throbber
Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`Clinical and epidemiological research
`
`EXTENDED REPORT
`Efficacy and safety of an interleukin 6 monoclonal
`antibody for the treatment of systemic lupus
`erythematosus: a phase II dose-ranging randomised
`controlled trial
`Daniel J Wallace,1 Vibeke Strand,2 Joan T Merrill,3 Serghei Popa,4 Alberto J Spindler,5
`Alicia Eimon,6 Michelle Petri,7 Josef S Smolen,8 Joseph Wajdula,9 Jared Christensen,10
`Cheryl Li,10 Annette Diehl,9 Michael S Vincent,10 Jean Beebe,10 Paul Healey,11
`Sudhakar Sridharan12
`
`ABSTRACT
`Objectives This phase II trial evaluated the efficacy
`and safety of an interleukin (IL) 6 monoclonal antibody
`for systemic lupus erythematosus (SLE).
`Methods Patients with active disease were randomised
`to placebo or PF-04236921 10 mg, 50 mg or 200 mg,
`subcutaneously, every 8 weeks with stable background
`therapy. SLE Responder Index (SRI-4; primary end point)
`and British Isles Lupus Assessment Group-based
`Composite Lupus Assessment (BICLA) were assessed at
`week 24. Post hoc analysis identified an enriched
`population based upon planned univariate analyses.
`Results 183 patients received treatment ( placebo,
`n=45; 10 mg, n=45; 50 mg, n=47; 200 mg, n=46).
`The 200 mg dose was discontinued due to safety
`findings and not included in the primary efficacy
`analysis. The SRI-4 response rates were not significant
`for any dose compared with placebo; however, the
`BICLA response rate was significant for 10 mg
`( p=0.026). The incidence of severe flares was
`significantly reduced with 10 mg (n=0) and 50 mg (n=2)
`combined versus placebo (n=8; p<0.01). In patients
`with greater baseline disease activity (enriched
`population), the SRI-4 ( p=0.004) and BICLA ( p=0.012)
`response rates were significantly different with 10 mg
`versus placebo. Four deaths (200 mg, n=3; 10 mg, n=1)
`occurred. The most frequently reported adverse events
`included headache, nausea and diarrhoea.
`Conclusions PF-04236921 was not significantly
`different from placebo for the primary efficacy end point
`in patients with SLE. Evidence of an effect with 10 mg
`was seen in a post hoc analysis. Safety was acceptable
`for doses up to 50 mg as the 200 mg dose was
`discontinued due to safety findings.
`Trial registration number NCT01405196;
`Pre-results.
`
`INTRODUCTION
`Systemic lupus erythematosus (SLE) is a chronic,
`systemic autoimmune disease associated with het-
`erogeneous immunological and clinical manifesta-
`tions, leading to sporadic and unpredictable flares
`of multisystem inflammation. SLE has a substantial
`detrimental impact on health-related quality of life
`
`(HRQOL) and participation in daily activities,
`including work within and outside the home.1
`The pleiotropic cytokine interleukin (IL) 6 has a
`range of biological effects and is primarily pro-
`duced by monocytes, fibroblasts and endothelial
`cells, and by T cells, B cells, keratinocytes and
`mesangial cells.2 IL-6 acts alone or alongside other
`cytokines to promote differentiation of B cells into
`immunoglobulin-producing cells, as well as prolif-
`eration and differentiation of T cells.3
`The spontaneous production of autoantibodies
`plays an important role in SLE pathogenesis,4 5
`which has been attributed to B cell hyperactivity.6
`Studies suggest that IL-6 is critically involved in the
`B cell hyperactivity of SLE, and may also mediate
`tissue damage.7 Moreover, IL-6 regulates hepatic
`synthesis of acute phase reactants,
`including the
`inflammatory biomarker C reactive protein (CRP),8
`and is involved in the differentiation of T helper 17
`(Th17) cells, which are understood to be pivotal in
`the induction of autoimmune diseases.9 Consistent
`with these observations, IL-6 production is higher in
`patients with active SLE than in healthy individuals,
`and serum IL-6 levels, as well as IL-6 levels mea-
`sured in skin lesions and the kidney, correlate with
`disease activity.10–14
`Targeting IL-6 signalling may offer a novel thera-
`peutic approach for SLE, supported by promising
`clinical and serological responses observed with the
`soluble IL-6 receptor inhibitor tocilizumab in a
`small, open-label phase I study.15 In this study, 16
`patients with mild-to-moderate SLE received one of
`three dose regimens of tocilizumab every 2 weeks
`for 12 weeks. Improvements in disease activity were
`seen and antidouble-stranded DNA (anti-dsDNA)
`levels decreased. It was noted that there was a clear
`dose-related reduction in complement
`levels and
`neutrophil count.
`PF-04236921 is a fully human immunoglobulin
`G2 monoclonal antibody that binds and neutralises
`IL-6 as demonstrated in the early phase I trials.16
`Here, we report the results of a phase II dose-
`ranging randomised controlled trial to assess the effi-
`cacy and safety of PF-04236921 in patients with
`active SLE.
`
`Handling editor Tore K Kvien
`
` (cid:377) Additional material is
`published online only. To view
`please visit the journal online
`(h t t p : / / d x . d o i . o r g / 1 0 . 1 1 3 6 /
`a n n r h e u m d i s - 2 0 1 6 - 2 0 9 6 6 8 )
`
`For numbered affiliations see
`end of article.
`
`Correspondence to
`Dr Daniel J Wallace, Division of
`Rheumatology, Cedars-Sinai
`Medical Center, Los Angeles,
`CA, 90048, USA;
`danielwallac@gmail.com
`
`Received 5 April 2016
`Revised 30 June 2016
`Accepted 11 July 2016
`Published Online First
`26 September 2016
`
`To cite: Wallace DJ, Strand V,
`Merrill JT, et al. Ann Rheum
`Dis 2017;76:534–542.
`
`534
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`Lassen - Exhibit 1029, p. 1
`
`

`

`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`METHODS
`Study design
`Following a 4-week screening period, patients were randomised
`(1:1:1:1) to receive placebo or PF-04236921 10 mg, 50 mg or
`200 mg. Randomisation was performed through an interactive
`voice response system according to a computer-generated ran-
`domisation schedule, with stratification by baseline disease activ-
`ity (SLE Disease Activity Index (SLEDAI)-2K score 6–9 vs ≥10;
`anti-dsDNA antibodies greater than vs less than the upper limit
`of normal (120 IU/mL)). Doses were administered as two sub-
`cutaneous injections at day 1, week 8 and week 16 over a
`24-week double-blind treatment phase, during which efficacy
`and safety data were recorded. Patients subsequently entered a
`28-week follow-up period.
`Consistent with entry criteria, stable (≥30 days before base-
`line) standard-of-care SLE medications including immunosup-
`pressives,
`antimalarials
`and corticosteroids were
`allowed.
`Corticosteroid doses were limited to prednisone ≤25 mg/day at
`baseline. Supplemental corticosteroids were allowed at baseline
`to no more than 10 mg/day above prestudy doses, but had to be
`tapered to the baseline dose by day 28. Subsequent dose increases
`were not allowed thereafter, and tapering was recommended
`based upon clinical
`judgement during the treatment phase,
`however no changes were permitted during the last 4 weeks of
`the 24-week treatment phase. Rescue medications for disease
`worsening were allowed during the treatment phase at investiga-
`tor discretion; however, such patients were considered treatment
`failures and non-responders for the efficacy analyses.
`
`Entry criteria
`Eligible patients were aged 18–75 years, had a clinical diagnosis
`of SLE according to American College of Rheumatology criteria,
`were serologically positive based upon current or historical posi-
`tive test results for antinuclear antibodies (ANA, human epithelial
`type 2; titre ≥1:80) and/or anti-dsDNA antibodies (>120 IU/L),
`and had active disease (SLEDAI-2K score of ≥6 and British Isles
`Lupus Assessment Group (BILAG) 2004 A disease in ≥1 organ
`system or BILAG B disease in ≥2 organ systems if no level A
`disease activity was present). Detailed exclusion criteria are
`included in the online supplementary material.
`
`End points
`The primary efficacy end point was the proportion of patients
`achieving the SLE Responder
`Index (SRI-4) at week 24.
`
`Figure 1 Patient disposition.
`*Treatment group terminated
`prematurely. AE, adverse event.
`
`Clinical and epidemiological research
`
`Responders were defined by a ≥4-point reduction in SLEDAI-2K
`score, no new BILAG A or two new BILAG B organ domain
`scores, and no significant deterioration (<0.3-point increase) in
`Physician’s Global Assessment score compared with baseline. In
`addition, responders could not be treatment failures, defined as:
`new or increased use of corticosteroids after day 28; new or
`increased use of immunosuppressives and/or antimalarials; death
`or hospitalisation due to worsening SLE; treatment discontinuation
`due to SLE; or a flare that would interfere with trial participation.
`Key secondary efficacy end points assessed at week 24 in-
`cluded the proportion of patients achieving BILAG-based
`Composite Lupus Assessment
`(BICLA) responses (responders
`defined by BILAG 2004 improvement (all A scores at baseline
`improved to B/C/D and all B scores improved to C or D), no
`new BILAG A scores and ≤1 new B score, no worsening of
`modified SLEDAI-2K score (modified to omit ‘low complement’
`and ‘leukopoenia’ parameters), no significant deterioration in
`Patient’s Global Assessment score (<10% worsening), and no
`treatment
`failure); ≥10%, ≥30% or ≥50% reductions
`in
`anti-dsDNA antibody levels; mean changes in complement
`levels (C3 and C4); the proportion of patients whose cortico-
`steroid dose was reduced by ≥25% from baseline, and to
`≤7.5 mg/day, for at least one visit up to and including week 24;
`mean changes in 36-item Short Form Health Survey (SF-36;
`V.2) summary and domain scores; mean changes in European
`Quality of Life 5 Dimensions (EQ-5D) visual analogue scale
`(VAS) scores; and mean changes in Functional Assessment of
`Chronic Illness Therapy (FACIT)-Fatigue scores.
`Exploratory efficacy end points at week 24 included the
`incidence of
`severe SLE flares using modified Safety of
`Estrogens
`in Lupus Erythematosus: National Assessment-
`SLEDAI Flare Index (SFI) or BILAG (defined for this proto-
`col as one new BILAG A or two new BILAG B organ domain
`scores).
`Additional details on pharmacokinetic, pharmacodynamic,
`biomarker and safety assessments are provided in the online
`supplementary material.
`
`Post hoc analysis of enriched population
`Prespecified descriptive univariate analyses were performed
`on the following baseline parameters to identify a population
`with an increased likelihood of achieving efficacy: age, gender,
`race, ethnicity, baseline SLEDAI-2K score, corticosteroid use,
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`535
`
`Lassen - Exhibit 1029, p. 2
`
`

`

`Clinical and epidemiological research
`
`Table 1 Patient demographics and disease characteristics at baseline
`
`Mean age, years (SD)
`
`Female, n (%)
`
`Race, n (%)
`
`White
`
`Black
`
`Asian
`
`Other
`Mean BMI, kg/m2 (SD)
`
`Mean SLE duration, years (SD)
`
`Mean SLEDAI-2K score (SD)
`
`SLEDAI-2K ≥10, n (%)
`
`BILAG 2004
`
`Placebo (n=45)
`
`10 mg (n=45)
`
`50 mg (n=47)
`
`200 mg (n=46)
`
`42.3 (13.0)
`
`38 (84.4)
`
`33 (73.3)
`
`4 (8.9)
`
`1 (2.2)
`
`7 (15.6)
`
`29.6 (7.1)
`
`9.1 (6.9)
`
`9.5 (2.2)
`
`22 (48.9)
`
`39.9 (11.5)
`
`43 (95.6)
`
`37 (82.2)
`
`3 (6.7)
`
`1 (2.2)
`
`4 (8.9)
`
`28.6 (6.9)
`
`7.9 (8.1)
`
`9.6 (2.7)
`
`22 (48.9)
`
`38.3 (10.5)
`
`43 (93.6)
`
`36 (76.6)
`
`8 (17.0)
`
`0 (0.0)
`
`3 (6.4)
`
`27.4 (6.9)
`
`7.5 (6.0)
`
`9.0 (2.7)
`
`19 (40.4)
`
`41.3 (11.3)
`
`43 (93.5)
`
`33 (71.7)
`
`9 (19.6)
`
`0 (0.0)
`
`4 (8.7)
`
`29.9 (8.1)
`
`8.6 (6.1)
`
`10.1 (3.9)
`
`22 (47.8)
`
`19 (42.2)
`
`16 (34.0)
`
`25 (54.3)
`
`BILAG A in ≥1 organ system, n (%)
`
`BILAG B in ≥2 organ systems, n (%)
`
`Mean BILAG numerical score (SD)
`
`BILAG A or B in organ domain, n (%)
`
`Cardiorespiratory
`
`Constitutional
`
`Gastrointestinal
`
`Haematological
`
`Mucocutaneous
`
`Musculoskeletal
`
`Neuropsychiatric
`
`Ophthalmic
`
`Renal
`
`20 (44.4)
`
`25 (55.6)
`
`18.4 (3.3)
`
`0 (0.0)
`
`3 (6.7)
`
`0 (0.0)
`
`1 (2.2)
`
`39 (86.7)
`
`44 (97.8)
`
`0 (0.0)
`
`1 (2.2)
`
`1 (2.2)
`
`27 (60.0)
`
`18.5 (4.1)
`
`33 (70.2)
`
`18.3 (4.1)
`
`26 (56.5)
`
`20.0 (5.2)
`
`2 (4.4)
`
`3 (6.7)
`
`0 (0.0)
`
`0 (0.0)
`
`39 (86.7)
`
`45 (100.0)
`
`0 (0.0)
`
`0 (0.0)
`
`1 (2.2)
`
`4 (8.5)
`
`2 (4.3)
`
`0 (0.0)
`
`0 (0.0)
`
`41 (87.2)
`
`46 (97.9)
`
`0 (0.0)
`
`1 (2.1)
`
`2 (4.3)
`
`6 (13.0)
`
`1 (2.2)
`
`0 (0.0)
`
`0 (0.0)
`
`37 (80.4)
`
`45 (97.8)
`
`5 (10.9)
`
`0 (0.0)
`
`3 (6.5)
`
`1.8 (0.3)
`
`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`Mean PhGA score (SD)
`
`Serologically positive (ANA ≥1:80 and/or anti-dsDNA >120 IU/mL), n (%)
`
`Anti-dsDNA >ULN (120 IU/mL), n (%)
`
`Detectable anti-dsDNA (≥28 IU/mL), n (%)
`
`Low C3 (<90 mg/dL), n (%)
`
`Low C4 (<16 mg/dL), n (%)
`
`Corticosteroid use, n (%)
`
`Corticosteroids >7.5 mg/day, n (%)
`
`Immunosuppressive use, n (%)
`
`Antimalarial use, n (%)
`
`Mean SF-36 score (SD)
`
`PCS score
`
`1.6 (0.4)
`
`36 (80.0)
`
`13 (28.9)
`
`27 (60.0)
`
`13 (28.9)
`
`10 (22.2)
`
`31 (68.9)
`
`23 (51.1)
`
`20 (44.4)
`
`34 (75.6)
`
`34.6 (10.2)
`
`1.7 (0.4)
`
`35 (77.8)
`
`7 (15.6)
`
`28 (62.2)
`
`12 (26.7)
`
`9 (20.0)
`
`32 (71.1)
`
`14 (31.1)
`
`18 (40.0)
`
`35 (77.8)
`
`1.6 (0.4)
`
`38 (80.9)
`
`10 (21.3)
`
`28 (59.6)
`
`11 (23.4)
`
`5 (10.6)
`
`36 (76.6)
`
`24 (51.1)
`
`21 (44.7)
`
`34 (72.3)
`
`32 (71.1)
`
`11 (23.9)
`
`21 (45.7)
`
`12 (26.7)*
`
`7 (15.6)*
`
`34 (73.9)
`
`18 (39.1)
`
`23 (50.0)
`
`26 (56.5)
`
`34.0 (8.0)
`
`39.6 (11.8)
`
`34.5 (8.4)
`
`42.7 (9.9)
`
`33.9 (9.6)
`
`39.2 (12.2)
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`MCS score
`
`Physical functioning
`
`Role physical
`
`Body pain
`
`General health
`
`Vitality
`
`Social functioning
`
`Role emotional
`
`Mental health
`
`Mean EQ-5D VAS score (SD)
`
`Mean FACIT-Fatigue score (SD)
`
`39.9 (9.7)
`
`51.4 (27.8)
`
`43.8 (26.8)
`
`39.5 (22.5)
`
`34.4 (18.7)
`
`35.0 (22.1)
`
`51.7 (25.2)
`
`61.3 (24.6)
`
`57.7 (18.7)
`
`56.7 (22.9)
`
`26.0 (11.8)
`
`48.6 (25.2)
`
`38.5 (24.8)
`
`37.8 (20.3)
`
`34.6 (19.0)
`
`38.9 (21.4)
`
`54.4 (24.5)
`
`56.5 (30.0)
`
`55.0 (20.6)
`
`55.2 (21.5)
`
`25.9 (11.4)
`
`51.3 (24.3)
`
`47.1 (21.7)
`
`39.9 (20.8)
`
`33.9 (11.9)
`
`41.2 (17.8)
`
`57.7 (22.1)
`
`61.2 (27.1)
`
`63.2 (16.2)
`
`57.6 (18.5)
`
`29.4 (10.3)
`
`45.0 (24.3)
`
`42.4 (25.9)
`
`36.3 (19.6)
`
`36.8 (18.7)
`
`37.2 (18.6)
`
`49.7 (24.8)
`
`53.6 (27.9)
`
`57.6 (21.4)
`
`49.8 (20.4)
`
`24.7 (11.6)
`
`*n=45 for the 200 mg group.
`ANA, antinuclear antibody; BILAG, British Isles Lupus Assessment Group; BMI, body mass index; dsDNA, double-stranded DNA; EQ-5D, European Quality of Life 5 Dimensions; FACIT,
`Functional Assessment of Chronic Illness Therapy; MCS, mental component summary; PCS, physical component summary; PhGA, Physician’s Global Assessment; SF-36, 36-item Short Form
`Health Survey; SLE, systemic lupus erythematosus; SLEDAI, Systemic Lupus Erythematosus Disease Activity Index; ULN, upper limit of normal; VAS, visual analogue scale.
`
`immunosuppressive use, anti-dsDNA antibodies, ANA and
`hypocomplementaemia. A post hoc analysis was conducted to
`evaluate whether influential covariates could define a more
`responsive population.
`
`Statistical analyses
`The primary analysis of SRI-4 responders at week 24 was based
`upon a generalised linear mixed model (GLMM) with stratifica-
`tion variables as covariates for each active treatment versus
`
`536
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`Lassen - Exhibit 1029, p. 3
`
`

`

`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`placebo comparison. Forty-five patients per group provided
`approximately 80% power to detect a 25% difference in SRI-4
`responder rates between PF-04236921 and placebo at week 24
`using a one-sided α of 0.05. No multiple comparison adjust-
`ments were made for multiple doses. Similar modelling was
`used for the secondary analysis of BICLA responders at week
`24. GLMM analyses for SRI and BICLA included all available
`data before each patient completed week 24 or discontinued.
`The model likelihood was adjusted for missed visits by discon-
`tinued patients based on patients with similar data patterns.
`The incidences of severe SFI flares and BILAG flares were com-
`pared across treatment groups using Fisher’s exact test. Mean
`changes in EQ-5D VAS, FACIT-Fatigue and SF-36 scores for each
`active treatment group were compared with placebo using an ana-
`lysis of covariance model, adjusted for baseline scores.
`Efficacy analyses were performed on the modified intent-
`to-treat population, which included all randomised patients who
`received at least one dose of study drug. After the 200 mg dose
`was stopped, prior to unblinding, the statistical analysis plan
`was amended to exclude this dose group from the primary ana-
`lysis. The safety population included all patients who received at
`least one dose of study drug.
`
`RESULTS
`Patients
`Of 423 screened patients, 183 were randomised and received
`treatment (figure 1).
`
`Clinical and epidemiological research
`
`Baseline characteristics were balanced between groups (table 1).
`Approximately 78% of patients were serologically positive at
`baseline; the remaining patients had historically positive ANA
`or anti-dsDNA, with current active SLE confirmed by independ-
`ent experts (based upon clinical history and SLE serologies).
`Rates of discontinuation due to adverse events (AEs), with-
`drawal of consent and loss to follow-up were generally low
`across groups (figure 1). Premature termination of the 200 mg
`dose accounted for 22 of the 50 study discontinuations. Based
`upon an assessment of fatalities due to serious infections and
`thromboembolic events, the data monitoring committee advised
`discontinuation of the 200 mg dose group (see safety outcomes
`for further details). Therefore, the primary efficacy outcomes
`are based upon a full analysis set of 137 patients who received
`placebo, 10 mg or 50 mg.
`
`Efficacy outcomes
`SRI-4 response rates (GLMM) at week 24 were numerically
`greater for 10 mg versus placebo; however, statistical signifi-
`cance was not achieved ( p=0.076; figure 2). There were signifi-
`cantly more BICLA responders
`for 10 mg versus placebo
`( p=0.026; figure 2). Neither outcome was significant for 50 mg
`versus placebo. A sensitivity analysis was performed for the SRI
`and BICLA using a logistic regression model; details are
`included in the online supplementary materials. The observed
`proportion of responders in the 200 mg group who had com-
`pleted week 24 prior to premature termination (n=22) was
`
`Figure 2 SRI-4 and BICLA responder rates at week 24 (A) in the total population, and (B) in the enriched population (GLMM model). *p<0.05 vs
`placebo; **p<0.01 vs placebo. BICLA, British Isles Lupus Assessment Group-based Composite Lupus Assessment; GLMM, generalised linear mixed
`model; SRI, Systemic Lupus Erythematosus Responder Index.
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`537
`
`Lassen - Exhibit 1029, p. 4
`
`

`

`Clinical and epidemiological research
`
`Table 2 Summary of efficacy outcomes at week 24
`
`SRI response rate, n/N (%)*
`
`OR vs placebo (90% CI)
`
`p Value
`
`BICLA response rate, n/N (%)*
`
`OR vs placebo (90% CI)
`
`p Value
`
`Treatment failures, n/N (%)
`
`p Value
`
`Patients with disease flares, n/N (%)
`
`Severe BILAG flares (new BILAG A or two new BILAG B organ domain scores)
`
`Severe SFI flares
`
`Proportion of patients with reductions in anti-dsDNA from baseline, n/N (%)‡
`
`≥10% reduction
`
`≥30% reduction
`
`≥50% reduction
`
`Mean change in C3 concentration from baseline, g/L (SD)§
`
`Mean change in C4 concentration from baseline, g/L (SD)§
`
`Placebo (n=45)
`
`10 mg (n=45)
`
`50 mg (n=47)
`
`16/42 (40.1)
`
`20/35 (59.9)
`
`14/36 (39.2)
`
`11/42 (25.1)
`
`11/45 (24.4)
`
`5/45 (11.1)
`
`8/45 (17.8)
`
`7/17 (41.2)
`
`3/16 (18.8)
`
`1/15 (6.7)
`
`2.2 (0.89 to 5.62)
`
`0.96 (0.38 to 2.41)
`
`0.076
`
`18/35 (49.7)
`
`0.528
`
`15/36 (40.5)
`
`2.95 (1.18 to 7.41)
`
`2.03 (0.82 to 5.06)
`
`0.026
`
`1/45 (2.2)
`
`0.005
`
`2/43 (4.7)
`
`0/43 (0.0)†
`
`9/15 (60.0)
`
`7/14 (50.0)
`
`4/14 (28.6)
`
`0.10
`
`4/47 (8.5)
`
`0.031
`
`0/44 (0.0)
`2/44 (4.5)†
`
`11/18 (61.1)
`
`6/18 (33.3)
`
`1/16 (6.3)
`
`−0.021 (0.176)
`
`0.0002 (0.0417)
`
`−0.100 (0.163)
`
`−0.169 (0.161)
`
`−0.0096 (0.0516)
`
`−0.0551 (0.0491)
`
`4/15 (26.7)
`
`5/24 (20.8)
`
`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`Patients whose corticosteroid dose was reduced by ≥25% from baseline, and to ≤7.5 mg/day,
`for at least one visit up to and including week 24, n/N (%)¶
`
`2/23 (8.7)
`
`LS mean change in SF-36 score from baseline (SE)
`
`PCS score
`
`MCS score
`
`Physical functioning
`
`Role physical
`
`Body pain
`
`General health
`
`Vitality
`
`Social functioning
`
`Role emotional
`
`Mental health
`
`LS mean change in EQ-5D VAS score from baseline (SE)
`
`LS mean change in FACIT-Fatigue score from baseline (SE)
`
`Placebo (n=45)
`
`10 mg (n=43)
`
`50 mg (n=46)
`
`3.08 (1.2)
`
`2.95 (1.4)
`
`4.87 (3.4)
`
`10.62 (3.4)
`
`7.92 (3.3)
`
`7.01 (2.5)
`
`6.42 (3.0)
`
`7.62 (3.5)
`
`6.49 (3.4)
`
`4.90 (2.5)
`
`5.99 (2.8)
`
`2.82 (1.5)
`
`6.04 (1.2)
`
`2.94 (1.4)
`
`10.96 (3.5)
`
`15.28 (3.5)
`
`13.38 (3.3)
`
`12.53 (2.6)
`
`10.30 (3.1)
`
`6.78 (3.5)
`
`6.65 (3.5)
`
`6.96 (2.6)
`
`10.30 (2.9)
`
`4.43 (1.6)
`
`5.67 (1.2)
`
`2.12 (1.4)
`
`12.49 (3.4)
`
`16.06 (3.3)
`
`13.94 (3.2)
`
`5.15 (2.5)
`
`7.45 (3.0)
`
`9.58 (3.4)
`
`10.80 (3.3)
`
`2.72 (2.5)
`
`6.18 (2.7)
`
`3.47 (1.5)
`
`Bold italic text denotes changes that were greater than the minimum clinically important difference (SF-36 PCS and MCS >2.5-point change from baseline;17 SF-36 domain scores
`>5-point change from baseline;17 EQ-5D >10-point change from baseline; FACIT-Fatigue score >4-point change from baseline).
`*Estimates from generalised linear mixed model. n/N represents the observed number of responders (n) for patients who completed through week 24 (N). Patients who discontinued from
`the study were not included in the denominator. Estimates from the generalised linear mixed model include all available data from completed and discontinued patients.
`†p<0.01 for combined 10 mg and 50 mg groups versus placebo (Fisher’s exact test).
`‡Patients with baseline anti-dsDNA above 31 IU/mL were included in the ≥10% reduction analysis (n=50); patients with baseline anti-dsDNA above 40 IU/mL were included in the ≥30%
`reduction analysis (n=48); patients with baseline anti-dsDNA above 54 IU/mL were included in the ≥50% reduction analysis (n=45).
`§Patients with complement data were included in the analyses of changes in C3 and C4 concentrations (placebo, n=41; 10 mg, n=39; 50 mg, n=38).
`¶Patients with a baseline corticosteroid dose >7.5 mg/day were included in the corticosteroid reduction analysis (n=62).
`BICLA, BILAG-based Composite Lupus Assessment; BILAG, British Isles Lupus Assessment Group; dsDNA, double-stranded DNA; EQ-5D, European Quality of Life 5 Dimensions; FACIT,
`Functional Assessment of Chronic Illness Therapy; LS, least squares; MCS, mental component summary; PCS, physical component summary; SF-36, 36-item Short Form Health Survey; SFI,
`modified Safety of Estrogens in Lupus Erythematosus: National Assessment (SELENA)-Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) Flare Index; SRI, Systemic Lupus
`Erythematosus Responder Index; VAS, visual analogue scale.
`
`similar to or worse than placebo for both SRI-4 (18.2% vs
`38.1% for placebo) and BICLA (27.3% vs 26.2% for placebo).
`Key efficacy outcomes
`are
`summarised in table 2.
`Treatment failure rates were significantly lower with 10 mg
`( p<0.01) and 50 mg ( p<0.05) versus placebo. No patients
`receiving 10 mg, and two receiving 50 mg experienced a
`severe SFI flare, compared with eight patients receiving
`placebo; severe SFI flare incidence was significantly lower for
`pooled 10 mg and 50 mg doses versus placebo ( p<0.01).
`Severe BILAG flare rates were also lower with PF-04236921
`vs placebo, although statistical significance was not achieved.
`Dose-dependent
`reductions
`in C3, C4 and CRP were
`observed.
`
`Across all groups, mean baseline SF-36 physical component
`summary (PCS) score (SD) was 34.3 (9.0) and mental compo-
`nent summary (MCS) score was 40.4 (10.9), which were
`approximately 1.5 SD and 1.0 SD <normative scores of 50,
`respectively.17 At week 24, trends towards improvements in
`SF-36 PCS scores, most SF-36 domain scores, FACIT-Fatigue
`and EQ-5D VAS scores were reported with 10 mg or 50 mg
`versus placebo. All HRQOL changes from baseline with 10 mg
`exceeded minimum clinically important differences (MCIDs).18
`
`Post hoc analysis of the enriched population
`Four univariate baseline parameters were associated with signifi-
`cant improvements in SRI-4 response rates for 10 mg versus
`
`538
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`Lassen - Exhibit 1029, p. 5
`
`

`

`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`Table 3 Summary of efficacy outcomes at week 24 in the enriched population
`
`Clinical and epidemiological research
`
`SRI response rate, n/N (%)*
`
`OR vs placebo (90% CI)
`
`p Value
`
`BICLA response rate, n/N (%)*
`
`OR vs placebo (90% CI)
`
`p Value
`
`Patients with disease flares, n (%)
`
`Placebo (n=33)
`
`10 mg (n=30)
`
`8/30 (27.7)
`
`15/21 (73.1)
`
`50 mg (n=38)
`
`12/28 (43.1)
`
`6/30 (19.7)
`
`7.09 (2.11 to 23.85)
`
`1.98 (0.67 to 5.86)
`
`0.004
`
`12/21 (55.7)
`
`0.151
`
`10/28 (34.7)
`
`5.11 (1.56 to 16.72)
`
`2.16 (0.71 to 6.59)
`
`0.012
`
`0.127
`
`BILAG flares (new BILAG A or two new BILAG B organ domain scores)
`
`Severe SFI flares
`
`5/33 (15.2)
`
`8/33 (24.2)
`
`0/30 (0.0)†
`
`0/28 (0.0)†
`
`Placebo (n=33)
`
`10 mg (n=28)
`
`LS mean change in SF-36 score from baseline (SE)
`
`PCS score
`
`MCS score
`
`Physical functioning
`
`Role physical
`
`Body pain
`
`General health
`
`Vitality
`
`Social functioning
`
`Role emotional
`
`Mental health
`
`LS mean change in EQ-5D VAS score from baseline (SE)
`
`LS mean change in FACIT-Fatigue score from baseline (SE)
`
`2.80 (1.4)
`
`2.04 (1.6)
`
`4.62 (4.0)
`
`8.44 (3.9)
`
`6.43 (3.9)
`
`6.80 (2.7)
`
`3.45 (3.5)
`
`5.80 (4.1)
`
`3.62 (3.7)
`
`4.20 (3.0)
`
`2.30 (3.2)
`
`1.16 (1.8)
`
`7.60 (1.5)‡
`
`2.69 (1.7)
`
`15.09 (4.4)
`
`14.63 (4.3)
`
`17.79 (4.2)
`
`14.13 (3.0)
`
`12.05 (3.8)
`
`10.02 (4.5)
`
`5.89 (4.1)
`
`6.78 (3.3)
`
`11.47 (3.5)
`
`5.28 (2.0)
`
`0/38 (0.0)†
`
`2/35 (5.7)†
`
`50 mg (n=37)
`
`5.07 (1.3)
`
`1.56 (1.5)
`
`12.44 (3.8)
`
`13.83 (3.7)
`
`11.56 (3.7)
`
`4.59 (2.6)
`
`4.19 (3.3)
`
`9.54 (3.9)
`
`8.98 (3.6)
`
`2.75 (2.9)
`
`6.10 (3.0)
`
`3.68 (1.8)
`
`Bold italic text denotes changes that were greater than the minimum clinically important difference (SF-36 PCS and MCS >2.5-point change from baseline;17 SF-36 domain scores
`>5-point change from baseline;17 EQ-5D >10-point change from baseline; FACIT-Fatigue score >4-point change from baseline).
`*Estimates from generalised linear mixed model. n/N represents the observed number of responders (n) for patients who completed through week 24 (N). Patients who discontinued from
`the study were not included in the denominator. Estimates from the generalised linear mixed model include all available data from completed and discontinued patients.
`†p<0.01 for combined 10 mg and 50 mg groups versus placebo (Fisher’s exact test).
`‡p<0.05 vs placebo.
`BICLA, BILAG-based Composite Lupus Assessment; BILAG, British Isles Lupus Assessment Group; EQ-5D, European Quality of Life 5 Dimensions; FACIT, Functional Assessment of Chronic
`Illness Therapy; LS, least squares; MCS, mental component summary; PCS, physical component summary; SF-36, 36-item Short Form Health Survey; SFI, modified Safety of Estrogens in
`Lupus Erythematosus: National Assessment (SELENA)-Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) Flare Index; SRI, Systemic Lupus Erythematosus Responder Index; VAS,
`visual analogue scale.
`
`placebo and defined an enriched population with greater disease
`activity at baseline: SLEDAI-2K score ≥10, corticosteroids
`≥7.5 mg/day, anti-dsDNA ≥28 IU/mL or hypocomplementaemia
`(C3 and C4). This included 101 patients ( placebo, n=33;
`10 mg, n=30; 50 mg, n=38), approximately 74% of the total
`population who had one or more of these characteristics.
`Efficacy outcomes in the enriched population are summarised
`in table 3. With 10 mg, SRI-4 and BICLA placebo-corrected
`response rates (GLMM) were greater in the enriched population
`than in the total population (45.4 vs 19.8 for SRI-4 and 36.0 vs
`24.6 for BICLA; figure 2) and significantly different
`than
`placebo for both SRI-4 ( p=0.004) and BICLA ( p=0.012).
`However, response rates in the 50 mg group were only margin-
`ally higher in the enriched population than in the total popula-
`tion. Notably, 10 mg was also associated with significant
`improvements in SF-36 PCS scores versus placebo ( p<0.05)
`and trends towards improvements in SF-36 MCS and domain
`scores. Changes from baseline in most SF-36 domain scores,
`EQ-5D VAS and FACIT-Fatigue scores exceeded MCID.
`
`Safety outcomes
`Rates of deaths, treatment-emergent AEs (TEAEs), serious AEs
`(SAEs) and AEs leading to treatment discontinuation during the
`treatment phase are summarised in table 4. The most frequent
`TEAEs (excluding infections and injection-site reactions) were
`headache, nausea and diarrhoea (nausea and diarrhoea were
`
`most commonly reported with placebo), and the most frequent
`infectious TEAEs were upper respiratory infection, cystitis and
`pharyngitis/laryngitis. More patients experienced non-infectious
`SAEs with placebo or 200 mg than with 10 mg or 50 mg. The
`higher SAE rate for placebo was largely due to a greater number
`of SLE flares. Serious infections occurred most frequently with
`200 mg. There were no cases of herpes zoster or malignancies.
`Four deaths occurred during the study. A 32-year-old woman
`died after receiving a single 10 mg dose due to a suspected pul-
`monary embolism (PE). A 54-year-old woman experienced
`severe shortness of breath and died on the way to the hospital
`after receiving a single 200 mg dose. Two additional patients
`(a 61-year-old woman and a 24-year-old woman) died after
`receiving two doses of 200 mg due to infectious causes
`combined with PEs (sepsis with PE and disseminated tubercu-
`losis with PE). A causal relationship with study medication
`could not be excluded for any of the events; therefore, the data
`monitoring committee recommended stopping further dosing of
`the 200 mg group. Additional details on the deaths are included
`in the online supplementary materials. In addition to the three
`deaths due to PEs listed above, there was one additional SAE
`that was due to a PE in a patient who received placebo.
`
`DISCUSSION
`While none of the treatment arms were significantly different
`than placebo for the primary end point, results of this trial
`
`Wallace DJ, et al. Ann Rheum Dis 2017;76:534–542. doi:10.1136/annrheumdis-2016-209668
`
`539
`
`Lassen - Exhibit 1029, p. 6
`
`

`

`Clinical and epidemiological research
`
`Table 4 TEAEs during the 24-week treatment phase
`
`Deaths, n (%)
`
`SAEs (excluding infections),
`n (%)*
`
`Serious infections, n (%)
`
`Sepsis
`
`Bronchitis
`
`Tuberculosis
`
`Bronchopneumonia
`
`Cellulitis
`
`Clostridium difficile colitis
`
`Placebo
`(n=45)
`
`0 (0.0)
`
`5 (11.1)
`
`2 (4.4)
`
`1 (2.2)
`
`1 (2.2)
`
`0 (0.0)
`
`0 (0.0)
`
`0 (0.0)
`
`1 (2.2)
`
`10 mg
`(n=45)
`
`1 (2.2)
`
`2 (4.4)
`
`1 (2.2)
`
`0 (0.0)
`
`0 (0.0)
`
`0 (0.0)
`
`0 (0.0)
`
`1 (2.2)
`
`0 (0.0)
`
`50 mg
`(n=47)
`
`0 (0.0)
`
`1 (2.1)
`
`2 (4.3)
`
`1 (2.1)
`
`0 (0.0)
`
`0 (0.0)
`
`0 (0.0)
`
`1 (2.1)
`
`0 (0.0)
`
`200 mg
`(n=46)
`
`3 (6.5)
`
`5 (10.9)
`
`4 (8.7)
`
`1 (2.2)
`
`0 (0.0)
`
`1 (2.2)
`
`1 (2.2)
`
`1 (2.2)
`
`0 (0.0)
`
`Sinusitis
`
`1 (2.2)
`
`0 (0.0)
`
`0 (0.0)
`
`0 (0.0)
`
`Any AEs (excluding
`infections and ISRs), n (%)
`
`34 (75.6)
`
`34 (75.6)
`
`32 (68.1)
`
`31 (67.4)
`
`Common AEs (≥5% in any treatment group, excluding infections and ISR), n (%)
`
`Headache
`
`Nausea
`
`Diarrhoea
`
`SLE
`
`Arthralgia
`
`Dizziness
`
`Cough
`
`2 (4.4)
`
`5 (11.1)
`
`5 (11.1)
`
`3 (6.7)
`
`3 (6.7)
`
`2 (4.4)
`
`2 (4.4)
`
`1 (2.2)
`
`4 (8.9)
`
`2 (4.4)
`
`2 (4.4)
`
`3 (6.7)
`
`1 (2.2)
`
`1 (2.2)
`
`4 (8.9)
`
`1 (2.2)
`
`5 (10.6)
`
`3 (6.4)
`
`2 (4.3)
`
`2 (4.3)
`
`2 (4.3)
`
`3 (6.4)
`
`0 (0.0)
`
`4 (8.5)
`
`5 (10.9)
`
`5 (10.9)
`
`3 (6.5)
`
`1 (2.2)
`
`2 (4.3)
`
`2 (4.3)
`
`1 (2.2)
`
`1 (2.2)
`
`Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-209668 on 26 September 2016. Downloaded from
`
`http://ard.bmj.com/
`
` on 19 July 2019 by guest. Protected by copyright.
`
`hospitalisation and are associated with significant morbidity and
`mortality.19 Trends towards improved HRQOL were reported
`with 10 mg by SF-36 PCS, FACIT-Fatigue and EQ-5D.
`Although a greater percentage of patients receiving 10 mg or
`50 mg achieved ≥10% decreases in anti-dsDNA antibodies from
`baseline versus placebo, applying a higher level of response
`(≥30% or ≥50%) did not reveal a clear effect. Dose-dependent
`decreases in complement were noted, consistent with results
`with another IL-6 inhibitor.15
`To determine if there was a subgroup of patients with higher
`response rates, a post hoc analysis of patients with high disease
`activity at baseline was performed. Efficacy of 10 mg appeared
`more pronounced in this enriched population based upon sig-
`nificantly greater SRI-4 and BICLA response rates versus
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket