control of chronic graft-versus-host disease. 26 Merrill JT, Manzi S, Aranow C, et al. Lupus community panel
Biol Blood Marrow Transplant 2018; 24: 19–26. proposals for optimising clinical trials: 2018. Lupus Sci Med 2018;
23 GlaxoSmithKline. Belimumab prescribing information 2017. 5: e000258.
https://www.gsksource.com/pharma/content/dam/ 27 Yurkovich M, Vostretsova K, Chen W, Avina-Zubieta JA. Overall and
GlaxoSmithKline/US/en/Prescribing_Information/Benlysta/pdf/ cause-specific mortality in patients with systemic lupus
BENLYSTA-PI-MG-IFU-COMBINED.PDF (accessed May 31, 2018). erythematosus: a meta-analysis of observational studies.
24 Wallace DJ, Kalunian K, Petri MA, et al. Efficacy and safety of Arthritis Care Res (Hoboken) 2014; 66: 608–16.
epratuzumab in patients with moderate/severe active systemic 28 Thong B, Olsen NJ. Systemic lupus erythematosus diagnosis and
lupus erythematosus: results from EMBLEM, a phase IIb, management. Rheumatology (Oxford) 2017; 56 (suppl 1): i3–13.
randomised, double-blind, placebo-controlled, multicentre study.
Ann Rheum Dis 2014; 73: 183–90.

www.thelancet.com Vol 392 July 21, 2018 231

You might also like