throbber
DI A BETES RESE ARCH A ND CLl N IC A L P RA CTICE 81 (2008) 161 1 6 8
`
`available at www.sciencedirect.com
`
`.. -.-,
`·•;" ScienceDjrect
`
`journal homepage: www .elsevier.com/locate/diabres
`
`ll"llnn&iio,nal Oi&betos ~
`
`n
`
`Dose-dependent improvement in glycemia with once-daily
`liraglutide without hypoglycemia or weight gain:
`A double-blind, randomized, controlled trial in Japanese
`patients with type 2 diabetes
`
`Y. Seino a,*, M.F. Rasmussen b, M. Zdravkovic b, K. Kaku c
`
`• Kansai Electric Power Hospital, 2 1 7 Fukushima Fukushima ku Osaka shi, 553 0003 Osaka, Japan
`b Nouo Nordisk NS, Bagsua.erd, Denmark
`c Department of Internal Medicine, Kawasaki Medical School, Okayama, Japan
`
`ARTICLE INFO
`
`ABSTRACT
`
`Article history:
`Received 6 November 2007
`Accepted 10 March 2008
`Published on line 20 May 2008
`
`Keywords:
`Liraglu ti.cl e
`GLP 1
`Type 2 diabetes mellitus
`lncretin
`
`Aims: To evaluate dose response efficacy and safety of once daily human GLP 1 analog
`liraglutide in Japanese subjects with type 2 diabetes.
`Methods : Patients (226, treated with diet with/without OADs, mean HbA1c 8.30%, mean BMl
`23.9 kwm2) were randomized after OAD discontinuation and washout to receive liraglutide
`0.1, 0.3, 0.6 or 0. 9 mg once daily, or placebo in double blind, parallel group design for 14
`weeks.
`Results: Liraglutide dose levels reduced HbA1c versus placebo (by 0.79%, 1.22%, 1.64% and
`1.85%, respectively; p < 0.0001 for linear contrast). Liraglutide 0.9 mw"day resulted in 75% of
`patients achieving HbA1c <7.0% and 57% achieving HbA1 c <6.5%. There were no major or
`minor hypoglycemic events. Llraglutide also reduced, with significant dose response (each
`p < 0.0001 for linear contrast) versus placebo: fasting plasma glucose (up to 2. 5 mmol/L),
`postprandial (0 3 h) glucose excursion (up to 12.8 mmol/(L h)); and increased postprandial
`insulin secretion (up to 23.0 µ.U/(mL h)) and beta cell function as evaluated by HOMA ~ (up
`to around 20.0 (µ,U/mL)/ (mwdL)). Body weight was unchanged; no development of liraglu
`tide antibodies was detected.
`Conclusions: Llraglutide was highly effective and well tolerated at doses up to 0.9 mwday in
`Japanese patients with type 2 diabetes, allowing glycemic control without weight gain or
`hypoglycemia.
`
`© 2008 Elsevier Ireland Ltd. All rights reserved.
`
`1.
`
`Introduction
`
`Type 2 diabetes is a multi.factorial disease, in which individuals,
`to a vaiying degree, exhibit failing beta cell function, weight
`gain and increasing insulin resistance over time. The relative
`
`importance of these factors differs between individuals and
`between populations, and it is well established that the
`pathophysiology of type 2 diabetes differs between Japanese
`and Caucasian patients. Insulin secretory capacity in Japanese
`patients with type 2 diabetes has been shown to be half of that
`
`• Corresponding author.
`E mail address: seino.yutaka@e2.kepco.co.jp (Y. Seino).
`see front matter © 2008 Elsevier Ireland Ltd. All rights reserved.
`0168 8227/$
`doil 0.1016/j. di ab res. 2008 .03.018
`
`MPI EXHIBIT 1038 PAGE 1
`
`MPI EXHIBIT 1038 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0001
`
`

`

`162
`
`d i a b e t e s r e s e a r c h a n d c l i n i c a l p r a c t i c e 8 1 ( 2 0 0 8 ) 1 6 1 1 6 8
`
`seen in Caucasian patients, a difference that is particularly
`pronounced for meal related secretion [1 3]. Japanese patients,
`in addition, typically have less insulin resistance compared
`to Caucasians and are generally less obese, with a typical
`body mass index (BMI) of 23 24 kg/m2 [4]. However, deteriorat
`ing glycemic control is often observed resulting in increased
`risk of microvascular and macrovascular complications. Avail
`able treatments for type 2 diabetes are often associated
`with undesirable effects (such as weight gain, hypoglycemia
`or edema)
`that
`limit
`their acceptability and potential
`for reaching treatment targets. There remains an urgent need
`for new therapies that address the multiple dysfunctions in
`type 2 diabetes without limitations of poor tolerability or
`acceptability.
`Glucagon like peptide 1 (GLP 1) is an incretin hormone
`with a broad spectrum of physiological actions. Analogs of
`GLP 1 may potentially be able to modulate the otherwise
`inevitable progression of type 2 diabetes [5 7]. Liraglutide is a
`long acting human GLP 1 analog that has a high degree of
`homology to native GLP 1 but, via acylation to myristic acid,
`achieves a longer plasma half life of 13 h and can thus be
`administered once daily [8 10].
`Previous studies with liraglutide in Japanese subjects have
`included single dose and 21 day stepwise dose escalation (up
`to 15 mg/kg) studies in healthy subjects and a 14 day dose
`escalation study (to 10 mg/kg) in subjects with type 2 diabetes
`[11]. Studies in non Japanese subjects with type 2 diabetes
`have shown that liraglutide is able to achieve sustained
`improvements in glycemic control with significant reduction
`in body weight and with a very low risk of hypoglycemia [7,12
`14]. Liraglutide has shown favorable effects on several
`parameters of beta cell function [10,15 17], and animals
`treated with liraglutide have shown increases in beta cell
`mass [18]. Recent data in non Japanese subjects with type 2
`diabetes also suggest potentially beneficial effects of liraglu
`tide on systolic blood pressure (SBP),
`triglycerides and
`cardiovascular biomarkers [19].
`The present study was conducted to assess the efficacy,
`safety and optimal dose for liraglutide during sustained
`treatment in Japanese subjects with type 2 diabetes.
`
`2.
`
`Subjects and methods
`
`This was a multicenter, double blind, randomized, parallel
`group phase 2 trial to evaluate the dose response relationship
`on glycemic control of four doses of liraglutide and placebo in
`Japanese subjects with type 2 diabetes. The primary efficacy
`endpoint was HbA1c after 14 weeks of treatment, with other
`measures of glycemic control (fasting plasma glucose (FPG),
`postprandial plasma glucose (PPG), self monitored plasma
`glucose profile) as secondary endpoints. The study was
`performed at 63 centers in Japan between March 2005 and
`May 2006. The study was performed in accordance with the
`Declaration of Helsinki, with informed consent of subjects and
`with approval of relevant ethics committees. Trial registration
`numbers were NCT00154414 and JapicCTI 050131.
`Patients included were to have type 2 diabetes treated
`with diet therapy with or without oral antidiabetic drug
`(OAD) monotherapy, HbA1c 7.0% and <10.0%, to be aged
`
`between 20 and 75 years and to have BMI <30 kg/m2. Patients
`treated with insulin or insulin sensitizer within 16 weeks, or
`receiving or expected to receive systemic corticosteroids,
`were excluded, as were those with impaired hepatic or renal
`function (serum glutamic oxaloacetic transaminase or
`serum glutamic pyruvic transaminase >80 IU/L, or serum
`creatinine 1.7 mg/dL), congestive heart failure (New York
`Heart Association class III or IV), unstable angina pectoris or
`myocardial
`infarction within 12 months, uncontrolled
`hypertension (SBP > 160 mmHg or diastolic blood pressure
`>100 mmHg), non stabilised proliferative retinopathy or
`maculopathy.
`The trial consisted of an 8 week run in period following
`screening (at which point OAD therapy, if any, was discontinued
`and during which an FPG <7.5 or 11.1 mmol/L at either of two
`visits was cause for exclusion), a 2 week dose titration period
`and a 12 week maintenance phase. Patients were randomized
`at the end of the run in period to liraglutide or placebo in four
`cohorts, each planned to contain 50 patients; in each cohort a
`planned 40 patients received one of four liraglutide doses
`(0.1 mg/day; 0.3 mg/day; 0.6 mg/day or 0.9 mg/day) and 10
`received placebo. Liraglutide doses in the 0.6 and 0.9 mg/day
`cohorts were increased from a starting dose of 0.3 0.6 mg/day
`after 1 week and, in the 0.9 mg/day cohort, by a further 0.3 mg/
`day after 2 weeks. Randomization was stratified by pre
`treatment (with or without OAD monotherapy) and was
`performed by sealed code by a central telephone registration
`centre; allocation to liraglutide or placebo in each cohort was
`blinded to subject and investigator. Dynamic allocation was
`employed in order to guarantee a balanced allocation within
`strata of pre trial treatment. Liraglutide was supplied by Novo
`Nordisk A/S (Copenhagen, Denmark) as a 6.25 mg/mL solution,
`and visually indistinguishable liraglutide vehicle as placebo.
`Trial medication was administered by abdominal subcutaneous
`injection using pre filled pen and needle set (FlexPen1,
`PenNeedle1, Novo Nordisk), once daily in the evening (at the
`same time every day for each subject, as far as possible).
`HbA1c and FPG were measured at baseline, after 2, 6 and 10
`weeks and at the end of the trial. A meal test, using a standard
`Japanese style breakfast, was performed at baseline and at
`the end of the trial; plasma glucose, insulin and glucagon
`being measured and the pre breakfast plasma glucose being
`taken as measure of FPG. Fasting pro insulin and C peptide
`levels were also recorded. All analyses were carried out by a
`central
`laboratory (Mitsubishi Kagaku BCL,
`Inc., Tokyo,
`Japan), except for a seven point plasma glucose profile which
`was measured before and approximately 2 h after each meal,
`and at bedtime by self monitoring at home using glucose
`meters (Glutest Ace1, Glutest PRO1, Sanwa Kagaku, Nagoya,
`Japan; Glucocard Diameter or Glucocard Diameter a, Arkray
`KDK Corp., Kyoto, Japan) before start of treatment and end of
`study, furthermore plasma liraglutide concentrations were
`measured using ELISA by Capio Diagnostics (Copenhagen,
`Denmark), and liraglutide antibodies were measured by
`radioimmunoassay by Novo Nordisk A/S Immunochemistry
`(Ma˚ løv, Denmark). Safety assessments included thyroid
`ultrasonography at screening and end of study, electrocar
`diography and clinical
`laboratory assessments including
`calcitonin. Beta cell function and insulin resistance were
`assessed from fasting insulin and plasma glucose values
`
`
`
`MPI EXHIBIT 1038 PAGE 2
`
`MPI EXHIBIT 1038 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0002
`
`

`

`D l ABE T E S R E S E ARC H AN D C LJ N I CA L P R AC T I C E 8r ( 2 008) 16 r r68
`
`163
`
`using the homeostasis model assessment (HOMA) 13 and
`HOMA R models, respectively (20):
`
`HOMA 13
`
`360 x fastinginsulin/(FPG 63)
`
`HOMA R
`
`(f?istinginsulin X FPG)/'WS
`
`where units of insulin and glucose µU/mL and mg/d.L, respec
`tively.
`Efficacy endpoints were analyzed using data from all
`patients who received trial product and for whom any efficacy
`data were recorded. Safety endpoints were recorded for any
`patient receiving any dose of trial product. HbA1c and other
`efficacy endpoints were analyzed using an ANOVA model with
`dose group and pre trial treatment (with and without OAD) as
`fixed effects and value at baseline as covariate. The existence
`of a monotonic dose response relationship was assessed for
`each endpoint using an F test for linear contrast with contrast
`coefficient of ( 2, 1, 0, 1, 2). A sequentially rejecting Dunnett
`test was used to perform pairwise comparison of HbA1c levels
`in liraglutide groups versus placebo. Required sample size was
`calculated as 32 per dose group, based on a two sided 5%
`significance level for the F test for linear contrast in the
`primary endpoint with 80% power. Randomization of a
`minimum of 200 subjects was planned, allowing for an
`expected dropout rate of 15%.
`
`3.
`
`Results
`
`A total of 372 patients were screened, of whom 226 were
`randomized to study cohorts and exposed to treatment
`
`(Table 1). Mean HbA1c was 8.30%; baseline characteristics
`were comparable across study groups (Table 1). Sixteen
`patients withdrew from the study, the largest number from
`the groups allocated to placebo (eight withdrawals).
`Mean HbA1c was reduced by all liraglutide dose levels
`relative to placebo (Fig. 1), the magnitude of the treatment
`effect ranging from 0.79 percentage points in the 0.1 mg/day
`group to 1.85 percentage points with 0.9 mg/day liraglutide
`(Table 2). HbA1c levels below ?.CJ% were achieved by 22%, 43%,
`62% and 75% of patients receiving liraglutide, 0.1, 0.3, 0.6 and
`0.9 mg/day, respectively, and 9% of those receiving placebo.
`The proportion of patients achievingHbA1c <6.5% ranged from
`7% to 57% in the liraglutide dose groups, and 2% in the placebo
`group (Table 2). A monotonic dose response relationship was
`confirmed in HbA1c level, with p < 0.0001 for linear contrast.
`Pairwise comparison showed statistically significant differ
`ences in HbA1c levels versus placebo in all liraglutide groups at
`14weeks.
`Self monitored seven point plasma glucose profiles
`showed reductions in mean glucose levels across the day;
`mean AUC values for glucose across the seven point profiles
`were lower than placebo with all liraglutide doses above
`0.1 mg/day (Table 2). Dose response relationship was con
`firmed with p < 0.0001 for linear contrast. FPG levels also
`showed reductions with all liraglutide dose levels versus
`placebo ranging from 0.76 to 2.48 mmol/L, with p < 0.0001 for
`linear contrast. A reduction in FPG was already evident at the
`2 week visit, which was the first timepoint following start of
`li.raglutide administration.
`Glucose levels following a standard breakfast after 14
`weeks showed a significant dose response in glucose AUCo-
`
`Table 1 - Patient flow and baseline characteristics of treatment groups
`Screened
`372
`
`Screening failure
`Met ~ 1 exclusion criterion
`Failed ;::1 inclusion criterion
`Other
`
`Randomized
`
`Withdrew
`Adverse event
`Non compliance with protocol
`Ineffective therapy
`Other
`Completed
`
`100
`39
`15
`
`Placebo
`46
`
`1
`0
`1
`6
`38
`
`0.1mg
`45
`
`0.3mg
`46
`
`0.6mg
`45
`
`0.9mg
`44
`
`0
`1
`1
`0
`43
`
`0
`1
`1
`1
`43
`
`0
`0
`1
`0
`44
`
`1
`0
`0
`1
`42
`
`Age (years)
`Mean (S.D.)
`Gender: male/female (n/n)
`Body weight (kg), mean (S.D.)
`BM! (kg/m2), mean (S.D.)
`Duration of diabetes (years) , mean (S.D,)
`
`57.5
`(8.7)
`29/17
`62.78 (10.88)
`23.77 (2.63)
`7.48 (5.65)
`
`56.5
`(8.4)
`3V14
`64.82 (10.29)
`24.26 (277)
`7.15 (5.14)
`
`56.8
`(8.8)
`32/14
`62.42 (11.18)
`23.93 (3.09)
`6.78 (4.69)
`
`60.0
`(7.0)
`28/17
`61.97 (9.40)
`23.74 (2.78)
`8.87 (6.77)
`
`55.5
`(7.6)
`31/13
`62.36 (10.65)
`23.59 (3.04)
`7.62 (4.92)
`
`Diabetic complications
`Retinopathy (n)
`Nephropathy (n)
`Neuropathy (n)
`Other (n)
`
`Treatment includes OAD (n)
`
`10
`6
`12
`2
`
`22
`
`17
`9
`14
`0
`
`21
`
`13
`9
`12
`0
`
`22
`
`11
`8
`7
`1
`
`19
`
`5
`3
`11
`0
`
`20
`
`MPI EXHIBIT 1038 PAGE 3
`
`MPI EXHIBIT 1038 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0003
`
`

`

`164
`
`DI AB E TE S R E SE AR C H AN D CLI N I C A L PRA CTI CE 81 (2008) 161 r 6 8
`
`9.0
`
`8.5 I
`
`8.0
`
`l 1.s
`
`(.) 7.0
`<(
`.0 6.5
`I
`6.0
`
`5.5
`
`0
`
`I
`
`Placebo
`
`0.1
`0.6
`0.3
`Liraglutide dose ( mg/day)
`
`0.9
`
`Fig. 1 - Dose-response for HbA1c with increasing doses of
`liraglutide. Data are means ± S.E. for HbA1c after 14 w eeks.
`
`3 h , with reductions versus placebo at all liraglutide dose levels
`and p < 0.0001 for linear contrast (Table 2). One hour PPG
`levels showed reductions with all liraglutide doses except for
`the lowest, while all liraglutide doses showed reductions in 2
`and 3 h PPG versus placebo (Fig. 2).
`Insulin levels during the 3 h following the standard
`breakfast were also increased by the t hree highest liraglutide
`dose levels (Table 2), also showing dose response (p < 0.0001
`
`for linear contrast). This was accompanied by significant dose
`relationship in beta cell function (HOMA B), with increases
`versus placebo at 0.3, 0.6 and 0.9 mg/day liraglutide (also
`p < 0.0001 for linear contrast). Pro insulin:insulin ratio and
`pro insulin:C peptide ratios were decreased in all liraglutide
`dose groups {p 0.0008 and p < 0.0001 for linear contrast,
`respectively). There was no evidence of change in insulin
`resistance (HOMA R).
`No relevant changes in body weight occurred during the
`study, a reduction of 0.95 kg (from baseline 62 kg) occurring in
`the placebo group and changes in the liraglutide groups
`ranging from +0.13 kg to 0.48 kg versus baseline (Table 2).
`No major hypoglycemic events were reported in any study
`group during the trial. Likewise, no episodes of minor
`hypoglycemia occurred (symptomatic events confirmed by
`plasma glucose <3.1 mmoVL).
`Plasma levels of liraglutide remained steady in all dose
`groups from 2 to 14 weeks; mean (S.D.) plasma concentrations
`at 14 weeks were 1.98 (0.93), 4.31 (1.68), 9.42 {4.13) and 10.08
`{4.21) nmoVL in subjects receiving 0.1, 0.3, 0.6 and 0.9 m!q'day
`liraglutide, respectively. No treatment related increase in
`liraglutide antibodies occurred during the study. An assay
`specific normal range was defined from a phase 2 study in
`Caucasians with type 2 diabetes (21). In this trial of Japanese
`subjects, antibody levels were above t he pre defined assay
`cut off level in 11/226 patients at baseline and in 13/207
`
`Table 2 - Effect of liraglutide on measures of glycemia, beta-cell function and body weight
`Placebo
`0.1mg
`0.3mg
`0.6mg
`
`0.9mg
`
`8.43 (1.02)
`8.52 (1.23)
`
`8.50 (0.84)
`7.78 (0.91)
`0.79 ( 1.08,
`0.50)
`
`HbA,c (%), mean (S.D.)
`Baseline
`Week 14
`Liraglutide placebo,
`mean (95% CQ
`Patients achieving HbA1c levels at week 14, n (%)
`< 5.8%
`0 (0%)
`0(0%)
`~ 5.8%, < 6.5%
`1 (2.2%)
`3 (6.7%)
`;,:6.5%, < 7.0%
`3 (6.5%)
`7 (15.6%)
`34 (73.9%)
`33 (73.3%)
`~ 7.0%
`7 point SMPG profile: AUC(7 point) (mmoV(L h)), mean (S.D.)
`Baseline
`170.8 (320)
`173.7 (32.0)
`Week 14
`165.6 (35.1)
`153.1 (30.3)
`Liraglutide. placebo,
`13.7 ( 25.3,
`mean (95% CQ
`2.1)
`Fasting plasma glucose (mmoVL), mean (S.D.)
`Baseline
`9.99 (1.71)
`10.03 (1. 73)
`Week 14
`9.77 (2.47)
`9.03 (1.76)
`Llraglutide placebo,
`0.76 ( 1.40,
`mean (95% CQ
`0.11)
`1 h postprandial plasma glucose (mmoVL), mean (S.D.)
`Baseline
`16.91 (2.42)
`16.57 (261)
`Week 14
`16.03 (2.72)
`15.00 (270)
`Llraglutide placebo,
`0.83 ( 1.81,
`mean (95% CQ
`0.15)
`2 h postprandial plasma glucose (mmoVL), mean (S.D.)
`Baseline
`15.97 (292)
`16.05 (3.04)
`Week 14
`14 .95 (3.49)
`13.57 (3.15)
`
`8.24 (0.92)
`7.17 (1.01)
`1.22 ( 1.50,
`0.93)
`
`8.21 (0.83)
`6.71 (0.92)
`1.64 ( 1.93,
`1.35)
`
`8.12 (0.98)
`6.45 (0.n)
`1.85 ( 2 14,
`1.56)
`
`1 (2.2%)
`12 (26,1%)
`7 (15.2%)
`23 (50.0%)
`
`164.7 (32.2)
`137.8 (29.0)
`25.0 ( 36.6,
`13.3)
`
`9.84 (1.73)
`8.42 (1.96)
`1.26 ( 1.89,
`0.61)
`
`16.08 (2.61)
`13.78 (2.87)
`1 .75 ( 2.73,
`0.76)
`
`6 (13.3%)
`18 (40.0%)
`4 (8.9%)
`15 (33.3%)
`
`9 (20.5%)
`16 (36.4%)
`8 (18.2%)
`9 (20.5%)
`
`167 .0 (35.1)
`123.5 (23.6)
`40.2 ( 51.9,
`28.6)
`
`1.55.5 (27. 7)
`116.8 (27.4)
`41.9 ( 53.8,
`29.9)
`
`9.84 (1.23)
`7.41 (1.23)
`2.27 ( 2.92,
`1.63)
`
`16.47 (2.99)
`12.99 (2.78)
`2.78 ( 3.74,
`1.81)
`
`9.34 (1.36)
`6.91 (122)
`248 ( 3.13,
`1.82)
`
`15.61 (289)
`11.39 (2.68)
`3.85 ( 4.84,
`2.86)
`
`14.n (2.74)
`11.48 (2.89)
`
`16.01 (3.78)
`10.74 (294)
`
`14.42 (3.44)
`8.89 (3.16)
`
`F test for
`linear
`contrast
`
`p < 0.0001
`
`Not applicable
`
`p < 0.0001
`
`p < 0.0001
`
`Not tested
`
`MPI EXHIBIT 1038 PAGE 4
`
`MPI EXHIBIT 1038 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0004
`
`

`

`DI ABE T ES RESEAR CH AN D CLIN IC AL PRAC TI CE 8 r (200 8) 1 6 r r6 8
`
`165
`
`Table 2 (Continued)
`
`Placebo
`
`0.1mg
`
`0.3mg
`
`0.6mg
`
`0.9mg
`
`liraglutide placebo,
`mean (95% CI)
`
`1.43 ( 2.58,
`0.29)
`
`2.83 ( 3.98,
`1.68)
`
`4.22 ( 5.35,
`3.09)
`
`5.23 ( 6.39,
`1.07)
`
`3 h postprandial plasma glucose mmol/L), mean (S.D.)
`Baseline
`13.86 (3.09)
`13.34 (2.91)
`Week 14
`12.57 (3.10)
`11.24 (2.68)
`liraglutide placebo,
`1 .56 ( 2.53,
`mean (95% CI)
`0.58)
`
`AUC(0 3 h) plasma glucose (mmol/(L h)), mean (S.D.)
`Baseline
`44.35 (6.88)
`44.50 (7.19)
`Week 14
`41.86 (8.19)
`38.64 (7.40)
`liraglutide placebo,
`3.30 ( 5.88,
`mean (95% CI)
`0.73)
`
`AUC (0 3 h) insulin (µ.U/(mL h)), mean (S.D.)
`Baseline
`58.84 (32.47)
`54.12 (30.98)
`Week 14
`59.38 (33.72)
`64.23 (33.17)
`liraglutide placebo,
`10.69 ( 6.00,
`mean (95% CI)
`27.39)
`
`Beta cell function (HOMA 13) ((µU/IllQ/(mg/dL)) , mean (S.D.)
`Baseline
`21.45 (14.75)
`19.36 (13.34)
`Week 14
`22.34 (21.77)
`27.56 (19.39)
`liraglutide placebo,
`7.79 ( 0.11,
`mean (95% CI)
`15.70)
`
`Insulin resistance (HOMA R), mean (S.D.)
`Baseline
`2 78 (1. 90)
`Week 14
`2.35 (1.33)
`liraglutide placebo,
`mean (95% CI)
`
`2.68 (1.69)
`2 .82 (1.98)
`0.54 ( 0.15,
`1.23)
`
`Pro insulin:insulin ratio ((pmol/L)/(µU/mL)), mean (S.D.)
`Baseline
`1 .56 (1.32)
`1.52 {1.17)
`Week 14
`1.58 (1.25)
`1.04 (0.66)
`liraglutide placebo,
`0.53 ( 0.87,
`mean (95% CI)
`0.20)
`
`Pro insulin:C peptide ratio ((pmol/L)/(ng/mL)), mean (S.D.)
`Baseline
`428 (2. 90)
`3.98 (2.45)
`Week 14
`3.87 (2.32)
`3.22 (2.35)
`liraglutide placebo,
`0.54 ( 1.25,
`mean (95% CI)
`0.18)
`
`12.49 (2.96)
`9.39 (2.86)
`2.79 ( 3.77,
`1.82)
`
`42.02 (6.72)
`34.16 (7.12)
`6.42 ( 9.01,
`3.84)
`
`71.69 (48.06)
`95.29 (69.12)
`19.69 (3.01,
`36.36)
`
`23.97 (18.51)
`36.82 (28.16)
`11.40 (3.58,
`19.21)
`
`3.02 (2.00)
`3.34 (3.12)
`0.83 (0.15,
`1.51)
`
`1.44 (1.03)
`1.04 (0.77)
`0.50 ( 0.83,
`0.17)
`
`4.05 (2.28)
`294 (2.08)
`0.84 ( 1.55,
`0.13)
`
`13.52 (3.48)
`8.13 (228)
`4.53 ( 5.49,
`3.57)
`
`12.27 (2.92)
`6.86 (1.88)
`5.23 ( 6.21,
`4.26)
`
`44.18 (8.54)
`31.46 (6.39)
`10.30 ( 12.85,
`7.76)
`
`40.84 (7.85)
`27.12 (6.41)
`12.80 ( 15.40,
`10.20)
`
`60.53 (32.04)
`102.56 (78.51)
`41.34 (25.12,
`57,55)
`
`22.06 (13.69)
`44.06 (30.27)
`20.99 (13.17,
`28.80)
`
`293 (1.63)
`261 (1.81)
`0.14 ( 0.54,
`0.82)
`
`1.50 (127)
`0.86 (0.65)
`0.69 ( 1.02,
`0.36)
`
`4.16 (3.56)
`241 (1.47)
`1.40 ( 2 11,
`0.69)
`
`63.83 (45.73)
`88.97 (67.74
`23.02 (6.52,
`39.52)
`
`22.99 (18.62)
`44.04 (32.44)
`19.82 (11.96,
`27.67)
`
`2.58 (1.86)
`2.05 (1.47)
`o.v ( 0.85,
`0.52)
`
`1.51 (1.01)
`1.00 (0.94)
`0.56 ( 0.89,
`0.23)
`
`3.60 (1.97)
`2.28 (1.53)
`1.32 ( 2.04,
`0.61)
`
`F test for
`linear
`contrast
`
`Not tested
`
`Not tested
`
`p < 0.0001
`
`p < 0.0001
`
`p < 0.0001
`
`p = 0.3521
`
`p = 0.0008
`
`p < 0.0001
`
`Body weight (kg), mean (S.D.)
`Baseline
`6200 (10.97)
`Week 14
`61.05 (10.89)
`liraglutide placebo,
`mean (95% CI)
`Values for baseline and week 14 are mean (S.D.). Liraglutide placebo differences (at week 14) were calculated using an ANOVA model with dose
`group and pre treatment as fixed effects and baseline value as covariate. SMPG, self measured plasma glucose.
`
`61.48 (10.55)
`61.00 (11.07)
`0.46 ( 022, 1.14)
`
`p = 0.2481
`
`64.26 (10.46)
`64.21 (10.67)
`0.87 (0.19, 1.55)
`
`61.54 (10.98)
`61.67 (11.39)
`1.08 (0.41, 1.75)
`
`61.52 (9.46)
`61.42 (9.68)
`0.84 (0.16, 1.51)
`
`patients at week 15 (post treatment). No subject's antibody
`levels at week 15 exceeded the maximum level recorded at
`baseline.
`A total of 154 patients (68%) experienced adverse events
`during the study, most commonly infections and gastro
`intestinal disorders. Distribution of adverse events was similar
`across treatment groups for infection/infestation disorders;
`incidences of gastrointestinal disorders were placebo, 24%;
`liraglutide 0.1 mg/day, 18%; 0.3 mg/day, 15%; 0.6 mg/day, 31 %;
`0.9 mg/day, 30%. Two patients, one receiving placebo, and one
`receiving 0.9 mg/day liraglutide, withdrew due to adverse
`
`events (abdominal discomfort and viral gastroenteritis; upper
`abdominal pain, respectively).
`Seven patients entering the study experienced serious
`adverse events; in five cases these events occurred before
`receiving any study medication. The two remaining events
`concerned a subject with suspected papillary thyroid carci
`noma (in the 0.6 mg/day liraglutide group) and a subject who
`suffered an alcohol related fall (in the 0.9 mg/day liraglutide
`group). Neither event was considered by investigators to be
`related to study treatment and both patients completed the
`study.
`
`MPI EXHIBIT 1038 PAGE 5
`
`MPI EXHIBIT 1038 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0005
`
`

`

`166
`
`d i a b e t e s r e s e a r c h a n d c l i n i c a l p r a c t i c e 8 1 ( 2 0 0 8 ) 1 6 1 1 6 8
`
`o Placebo
`.t. 0.1 mg/day
`• 0.3 mg/clay
`♦ 0.6 mg/day
`■ 0.9 mg/day
`
`___
`--
`
`2---__
`
`P ~;=:~~=:;
`----------- ~-----------
`---
`~---- -----~
`----- ----..
`--------
`
`.
`1>
`
`18
`
`16
`
`14
`
`12
`
`10
`
`8
`
`6
`
`4
`
`-:co
`0
`E
`E
`Q)
`(/)
`0
`()
`:J
`0)
`
`(v
`E
`(/)
`(v
`
`0::
`
`2
`o~~ - - - - - -~ - - - - -~ - - - - -~
`1 hour
`Before
`2 hours
`3 hours
`breakfast
`after breakfast
`
`Fig. 2 – Postprandial plasma glucose profiles following a
`standard breakfast after 14 weeks’ treatment with
`liraglutide. Data are means W S.E.
`
`4.
`
`Discussion
`
`In this study, we investigated efficacy and safety from a dose
`response perspective with four doses of liraglutide and
`placebo in Japanese patients with type 2 diabetes. We
`demonstrated significantly dose dependent improvements
`in key parameters of glycemia HbA1c, FPG, PPG, seven point
`self monitored plasma glucose
`and beta cell
`function
`following 14 weeks’ treatment with liraglutide. Treatment
`was weight neutral, no major or minor hypoglycemic episodes
`occurred, and there was no treatment related development of
`liraglutide antibodies during the trial.
`The magnitude of HbA1c reduction with liraglutide in this
`study is comparable to those reported in non Japanese
`subjects by Vilsbøll and colleagues, where reductions of up
`to 1.74% points were achieved from similar baseline HbA1c
`levels of 8.1 8.5% [14]. Madsbad and colleagues reported a
`0.74% point reduction in HbA1c with liraglutide, 0.75 mg/day,
`from a somewhat better controlled baseline of 7.4% [13].
`A clear dose response relationship for HbA1c reduction was
`shown in this study. All doses of liraglutide tested showed a
`degree of efficacy, with a plateau of dose response at 0.9 mg/
`day. A dose of 0.9 mg/day appears to be highly effective and, of
`the doses tested, offers the optimal efficacy/tolerability
`balance in Japanese patients with type 2 diabetes. Indeed,
`the reductions in HbA1c in this study allowed a large majority
`of patients receiving the higher doses of liraglutide to achieve
`satisfactory glycaemic control: in the 0.9 mg/day group 75% of
`patients achieved HbA1c <7.0% and more than 50% achieved
`HbA1c <6.5% (‘good’ or ‘excellent’ categories in current Japan
`Diabetes Society guidance) without safety concerns or limited
`tolerability [22]. As in previous studies with liraglutide in non
`Japanese subjects [7,12,13,21], the risk of hypoglycemia is low
`with this agent, with no confirmed minor or major hypogly
`cemic episodes reported in this study. This combination of
`efficacy and hypoglycemic tolerability is highly encouraging.
`Previous longer term studies with liraglutide, largely in
`Caucasians, have reported significant weight
`reduction
`[7,12,14]. In the present study, body weight remained stable.
`
`This is likely to reflect the very different initial characteristics
`typical for Japanese patients: initial BMI values in the studies
`by Feinglos et al. and Nauck et al. were 34.5 kg/m2 and 31.3
`32.4 kg/m2, respectively, compared with 23.9 kg/m2 in our
`Japanese cohort [7,12].
`Japanese and Caucasian patients also differ, typically, in
`the nature of their type 2 diabetes. Japanese subjects have
`greatly impaired first phase insulin secretion from an early
`stage in type 2 diabetes, which accelerates the onset of
`postprandial hyperglycemia, whereas in some other groups
`type 2 diabetes is a disease of profound insulin resistance [23
`26]. The improvements in overall metabolic control as well as
`in PPG levels with liraglutide in Japanese individuals are
`therefore welcome and consistent with previous data with
`liraglutide in non Japanese subjects [15]. Taken together,
`these data show that
`liraglutide is highly effective in
`populations where either increased insulin resistance or
`impaired insulin secretion is the main contributor to meta
`bolic derangement. The 31% reduction in postprandial glucose
`AUC seen in this study with 0.9 mg/day liraglutide compares
`favorably with the 23% reduction reported in Caucasian
`subjects by Juhl et al.
`[15]. Evidence from this study of
`improvements in beta cell function was also encouraging.
`Pro insulin:insulin ratio, a postulated indicator of insulin
`secretory dysfunction [27,28] was reduced by all doses of
`liraglutide versus placebo, as seen in previous studies with
`liraglutide [13]. Beta cell function (HOMA B) was improved by
`liraglutide, as seen by Madsbad et al.
`[13]; the lack of
`improvement in HOMA R is likely to reflect the lack of effect
`on body weight and the low initial levels of insulin resistance
`in these Japanese subjects.
`Liraglutide was well tolerated in this study, with adverse
`event profiles comparable to placebo and few withdrawals
`from treatment. No safety concerns were raised.
`In conclusion, we have shown that liraglutide, at doses of
`up to 0.9 mg/day, is effective in improving glycaemic control
`and well tolerated in Japanese patients with type 2 diabetes,
`with encouraging indications of beneficial effects on beta cell
`function. An agent that allows three quarters of a population
`of subjects with type 2 diabetes to achieve good glycaemic
`control, with a low risk of hypoglycemia and no weight gain, is
`well tolerated and requires only a single daily administration
`will be welcome in clinical practice.
`
`Conflict of interest statement
`
`We have a competing interest to declare. Professor Seino and
`Professor Kaku and currently on an advisory board for Novo
`Nordisk Pharma Ltd., Tokyo,
`Japan. Mads Rasmussen is
`employed by, and holds shares in, Novo Nordisk A/S,
`Copenhagen, Denmark.
`
`Acknowledgments
`
`This study was conducted as part of the development program
`for liraglutide, and was supported by Novo Nordisk Pharma
`Ltd., Japan. A list of participating investigators is presented in
`Appendix A.
`
`
`
`MPI EXHIBIT 1038 PAGE 6
`
`MPI EXHIBIT 1038 PAGE 6
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1038-0006
`
`

`

`d i a b e t e s r e s e a r c h a n d c l i n i c a l p r a c t i c e 8 1 ( 2 0 0 8 ) 1 6 1 1 6 8
`
`167
`
`Appendix A. Investigators
`
`Takashi Sasaki, Director, Keijinkai Sasaki Hospital, Sap
`poro City, Hokkaido; Fuminobu Okuguchi, Director, Okuguchi
`Clinic of Internal Medicine, Sendai City, Miyagi; Kenichi
`Yamada, Director, Kenichi Yamada Clinic, Tagajo City, Miyagi;
`Hiroaki Seino, Director, Diabetic Centre, Ohta Nishinouchi
`Hospital, Koriyama City, Fukushima; Takeshi Osonoi, Direc
`tor, Iryo Houjin Kenseikai, Naka Memorial Clinic, Naka City,
`Ibaraki; Yukiko Onishi, Director of Clinical Research Division,
`The Institute for Adult Diseases Asahi Life Foundation
`Marunouchi Hospital, Tokyo; Yasuhiko Iwamoto, Director,
`Diabetic Centre, Tokyo Women’s Medical University School of
`Medicine, Tokyo; Kouichi Hirao, President, H.E.C Science
`Clinic, Yokohama City, Kanagawa; Rika Usuda, Director,
`Toyama Prefectural Central Hospital, Toyama City, Toyama;
`Toshinari Takamura, Assistant Professor, Kanazawa Univer
`sity Hospital Endocrinology and Metabolism Department,
`Kanazawa City Ishikawa; Tatsuhide Inoue, Director of Live
`hood Support, Shizuoka Kenritsu Sougo Hospital, Shizuoka
`City Shizuoka; Nobuyuki Abe, Director, Internal Medicine, ABE
`Hospital, Ohita City Ohita; Hideaki Jinnouchi, Sub Director,
`Diabetes Care Center,
`Jinnouchi Clinic, Kumamoto City,
`Kumamoto; Shinji Taneda, Director, Manda Memorial Hospi
`tal, Sapporo City, Hokkaido; Masato Odawara, Professor,
`Tokyo Medical University Hospital, Tokyo; Keiko Arai, Direc
`tor, Arai Clinic, Yokohama City, Kanagawa; Hiroshi Tanaka,
`Dept. Chief Director, Social
`Insurance Chukyo Hospital
`Endocrinology and Metabolism Department, Nagoya City,
`Aichi; Syuji Nakamura, Director, Iryo Houjin Shadan Kowakai
`Heiwadai
`Hospital, Miyazaki City, Miyazaki;
`Hiroki
`Yokoyama, Director,
`Jiyugaoka Clinic,
`Internal Medicine,
`Obihiro City, Hokkaido; Takashi Sasaki, Director, Kashiwa
`Hospital,
`Jikei University School of Medicine Diabetes,
`Metabolism and Endocrinology Dept., Kashiwa City, Chiba;
`Masami Nemoto, Chief Director, Jikei University School of
`Medicine Diabetes, Metabolism and Endocrinology Dept.,
`Tokyo; Takashi Kadowaki, Professor, University of Tokyo
`Hospital Department of Metabolic Diseases, Tokyo; Kiyokazu
`Matoba, President, Matoba Diabetes Clinic, Ebina City, Kana
`gawa; Kunikazu Kondo, Director, Aichi Prefectural Welfare
`Federation of Agricultural Co operative Associations Anjo
`Kosei Hospital, Anjo City, Aichi; Hiroshi Sobajima, Internal
`Department Director, Ogaki Municipal Hospital Department of
`Diabetology and Nephrology, Ogaki City, Gifu; Yutaka Noto,
`Director, National Hospital Organisation Kanazawa Medical
`Center, Kanazawa City, Ishikawa; Yukihiro Nagai, Director,
`Nagai Internal Medicine Clinic, Kanazawa City,
`Ishikawa;
`Akira Okada, Director, Okada Clinic, Fukuoka City, Fukuoka;
`Yoshifumi Yokomizo, Director, Iryo Houjin Yokomizo Internal
`Medicine Clinic, Kitakyusyu City Fukuoka; Shigeru Miyazaki,
`Internal Depart

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