throbber
O R I G I N A L P A P E R
`One year of liraglutide treatment offers sustained and
`more effective glycaemic control and weight reduction
`compared with sitagliptin, both in combination with
`metformin, in patients with type 2 diabetes: a
`randomised, parallel-group, open-label trial
`
`R. Pratley,1 M. Nauck,2 T. Bailey,3 E. Montanya,4 R. Cuddihy,5 S. Filetti,6 A. Garber,7 A. B. Thomsen,8
`H. Hartvig,9 M. Davies,10 for the 1860-LIRA-DPP-4 Study Group
`
`S U M M A R Y
`
`Aim: The aim of this study was to compare the efficacy and safety of once-daily
`human glucagon-like peptide-1 analogue liraglutide with dipeptidyl peptidase-4
`inhibitor sitagliptin, each added to metformin, over 52 weeks in individuals with
`type 2 diabetes. Methods: In an open-label, parallel-group trial, metformin-trea-
`ted participants were randomised to liraglutide 1.2 mg ⁄ day (n = 225), liraglutide
`1.8 mg ⁄ day (n = 221) or sitagliptin 100 mg ⁄ day (n = 219) for 26 weeks (main
`phase). Participants continued the same treatment
`in a 26-week extension.
`Results: Liraglutide (1.2 or 1.8 mg) was superior to sitagliptin for reducing HbA1c
`from baseline (8.4–8.5%) to 52 weeks: )1.29% and )1.51% vs. )0.88% respec-
`tively. Estimated mean treatment differences between liraglutide and sitagliptin
`were as follows: )0.40% (95% confidence interval )0.59 to )0.22) for 1.2 mg
`and )0.63% ()0.81 to )0.44) for 1.8 mg (both p < 0.0001). Weight loss was
`greater with liraglutide 1.2 mg ()2.78 kg) and 1.8 mg ()3.68 kg) than sitagliptin
`()1.16 kg)
`(both p < 0.0001). Diabetes Treatment Satisfaction Questionnaire
`scores increased significantly more with liraglutide 1.8 mg than with sitagliptin
`(p = 0.03). Proportions of participants reporting adverse events were generally
`comparable; minor hypoglycaemia was 8.1%, 8.3% and 6.4% for
`liraglutide
`1.2 mg, 1.8 mg and sitagliptin respectively. Gastrointestinal side effects, mainly
`nausea, initially occurred more frequently with liraglutide, but declined after several
`weeks. Conclusion: Liraglutide provides greater sustained glycaemic control and
`body weight
`reduction over 52 weeks. Treatment satisfaction was significantly
`greater with 1.8 mg liraglutide, similar to 26-week results. The safety profiles of
`liraglutide and sitagliptin are consistent with previous reports.
`
`What’s known
`Results of independent trials and several 26-week
`head-to-head trials suggest that GLP-1 receptor
`agonists produce greater glycaemic and weight
`reductions compared with DPP-4 inhibitors. Our
`26-week trial showed that the human once-daily
`GLP-1 analogue liraglutide effected greater
`glycaemic control and weight loss than the DPP-4
`inhibitor sitagliptin.
`
`What’s new
`Longer-term sustainability of the 26-week efficacy
`and safety results with liraglutide and sitagliptin, as
`well as the maintenance of the greater comparative
`efficacy with liraglutide, was not known. This report
`shows that 26-week improvements were sustained
`after 52 weeks of treatment, with liraglutide
`producing significantly greater glycaemic and
`weight reductions than sitagliptin.
`
`Introduction
`
`Metformin is now standard first-line treatment (in
`addition to lifestyle modifications) for type 2 diabe-
`tes
`(T2D)
`(1). The progressive nature of T2D,
`including declining beta-cell function, usually neces-
`sitates addition of other antihyperglycaemic agents to
`metformin, as blood glucose levels rise. However,
`current guidelines vary with respect to second-line
`therapy (1,2). A meta-analysis of currently available
`non-insulin antihyperglycaemic agents added to met-
`formin revealed that, while reductions in glycosylated
`
`haemoglobin (HbA1c) were similar across
`several
`drug classes (including sulphonylureas, thiazolinedi-
`ones
`and alpha-glucosidase
`inhibitors;
`reduction
`range: 0.64–0.97%), treatment side effects (such as
`weight gain and ⁄ or hypoglycaemia) varied consider-
`ably (3). Therefore, head-to-head studies of glucose-
`lowering agents are needed to compare overall
`clinical efficacy and safety when added to metformin.
`Treatment intensification with incretin-based ther-
`apies is appealing given that they provide good gly-
`caemic control with a low risk of hypoglycaemia,
`because of
`the glucose-dependent
`stimulation of
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`doi: 10.1111/j.1742-1241.2011.02656.x
`
`1Diabetes and Metabolism
`Translational Medicine Unit,
`University of Vermont College
`of Medicine, Burlington, VT,
`USA
`2Diabetes Center,
`Diabeteszentrum Bad
`Lauterberg, Bad Lauterberg im
`Harz, Germany
`3Director, AMCR Institute,
`Escondido, CA, USA
`4Endocrine Unit, IDIBELL-
`Hospital Universitari Bellvitge,
`Barcelona, Spain
`5Medical Director, International
`Diabetes Center, Minneapolis,
`MN, USA
`6Department of Clinical
`Sciences, Sapienza University of
`Rome, Rome, Italy
`7Departments of Medicine,
`Biochemistry, and Molecular and
`Cellular Biology, Baylor College
`of Medicine, Houston, TX, USA
`8Medical & Science Department,
`Novo Nordisk A ⁄ S, Soeborg,
`Denmark
`9Biostatistical Department, Novo
`Nordisk A ⁄ S, Soeborg, Denmark
`10Department of Cardiovascular
`Sciences, University of Leicester,
`Leicester, UK
`
`Correspondence to:
`Dr Richard Pratley, Diabetes and
`Metabolism Translational
`Medicine Unit, Colchester
`Research Facility 110C,
`University of Vermont College of
`Medicine, Burlington, VT 04556,
`USA
`Tel.: + 1 802 656 5811
`Fax: + 1 802 656 8031
`Email: richard.pratley@uvm.edu
`
`Disclosures
`RP has attended advisory panels
`for GlaxoSmithKline; has
`received research
`
`397
`
`MPI EXHIBIT 1126 PAGE 1
`
`

`

`398
`
`Liraglutide vs. sitagliptin 1-year results
`
`grants ⁄ participated in clinical
`trials for Merck & Co., Novartis
`Pharmaceuticals, Novo Nordisk
`Inc., Roche, Takeda Global
`Research Development Center
`Inc., Eli Lilly & Co., Merck & Co.,
`Novartis Pharmaceuticals
`Corporation, Novo Nordisk A ⁄ S,
`Pfizer Inc., sanofi-aventis; has
`acted as consultant for
`GlaxoSmithKline; has received
`honoraria from Novartis
`Pharmaceuticals Corp, Novo
`Nordisk Inc., Roche and Takeda
`Global Research Development
`Center and has attended
`speakers’ bureaux for Merck &
`Co. MN has received research
`grants from Bayer Vital Pharma,
`Eli Lilly & Co., Menarini ⁄ Berlin-
`Chemie, Merck Sharp & Dohme,
`Novartis Pharma and Novo
`Nordisk A ⁄ S; has accepted
`honoraria for membership in
`advisory boards and consulting,
`and has received honoraria for
`speaking on incretin-based
`antidiabetic medications from
`Amylin Pharmaceuticals,
`AstraZeneca, Bayer Vital
`Pharma, Berlin-
`Chemie ⁄ Menarini, Biovitrum,
`Boehringer Ingelheim, Eli Lilly &
`Co., GlaxoSmithKline, Hoffman
`La Roche, Novartis Pharma,
`Novo Nordisk A ⁄ S, sanofi-
`aventis Pharma and Takeda. TB
`has attended advisory panels for
`Amylin Pharmaceuticals; has
`received research support from
`Animas Corporation, Becton
`Dickinson, CPEX
`Pharmaceuticals, Dexcom, Eli
`Lilly & Co., GlaxoSmithKline,
`Medtronic MiniMed, Merck,
`Novo Nordisk Inc., Resmed and
`sanofi-aventis; and has attended
`speakers’ bureaux for Amylin
`Pharmaceuticals Inc., Dexcom,
`Eli Lilly & Co., Medtronic
`MiniMed, Novo Nordisk Inc.,
`Roche Diagnostics and sanofi-
`aventis. EM has attended
`advisory panels for Merck Sharp
`& Dohme, Novartis, Novo
`Nordisk and sanofi-aventis.
`RC has attended advisory boards
`for Novo Nordisk, Bayer, Roche,
`CeQur, Eli Lilly and Abbott; has
`received support for educational
`activities from Lifescan, Eli Lilly,
`Merck, Novartis and sanofi-
`aventis; has or is principal or co-
`investigator for sponsored
`clinical trials research for
`Amylin, Abbott, Bayer, Daiichi
`Sankyo, Dexcom, Edwards
`Lifesciences, Eli Lilly, Hygeia,
`Intarcia, Johnson &
`Johnson ⁄ Lifescan, Mannkind,
`Medtronic, Merck, Novo
`Nordisk, Quotient Diagnostics,
`ResMed, Roche, sanofi-aventis,
`
`insulin secretion and inhibition of glucagon release,
`and do not produce weight gain (3–6). Glucagon-like
`peptide-1 (GLP-1) receptor agonists and dipeptidyl
`peptidase-4 (DPP-4) inhibitors are two distinct clas-
`ses of incretin-based therapies. While 26-week, head-
`to-head studies suggest that GLP-1 receptor agonists
`have greater glycaemic and weight reduction efficacy
`than DPP-4 inhibitors (7–9),
`longer-term results
`have not been reported.
`In a 26-week, head-to-head trial of the once-daily
`human GLP-1 analogue liraglutide and the DPP-4
`inhibitor sitagliptin, both in combination with met-
`liraglutide (1.2 or 1.8 mg ⁄ day) was signifi-
`formin,
`cantly more effective than sitagliptin (100 mg ⁄ day)
`for reducing HbA1c ()1.24% and )1.50% vs. )0.90%
`respectively),
`fasting
`plasma
`glucose
`(FPG)
`()1.87 mmol ⁄ l [)33.66 mg ⁄ dl] and )2.14 mmol ⁄ l
`[)38.52 mg ⁄ dl] vs. )0.83 mmol ⁄ l [)14.94 mg ⁄ dl],
`respectively) and body weight ()2.86 and )3.38 kg
`vs. )0.96 kg respectively) (8). Incidence of minor
`hypoglycaemia was low (around 5%) and compara-
`ble across treatment groups. Nausea incidence was
`greater with liraglutide than with sitagliptin during
`therapy initiation, but generally declined after several
`weeks of treatment.
`Trial participants could continue treatment in a
`26-week extension phase designed to evaluate the
`sustainability of efficacy and safety effects of liraglu-
`tide and sitagliptin. This report shows that 26-week
`improvements were sustained after 52 weeks of treat-
`ment, with liraglutide producing greater glycaemic
`and weight reductions than sitagliptin.
`
`Methods
`
`Study design
`inclu-
`Details of
`study design and participant
`sion ⁄ exclusion criteria have been reported previously
`(8). Briefly, in a multinational, randomised, parallel-
`group, open-label, active-comparator trial, participants
`with T2D previously treated with metformin mono-
`therapy (‡ 1500 mg ⁄ day) for a minimum of 3 months,
`but with suboptimal glycaemic control (HbA1c 7.5–
`10%), were randomised (1 : 1 : 1) to treatment with
`either liraglutide 1.2 or 1.8 mg ⁄ day (subcutaneous
`injection) or sitagliptin 100 mg ⁄ day (orally) while
`continuing on existing metformin therapy.
`After completing the 26-week main phase, partici-
`pants choosing to enrol
`in the extension provided
`written informed consent and continued for another
`26 weeks
`in their originally
`assigned treatment
`groups. The protocol, including the extension, was
`institutional review board-approved, followed Good
`Clinical Practice guidelines and conformed to the
`Declaration of Helsinki. The 52-week trial was
`
`initiated on 16 June 2008 and completed on 10
`December 2009.
`Additional withdrawal criteria during the exten-
`sion were: elevated FPG > 11.1 mmol ⁄ l (200 mg ⁄ dl)
`with no treatable intercurrent cause or acute pancre-
`atitis (defined as a minimum two out of three of the
`following: characteristic abdominal pain, amylase
`and ⁄ or lipase > 3 · upper normal range or charac-
`teristic findings on computed tomography ⁄ magnetic
`resonance imaging).
`
`Outcomes
`included
`Efficacy outcomes assessed at 52 weeks
`change in HbA1c, FPG, body weight, proportion of
`participants achieving HbA1c < 7% or £ 6.5%, pro-
`portion of participants reaching the composite end-
`point of HbA1c < 7.0% with no weight gain and no
`confirmed major
`(participant unable
`to
`treat
`him ⁄ herself) or minor (plasma glucose < 3.1 mmol ⁄ l
`[56 mg ⁄ dl]) hypoglycaemia. Other measures included
`fasting C-peptide, fasting pro-insulin : insulin ratio,
`and homeostasis model assessment analyses of beta-
`cell
`function (HOMA-B) and insulin resistance
`(HOMA-IR). Change in Diabetes Treatment Satisfac-
`tion Questionnaire (DTSQ) scores from baseline was
`not assessed in participants from Slovakia, Serbia or
`Slovenia (118 ⁄ 665 [17.7%]) because of the lack of
`validated questionnaires in their native languages.
`Safety and tolerability assessments at 52 weeks
`included incidence of adverse events (AEs) and hyp-
`oglycaemia, as well as various clinical and laboratory
`variables. AEs of special
`interest
`included nausea,
`thyroid AEs and pancreatitis.
`
`Statistical analysis
`Methods for statistical analyses were similar to those
`reported for the first 26 weeks (8). Glycaemic effi-
`cacy, as measured by change in HbA1c from baseline
`to week 52 of
`liraglutide vs. sitagliptin treatment,
`was assessed by a non-inferiority comparison with a
`margin of 0.4%, followed by a superiority compari-
`son. Both tests used two-sided hypotheses, with a
`p-value of < 0.05 considered significant. Analysis of
`covariance, with treatment and country as fixed
`effects and baseline measure as a covariate, was used
`for continuous efficacy end-points. Logistic regres-
`sion was used to analyse categorical variables, includ-
`ing the participant proportions achieving HbA1c
`targets and composite end-point (HbA1c < 7.0% with
`no weight gain and no confirmed major or minor
`hypoglycaemia), with treatment and country as fixed
`effects, and baseline HbA1c (and body weight for
`composite) as covariates. Efficacy assessments were
`performed on the full analysis set: all randomised
`participants exposed to at least one dose of the drug.
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 2
`
`

`

`Liraglutide vs. sitagliptin 1-year results
`
`399
`
`Takeda and Valeritas; and is an
`employee of the International
`Diabetes Center at Park Nicollet.
`All honoraria, speaking fees,
`consulting fees and research and
`educational support are paid
`directly to the non-profit
`International Diabetes Center of
`which RP is a salaried employee;
`he receives no personal
`payments for any of these
`activities. SF has acted as
`consultant and attended
`speakers’ bureaux for Novo
`Nordisk A ⁄ S. AG is an advisor
`and speaker for Novo Nordisk,
`Merck, GlaxoSmithKline and
`Sankyo. ABT is an employee of
`Novo Nordisk and was directly
`involved in study conduct. HH is
`an employee of Novo Nordisk
`and was directly involved in
`study conduct. MD has attended
`advisory panels for Eli Lilly &
`Co., Merck Sharp & Dohme Ltd.,
`Novartis Pharmaceuticals, Novo
`Nordisk Pharma Ltd., Schering-
`Plough, Roche and Bristol-Myers
`Squibb; has received research
`support from Eli Lilly & Co.,
`Merck Sharp & Dohme Ltd.,
`Novartis Pharmaceuticals, Novo
`Nordisk Pharma UK and
`GlaxoSmithKline; and has
`received honoraria for lectures
`from Eli Lilly & Co., Merck Sharp
`& Dohme Ltd., Novartis
`Pharmaceuticals and Novo
`Nordisk Pharma Ltd.
`
`Clinical trial registration
`number: clinicaltrials.gov,
`NCT00700817
`
`Re-use of this article is
`permitted in accordance with
`the Terms and Conditions set
`out at http://wileyonlinelibrary.
`com/onlineopen#OnlineOpen_
`Terms
`
`Missing data were imputed using the last observation
`carried forward (LOCF) method.
`The safety analysis set
`included all participants
`exposed to at least one dose of the drug they were
`randomised to. Serum calcitonin values were analy-
`sed using a repeated measures model, with time,
`gender, treatment and treatment-by-time interaction
`as fixed effects and participant as a random effect.
`Hypoglycaemia was analysed using a general
`linear
`model with treatment as a fixed effect. For each
`week of the extension, the proportions of partici-
`pants experiencing nausea were analysed using Fish-
`er’s exact test. Only summary statistics are reported
`for other safety parameters. Data are reported as
`least square means with 95% confidence interval
`(CI), unless otherwise noted. The significance level
`is p < 0.05.
`
`Results
`
`Participant disposition and baseline
`characteristics
`After screening, 665 participants were randomised
`into three treatment arms: liraglutide 1.2 mg, 1.8 mg
`and sitagliptin (Figure 1). As previously reported, the
`groups were well matched for baseline characteristics
`(8). Of participants completing 26 weeks, 497 ⁄ 554
`(90%) entered into the extension, with 436 ⁄ 497
`(88%) completing 52 weeks. A lower proportion of
`randomised participants withdrew from the exten-
`sion compared with the main phase, and withdrawal
`because of AEs was also lower in the extension.
`Patient withdrawal because of AEs in the main phase
`was higher for both liraglutide groups than for sitag-
`liptin, whereas only the liraglutide 1.8 mg group had
`a slightly higher AE withdrawal rate in the extension.
`
`Efficacy outcomes
`Mean HbA1c decreased more substantially with either
`dose of liraglutide compared with sitagliptin during
`the first 12 weeks, and these reductions were gener-
`ally maintained up to week 52 (Figure 2A). Mean
`reductions in HbA1c from baseline to week 52 with
`liraglutide 1.2 mg ()1.29% [95% CI: )1.43 to
`)1.15]) and 1.8 mg ()1.51% [)1.65 to )1.37]) were
`significantly
`greater
`compared with
`sitagliptin
`()0.88% [)1.02 to )0.74]). Estimated mean treat-
`ment differences were )0.40% (95% CI )0.59 to
`)0.22)
`for
`liraglutide 1.2 mg vs.
`sitagliptin and
`)0.63% ()0.81 to )0.44) for liraglutide 1.8 mg vs.
`sitagliptin (p < 0.0001 for both doses.)
`As with HbA1c, liraglutide was more effective for
`reducing FPG compared with sitagliptin (Figure 2B).
`FPG declined rapidly from baseline during weeks 0–4
`in all treatment groups and the reductions were gen-
`
`erally sustained up to 52 weeks. FPG reductions from
`baseline at week 52 were )1.71 mmol ⁄ l (95% CI
`)2.04 to )1.38) ()30.78 mg ⁄ dl [)36.78 to )24.78])
`for 1.2 mg liraglutide and )2.04 mmol ⁄ l ()2.37 to
`)1.71)
`()36.72 mg ⁄ dl
`[)42.72 to )30.72])
`for
`1.8 mg liraglutide vs. )0.59 mmol ⁄ l ()0.92 to )0.26)
`()10.62 mg ⁄ dl [)16.62 to )4.62]) for sitagliptin.
`Estimated mean treatment differences between lira-
`glutide and sitagliptin were )1.13 mmol ⁄ l (95% CI
`)1.57 to )0.68) ()20.34 mg ⁄ dl [)28.26 to )12.24])
`for 1.2 mg and )1.45 mmol ⁄ l
`()1.89 to )1.01)
`()26.1 mg ⁄ dl
`[)34.02
`to )18.18])
`for
`1.8 mg
`(p < 0.0001 vs. sitagliptin for both doses).
`Weight loss was considerably greater with liraglu-
`tide compared with sitagliptin (Figure 2C). Most
`weight loss occurred during the first 26 weeks and
`was
`sustained in the extension in all
`treatment
`groups (Figure 2C). At week 52, weight loss with
`liraglutide 1.2 mg was )2.78 kg (95% CI )3.39 to
`)2.17) compared with )3.68 kg ()4.29 to )3.07) for
`1.8 mg and )1.16 kg ()1.77 to )0.55) for sitagliptin.
`Estimated mean treatment differences were )1.62 kg
`(95% CI )2.43 to )0.82) for liraglutide 1.2 mg and
`)2.53 kg ()3.33 to )1.72) for liraglutide 1.8 mg vs.
`sitagliptin (p < 0.0001 for both doses). Weight loss
`with liraglutide 1.8 mg was significantly greater than
`that with liraglutide 1.2 mg (p = 0.03). The 26-week-
`reductions
`in waist circumference were generally
`maintained at week 52 in all groups and were signifi-
`cantly larger with liraglutide (both doses) than sitag-
`liptin (Table 1).
`As with the main study results (8), postprandial
`plasma glucose data were highly variable and difficult
`to interpret, and are excluded from this report. As
`this was a multinational study, data variability may
`have resulted from the varying meal content, time of
`meals and timing of postprandial glucose measure-
`ments across different countries.
`Overall, the magnitude of HbA1c reduction from
`baseline increased with the higher baseline HbA1c
`categories in all groups (Figure 3A). After 52 weeks,
`mean reductions in HbA1c were significantly greater
`with liraglutide 1.8 mg than with sitagliptin across all
`baseline HbA1c categories. The reductions were sig-
`nificantly larger with liraglutide 1.2 mg than with
`sitagliptin for two baseline HbA1c categories: > 8%
`to £ 8.5% and > 9%.
`Proportions of participants achieving target HbA1c
`< 7% (American Diabetes Association [ADA] target)
`or £ 6.5% (American Association of Clinical Endo-
`crinologists
`[AACE]
`target)
`increased during the
`extension in all treatment groups (Figure 3B). Over-
`all,
`liraglutide (both doses) was significantly more
`effective than sitagliptin in allowing patients to reach
`target HbA1c after 52 weeks.
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 3
`
`

`

`400
`
`Liraglutide vs. sitagliptin 1-year results
`
`Figure 1 Trial flow chart with participant demographics at baseline. Demographic data are mean ± SD, unless otherwise
`noted. *Participants were withdrawn if they fulfilled withdrawal criteria, decided that they no longer wanted to participate,
`or did not attend any visit after randomisation. BMI, body mass index; FAS, full analysis set; FPG, fasting plasma glucose
`
`The estimated proportion of participants reaching
`the composite end-point of HbA1c < 7.0%, with
`no weight gain and no confirmed major or minor
`hypoglycaemia, increased during the extension in all
`treatment groups (Figure 3C). After 52 weeks, a sig-
`nificantly greater percentage of participants achieved
`the
`composite
`end-point with liraglutide
`(both
`groups) than with sitagliptin, with an odds ratio
`(OR) vs. sitagliptin of 2.80 (95% CI 1.74 to 4.48)
`and 4.37 (2.74 to 6.98) for 1.2 and 1.8 mg liraglutide
`respectively (both doses p < 0.0001). Liraglutide
`
`1.8 mg was more effective than liraglutide 1.2 mg
`(OR: 1.56 [1.04 to 2.35], p = 0.03).
`Overall, the improved status of several indicators
`of beta-cell function (fasting C-peptide, fasting pro-
`insulin:insulin ratio and HOMA-B) at week 26 was
`maintained at week 52, with liraglutide effecting
`significantly greater improvements than sitagliptin
`(Table 1). The reduction in HOMA-IR became sig-
`nificantly greater with liraglutide 1.8 mg than sitag-
`liptin during the extension. As observed at week 26,
`mean heart rate continued to be slightly but signifi-
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 4
`
`

`

`Liraglutide vs. sitagliptin 1-year results
`
`401
`
`and sitagliptin respectively) with no consistent
`pattern with respect to system organ class (Table 2).
`Three deaths occurred during the 52-week period.
`Two deaths during the first 26 weeks, one in a
`participant with pancreatic carcinoma (liraglutide
`1.8 mg) and one because of cardiac arrest (sitaglip-
`tin), were reported previously and considered unli-
`kely to be related to the trial drugs (8). One sudden
`cardiac death during the extension occurred in a 66-
`year-old man randomised to sitagliptin and was
`judged as unlikely to be related to the trial drug by
`the investigator.
`Gastrointestinal disorders, as well as infections and
`infestations, were the most commonly reported mild-
`to-moderate AEs with liraglutide. The incidence
`of nausea,
`the most prevalent gastrointestinal AE
`with liraglutide, declined after the first 3 weeks of
`treatment and remained low during the extension
`(Figure 4). For each week of the extension, the pro-
`portions of participants experiencing nausea did not
`differ significantly between liraglutide (1.2 or 1.8 mg)
`and sitagliptin treatment groups.
`One episode of major hypoglycaemia (blood glu-
`cose 3.6 mmol ⁄ l [64.8 mg ⁄ dl]) occurred during the
`first 26 weeks in a participant on liraglutide 1.2 mg
`(8). Third-party assistance was required, but no sei-
`zures or coma occurred. The participant recovered
`and the episode was categorised as possibly related to
`the trial product by the investigator. No major hypo-
`glycaemic episodes occurred during the extension.
`Minor hypoglycaemia rates were low and comparable
`between treatment
`groups over 52 weeks,
`after
`excluding an outlier in the 1.8 mg liraglutide group
`with 21 minor events during the first 26 weeks and
`two events in the extension (leading to participant
`withdrawal
`from the trial). Adjusted minor hypo-
`glycaemia rates were 0.143, 0.154 and 0.137 hypogly-
`caemic episodes per patient per year for liraglutide
`1.2 mg, 1.8 mg and sitagliptin respectively.
`One case of ‘non-acute’ pancreatitis was reported
`during the extension in a 54-year-old man, with a
`medical history of hepatitis and hyperlipidaemia,
`treated with liraglutide 1.8 mg for 227 days. Initially,
`the participant experienced abdominal pain, nausea,
`vomiting for 1 day and black stools for 3 days. The
`participant was instructed to stop aspirin and initiate
`omeprazole treatment. Upon later hospitalisation for
`a different condition, laboratory tests showed slightly
`increased levels of amylase (2.6 lkat ⁄ l, normal range:
`0–1.67 lkat ⁄ l) and lipase (1.44 lkat ⁄ l, normal range:
`0–1 lkat ⁄ l). The investigator decided to withdraw
`the participant, although the specific withdrawal cri-
`teria for acute pancreatitis were not met. The event
`was rated as mild and possibly related to the trial
`drug by the investigator.
`
`Figure 2 Effect of 1.2 mg liraglutide, 1.8 mg liraglutide or
`100 mg sitagliptin on glycaemic control and body weight
`from baseline to 52 weeks. (A) Mean HbA1c values. (B)
`Mean fasting plasma glucose (FPG) values. (C) Mean
`change in body weight. Error bars are 1.96 · SE,
`corresponding to the 95% CI
`
`cantly elevated with liraglutide compared with sitag-
`liptin at week 52 (Table 1).
`The increase in DTSQ scores at week 26 was gen-
`erally sustained at week 52 in all treatment groups.
`The improvement in overall treatment satisfaction,
`measured as the increase in DTSQ scores between
`weeks 0 and 52, was significantly higher with liraglu-
`tide 1.8 mg (baseline: 28.0) than with sitagliptin
`(baseline: 27.1): 4.3 (95% CI 3.3 to 5.3) vs. 3.0 (2.0
`to 4.0) (p = 0.03). By contrast, the increase from
`baseline (27.8) with liraglutide 1.2 mg (3.3 [2.3 to
`4.3]) was not statistically different from sitagliptin.
`
`Safety outcomes
`The majority (‡ 97%) of treatment-emergent AEs in
`all groups over 52 weeks were mild or moderate. The
`proportion of participants reporting serious AEs was
`low and comparable between treatment groups
`(4.5%, 6.0% and 5.5% for liraglutide 1.2 mg, 1.8 mg
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 5
`
`

`

`402
`
`Liraglutide vs. sitagliptin 1-year results
`
`1.63 ()7.74 to 11.00)
`0.09 (0.03 to 0.15)
`)0.09 ()0.13 to )0.05)
`25.76 (19.39 to 32.13)
`)1.36 ()2.05 to )0.67)
`
`)2.27 ()11.76 to 7.22)
`0.01 ()0.05 to 0.07)
`)0.01 ()0.05 to 0.03)
`3.98 ()2.45 to 10.45)
`)0.41 ()1.1 to 0.28)
`
`1.67 ()11.2 to 14.51); 0.80
`0.04 ()0.05 to 0.12); 0.38
`)0.06 ()0.11 to )0.02); 0.005
`18.60 (9.82 to 27.38); < 0.001
`)0.85 ()1.79 to 0.08); 0.07
`
`3.90 ()8.76 to 16.57); 0.55
`0.08 (0.00 to 0.16); 0Æ06
`)0.08 ()0.13 to )0.04); < 0.001
`21.77 (13.12 to 30.42); < 0.001
`)0.94 ()1.86 to )0.02); 0.04
`
`)2.55 ()4.37 to )0.72)
`)0.87 ()1.99 to 0.25)
`3.09 (1.91 to 4.27)
`
`)1.03 ()2.85 to 0.79)
`)1.47 ()2.59 to )0.35)
`0.09 ()1.09 to 1.27)
`
`0.66 ()1.79 to 3.10); 0.60
`0.94 ()0.57 to 2.45); 0.22
`1.63 (0.03 to 3.22); 0.05
`
`)1.53 ()3.97 to 0.92); 0.22
`0.60 ()0.90 to 2.11); 0.43
`3.00 (1.40 to 4.59); < 0.001
`
`)0.09 ()0.21 to 0.03)
`0.09 ()0.01 to 0.19)
`)0.19 ()0.27 to )0.11)
`0.02 (0.00 to 0.04)
`)0.32 ()0.52 to )0.12)
`)0.10 ()0.14 to )0.06)
`–0.03 ()0.05 to )0.01)
`
`0.03 ()0.15 to 0.09)
`0.17 (0.07 to 0.27)
`)0.15 ()0.23 to )0.07)
`0.01 ()0.01 to 0.03)
`)0.23 ()0.43 to )0.03)
`)0.06 ()0.10 to )0.02)
`–0.03 ()0.05 to )0.01)
`
`)0.04 ()0.20 to 0.12); 0.61
`)0.08 ()0.20 to 0.05); 0.25
`0.04 ()0.06 to 0.13); 0.47
`0.00 ()0.03 to 0.04); 0.92
`0.12 ()0.13 to 0.38); 0.34
`)0.01 ()0.06 to 0.04); 0.67
`0.00 ()0.03 to 0.04); 0.83
`
`)0.12 ()0.28 to 0.03); 0.12
`)0.08 ()0.21 to 0.05); 0.23
`)0.04 ()0.14 to 0.06); 0.43
`0.01 ()0.02 to 0.05); 0.53
`)0.09 ()0.34 to 0.16); 0.49
`)0.05 ()0.09 to 0.00); 0.05
`)0.00 ()0.04 to 0.03); 0.93
`
`)3.02 ()3.76 to )2.28)
`
`)1.23 ()1.97 to )0.49)
`
`)1.13 ()2.12 to )0.13); 0Æ03
`
`)1.79 ()2.78 to )0.79); < 0.001
`
`Table 1 Changes in secondary end-points from baseline to week 52
`
`Mean change from baseline [95% CI]
`
`Estimated treatment difference; p-value
`
`Liraglutide 1.2 mg
`
`Liraglutide 1.8 mg
`
`Sitagliptin 100 mg
`
`Liraglutide 1.2 mg–sitagliptin
`
`Liraglutide 1.8 mg–sitagliptin
`
`Beta-cell function and insulin resistance
`)0.60 ()10.11 to 8.91)
`Fasting insulin (pmol ⁄ l)
`0.05 ()0.01 to 0.11)
`Fasting C-peptide (nmol ⁄ l)
`)0.07 ()0.11 to )0.03)
`Fasting pro-insulin:insulin ratio
`HOMA-B (%)
`22.58 (16.09 to 29.07)
`)1.27 ()1.96 to )0.58)
`HOMA-IR (%)
`Blood pressure
`Systolic blood pressure (mmHg)
`Diastolic blood pressure (mmHg)
`Heart rate (beats ⁄ min)
`Lipid profiles
`Total cholesterol (mmol ⁄ l)
`LDL cholesterol (mmol ⁄ l)
`VLDL cholesterol (mmol ⁄ l)
`HDL cholesterol (mmol ⁄ l)
`Triglycerides (mmol ⁄ l)
`Free fatty acids (mmol ⁄ l)
`Apolipoprotein B (g ⁄ l)
`Physical measure(s)
`Waist circumference (cm)
`
`)0.01 ()0.13 to 0.11)
`0.09 ()0.01 to 0.19)
`)0.11 ()0.19 to )0.03)
`0.01 ()0.01 to 0.03)
`)0.10 ()0.30 to 0.10)
`)0.07 ()0.11 to )0.03)
`)0.03 ()0.05 to )0.01)
`
`)2.36 ()3.10 to )1.62)
`
`)0.37 ()2.19 to 1.45)
`–0.53 ()1.65 to 0.59)
`1.72 (0.54 to 2.90)
`
`Data are least square means (95% CI); p-values (for treatment differences). CI, confidence interval; HDL, high-density lipoprotein; HOMA-B, homeostasis model assessment of beta-cell function; HOMA IR, HOMA
`insulin resistance; LDL, low-density lipoprotein; VLDL, very low-density lipoprotein.
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 6
`
`

`

`Liraglutide vs. sitagliptin 1-year results
`
`403
`
`Figure 3 Mean reductions in HbA1c by baseline category and proportions of participants reaching target end-points with
`1.2 mg liraglutide, 1.8 mg liraglutide and 100 mg sitagliptin from weeks 0–52. (A) Mean reductions in HbA1c from
`baseline to week 52 by baseline HbA1c category. (B) Percentage of participants achieving target HbA1c < 7% (ADA) or
`£ 6.5% (AACE). (C) Percentage of participants reaching the composite end-point of HbA1c < 7.0%, with no weight gain
`and no confirmed major or minor hypoglycaemia. In (B) and (C), solid bar portions represent percentages from weeks
`0–26, while shaded portions represent percentages from weeks 27–52. *p < 0.05 vs. sitagliptin; **p £ 0.01 vs. sitagliptin;
`***p < 0.001 vs. sitagliptin. p-values are derived from a logistic regression model with treatment and country as fixed
`effects and baseline HbA1c and body weight (for composite) as covariate(s). AACE, American Association of Clinical
`Endocrinologists; ADA, American Diabetes Association
`
`Mean changes in serum calcitonin, an indicator
`of C-cell hyperplasia, were small and there were no
`statistically significant differences between treatment
`groups. Mean calcitonin levels remained below the
`upper normal limit for both genders from baseline
`to 52 weeks. The proportions of subjects reporting
`thyroid-related treatment-emergent AEs were compa-
`rable across treatment groups (5.0%, 5.5% and 4.6%
`for liraglutide 1.2 mg, 1.8 mg and sitagliptin respec-
`
`tively) (Table S1). No cases of thyroid malignancy
`were found during the trial period (Table S2).
`
`Discussion
`
`Liraglutide produced sustained and greater reduc-
`tions in HbA1c, FPG and body weight compared
`with sitagliptin after 52 weeks of treatment, similar
`to results previously reported after week 26 (8).
`
`ª 2011 Blackwell Publishing Ltd Int J Clin Pract, April 2011, 65, 4, 397–407
`
`MPI EXHIBIT 1126 PAGE 7
`
`

`

`404
`
`Liraglutide vs. sitagliptin 1-year results
`
`Table 2 Participants with treatment-emergent adverse events during weeks 0–52
`
`Adverse events
`
`Serious adverse events*
`Deaths
`Severe adverse events
`Gastrointestinal disorders
`Musculoskeletal and connective
`tissue disorders
`Infections and infestations
`Neoplasms (benign, malignant
`and unspecified)
`Cardiac disorders
`Investigations
`Nervous system disorders
`Renal and urinary disorders
`Adverse events (of any severity)
`reported by > 5% of participants
`Gastrointestinal disorders
`Nausea
`Vomiting
`Diarrhoea
`Constipation
`Dyspepsia
`Infections and infestations
`Nasopharyngitis
`Influenza
`Nervous system disorders
`Headache
`Musculoskeletal and connective
`tissue disorders
`General disorders and administration-site
`conditions
`Metabolism and nutrition disorders
`Decreased appetite
`Investigations
`Skin and subcutaneous tissue disorders
`Respiratory, thoracic and mediastinal
`disorders
`Injury, poisoning and procedural
`complications
`Vascular disorders
`
`Liraglutide
`1.2 mg/day
`(n = 221)
`
`Liraglutide
`1.8 mg/day
`(n = 218)
`
`Sitagliptin
`100 mg/day
`(n = 219)
`
`10 (4.5)
`0
`12 (5.4)
`4 (1.8)
`3 (1.4)
`
`3 (1.4)
`1 (0.5)
`
`2 (0.9)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`158 (71.5)
`
`80 (36.2)
`48 (21.7)
`18 (8.1)
`20 (9.0)
`10 (4.5)
`8 (3.6)
`74 (33.5)
`27 (12.2)
`13 (5.9)
`40 (18.1)
`21 (9.5)
`39 (17.6)
`
`13 (6.0)
`1 (0.5)
`15 (6.9)
`5 (2.3)
`1 (0.5)
`
`3 (1.4)
`3 (1.4)
`
`1 (0.5)
`1 (0.5)
`0 (0.0)
`0 (0.0)
`167 (76.6)
`
`94 (43.1)
`60 (27.5)
`23 (10.6)
`27 (12.4)
`13 (6.0)
`15 (6.9)
`77 (35.3)
`32 (14.7)
`4 (1.8)
`48 (22.0)
`29 (13.3)
`45 (20.6)
`
`12 (5.5)
`2 (0.9)
`13 (5.9)
`4 (1Æ8)
`1 (0.5)
`
`3 (1.4)
`1 (0.5)
`
`1 (0.5)
`0 (0.0)
`2 (0.9)
`1 (0.5)
`139 (63.5)
`
`52 (23.7)
`12 (5.5)
`11 (5.0)
`14 (6.4)
`8 (3.7)
`5 (2.3)
`75 (34.2)
`31 (14.2)
`8 (3.7)
`44 (20.1)
`27 (12.3)
`45 (20.5)
`
`31 (14.0)
`
`32 (14.7)
`
`13 (5.9)
`
`25 (11.3)
`8 (3.6)
`21 (9.5)
`22 (10.0)
`16 (7.2)
`
`27 (12.4)
`12 (5.5)
`27 (12.4)
`20 (9.2)
`18 (8.3)
`
`19 (8.7)
`3 (1.4)
`16 (7.3)
`22 (10.0)
`21 (9.6)
`
`20 (9.0)
`
`20 (9.2)
`
`21 (9.6)
`
`16 (7.2)
`
`15 (6.9)
`
`10 (4.6)
`
`Data are number (%) of participants. A participant could experience more than one adverse
`event. *Liraglutide 1.2 mg group: acute myocardial infarction, myocardial infarction, epiglottic
`carcinoma, thyroid disorder, hypertensive crisis (relapsing), hypoesthesia, coxarthrosis defor-
`mans, worsening of coxarthrosis, haemorrhagic anaemia, haematochezia, infected seb

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