throbber
E m e r g i n g T r e a t m e n t s a n d T e c h n o l o g i e s
`O R I G I N A L
`A R T I C L E
`
`The Effect of Liraglutide, a Long-Acting
`Glucagon-Like Peptide 1 Derivative, on
`Glycemic Control, Body Composition, and
`24-h Energy Expenditure in Patients
`With Type 2 Diabetes
`
`1
`HELLE HARDER, MSC
`LENE NIELSEN, MSCODONT
`
`2
`
`2
`TU D.T. THI, MSC
`ARNE ASTRUP, MD, DRMEDSCI
`
`1
`
`OBJECTIVE — Glucagon-like peptide (GLP)-1 is a gut hormone that exerts incretin effects
`and suppresses food intake in humans, but its therapeutic use is limited due to its short half-life.
`This was a randomized, double-blind, parallel-group, placebo-controlled trial investigating the
`effect of the long-acting GLP-1 derivative liraglutide (NN2211) on glycemic control, body
`weight, body composition, and 24-h energy expenditure in obese subjects with type 2 diabetes.
`
`RESEARCH DESIGN AND METHODS — Thirty-three patients (mean ⫾ SD) aged
`60.0 ⫾ 9.5 years, with HbA1c 7.5 ⫾ 1.2% and BMI 36.6 ⫾ 4.1 kg/m2, were randomized to
`treatment with a single daily subcutaneous dose of 0.6 mg liraglutide (n ⫽ 21) or placebo (n ⫽
`12) for 8 weeks. In addition to weight and glycemic parameters, body composition was assessed
`by dual-energy X-ray absorptiometry (DEXA) scanning and 24-h energy expenditure in a respi-
`ratory chamber.
`
`RESULTS — After 8 weeks, liraglutide reduced fasting serum glucose (liraglutide, ⫺1.90
`mmol/l, and placebo, 0.27 mmol/l; P ⫽ 0.002) and HbA1c (liraglutide, ⫺0.33%, and placebo,
`0.47%; P ⫽ 0.028) compared with placebo. No change in body weight was detected (liraglutide,
`⫺0.7 kg, and placebo, ⫺0.9 kg; P ⫽ 0.756). There was a nonsignificant trend toward a decrease
`in total fat mass (liraglutide, ⫺0.98%, and placebo, ⫺0.12%; P ⫽ 0.088) and toward an increase
`in lean body mass (liraglutide, 1.02%, and placebo, 0.23%; P ⫽ 0.118) in the liraglutide group
`compared with the placebo group. Twenty-four-hour energy expenditure was unaffected by the
`treatment (liraglutide, ⫺12.6 kJ/h, and placebo, ⫺13.7 kJ/h; P ⫽ 0.799).
`
`CONCLUSIONS — Eight weeks of 0.6-mg liraglutide treatment significantly improved gly-
`cemic control without increasing weight in subjects with type 2 diabetes compared with those on
`placebo. No influence on 24-h energy expenditure was detected.
`
`Diabetes Care 27:1915–1921, 2004
`
`G lucagon-like peptide (GLP)-1 has
`
`several effects that make it a poten-
`tial candidate molecule for the
`treatment of type 2 diabetes. GLP-1 is a
`
`peptide hormone secreted from the L-cells
`in the lower gut, i.e., the distal jejunum,
`ileum, and colon/rectum, in response to
`ingestion of carbohydrates, lipids, and
`
`● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ●
`
`From the 1Department of Human Nutrition, LMC, The Royal Veterinary and Agricultural University, Fred-
`eriksberg, Denmark; and 2Novo Nordisk, Bagsvaerd, Denmark.
`Address correspondence and reprint requests to Arne Astrup, Research Department of Human Nutrition,
`LMC, The Royal Veterinary and Agricultural University, Rolighedsvej 30, DK-1958 Frederiksberg, Denmark.
`E-mail: ast@kvl.dk.
`Received for publication 8 January 2004 and accepted in revised form 15 May 2004.
`H.H. left the Department of Human Nutrition at The Royal Veterinary and Agricultural University to take
`a position at Novo Nordisk in September 2003.
`Abbreviations: AUC, area under the curve; DEXA, dual-energy X-ray absorptiometry; GLP, glucagon-like
`peptide; HOMA, homeostasis model assessment; OHA, oral hypoglycemic agent; VAS, visual analog score.
`A table elsewhere in this issue shows conventional and Syste`me International (SI) units and conversion
`factors for many substances.
`© 2004 by the American Diabetes Association.
`
`mixed meals (1–3). GLP-1 stimulates
`postprandial insulin secretion and acts as
`an incretin hormone, thus potentiating
`glucose-stimulated insulin release. The
`incretin effect refers to the increased insu-
`lin response elicited by oral glucose com-
`pared with the response resulting from an
`isoglycemic intravenous infusion (4). In
`addition, GLP-1 has a suppressive effect
`on glucagon release and hepatic glucose
`output (5).
`Treatment of hyperglycemia in type 2
`diabetic patients with GLP-1 has received
`much attention, but additional therapeu-
`tic indications could be envisioned.
`Within the last few years, the ability of
`GLP-1 to decrease appetite and energy in-
`take has been established (6). A reduced
`food intake would support weight reduc-
`tion attempts in patients with type 2 dia-
`betes. The appetite-reducing effect might
`partly be related to the ability of GLP-1 to
`decrease gastric emptying and reduce gas-
`tric motility (2). The mechanisms in-
`volved in the regulation of appetite and
`food intake are, however, complex and
`not fully understood; this also applies for
`the mechanisms mediating the anorectic
`effects of GLP-1. Furthermore, GLP-1 re-
`ceptors have been found in various tis-
`sues, such as pancreas, stomach,
`intestine, brain, and heart (7). The ano-
`rectic effect of GLP-1 has been shown in
`both healthy lean and obese people as
`well as in type 2 diabetic patients (6,8 –
`12). This effect has been reported from
`studies (13,14) of up to 2 days in dura-
`tion. In addition, obese subjects have
`been shown (10) to have an attenuated
`GLP-1 release in response to meals. A
`study (15) in obese type 2 diabetic sub-
`jects with continuous infusion of GLP-1
`using insulin pumps demonstrated that,
`aside from a glucose-lowering effect,
`GLP-1 also exhibited weight-reducing
`properties. After 6 weeks, GLP-1–treated
`subjects experienced a significant weight
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`1915
`
`MPI EXHIBIT 1085 PAGE 1
`
`

`

`Effects of liraglutide in type 2 diabetes
`
`loss of 1.9 kg compared with baseline; the
`weight loss was, however, not significant
`when compared with placebo (15).
`It is estimated that ⬎80% of type 2
`diabetic patients are obese, and even more
`might be overweight, with abdominal fat
`accumulation, and hence benefit from
`weight loss. It would be desirable if pe-
`ripherally administered GLP-1 was able
`to reduce energy intake and thereby in-
`duce weight loss. A major disadvantage is
`that native GLP-1 has a very short half-life
`in plasma due to rapid degradation by
`⬍1.5 min af-
`dipeptidyl-peptidase IV (t0.5
`ter intravenous injection) (16), which is
`why modifications are needed for clinical
`use. Liraglutide (NN2211) is a long-
`acting GLP-1 derivative developed for the
`treatment of type 2 diabetes. Liraglutide
`⫽ 13 h) suit-
`has a prolonged action (t0.5
`able for once-daily injection. The mecha-
`nism of protraction is a combination of
`albumin binding and self-association, re-
`sulting in slow absorption from subcutis,
`stability against dipeptidyl-peptidase IV,
`and a long plasma half-life. The aim of the
`present study was to examine if liraglutide
`administered subcutaneously once daily
`for 8 weeks in obese type 2 diabetic sub-
`jects would decrease body weight and to-
`tal fat mass and have an effect on 24-h
`energy expenditure, in addition to im-
`provement of glycemic control.
`
`RESEARCH DESIGN AND
`METHODS — Thirty-five type 2 dia-
`betic patients (age 60.0 ⫾ 9.5 years
`[mean ⫾ SD]) were recruited from the
`investigators’ own patient files and/or ad-
`vertisement in local newspapers and
`screened medically for their eligibility for
`the trial. Inclusion criteria were 1) diet
`treated and/or subjects in monotherapy
`with sulfonylurea or repaglinide (No-
`voNorm); 2) HbA1c for diet-treated sub-
`jects of 7–12%, both inclusive; 3) HbA1c
`for sulfonylurea-treated subjects ⱕ10%;
`and 4) BMIⱖ27 kg/m 2. The key exclu-
`sion criteria were 1) New York Heart As-
`sociation class III and IV heart failure, 2)
`uncontrolled hypertension (systolic
`blood pressure ⱖ180 mmHg and/or dia-
`stolic blood pressure ⱖ105 mmHg), 3)
`serum creatinine ⬎150 ␮mol/l, and 4)
`alanine aminotransferase three or more
`times the upper-normal range. Twenty
`patients were treated with sulfonylureas,
`3 with NovoNorm, and 12 with diet only.
`The protocol (file no. 02-024/01) was ap-
`proved by the Ethical Committee of Fre-
`
`deriksberg and Copenhagen. The trial
`was performed in accordance with the
`Helsinki II Declaration. Written consent
`was obtained from each participant be-
`fore beginning the study.
`The study was double blind and pla-
`cebo controlled with two parallel arms.
`The subjects attended a screening visit to
`assess their eligibility. Eligible subjects on
`oral hypoglycemic agents (OHAs) were
`asked to discontinue current OHA treat-
`ment for a 2-week washout period before
`the start of dosing. Eligible subjects were
`randomized to receive a single daily dos-
`ing of liraglutide (0.6 mg) or placebo for 8
`weeks (long term). The trial product was
`administered as a subcutaneous injection
`in the abdomen or in the thigh in the
`morning using a NovoPen (1.5 with No-
`vofine 30-gauge 0.3- to 8-mm needle) as
`the dispensing device. The 24-h energy
`expenditure was measured in a respira-
`tory chamber before the intervention pe-
`riod (baseline) and again after 3 days on
`liraglutide or placebo. The 24-h energy
`expenditure was performed after 3 days
`(short term) only to avoid confounding
`due to possible weight loss later in the
`intervention period. Thus, no long-term
`energy expenditure assessment was done.
`To assess appetite and spontaneous en-
`ergy intake, three meal tests were con-
`ducted through the intervention period.
`Two were carried out following each re-
`spiratory chamber visit, and the third one
`was conducted at the end of the interven-
`tion period (8 weeks). Subjective appetite
`ratings were assessed by visual analog
`scores (VASs) in connection with the
`fixed-energy breakfast meal (17). Sponta-
`neous energy intake was assessed in con-
`nection to an ad libitum lunch meal 4 h
`after the fixed-energy breakfast meal.
`Body composition (assessed by dual-
`energy X-ray absorptiometry [DEXA]
`scan) was evaluated at baseline and after 8
`weeks (end of intervention).
`
`Analytical determinations
`HbA1c was analyzed using a high-
`performance liquid chromatography,
`ion-exchange chromatography assay
`(normal range 4.3–5.8%; Tosoh). Serum
`concentrations of liraglutide, insulin, and
`C-peptide were analyzed by enzyme-
`linked immunosorbent assay methods.
`Serum concentration of glucose was mea-
`sured using an enzymatic glucose oxidase
`method (normal range [fasting] 3.9 – 6.6
`mmol/l). Plasma glucagon was analyzed
`
`by MDS Pharma Services, Switzerland,
`using a radioimmunoassay (Linco Re-
`search, St. Charles, MO). Standard labo-
`ratory analyses were performed by a
`central laboratory (if not otherwise stat-
`ed): Capio Diagnostik, Copenhagen,
`Denmark. Homeostasis model assess-
`ment (HOMA) analysis: An estimate of
`␤-cell function (HOMA-S) and of insulin
`resistance (HOMA-R) was calculated by
`HOMA: ␤-cell function (%) ⫽ 20 ⫻ in-
`sulin/(glucose ⫺ 3.5) and insulin resis-
`tance ⫽ insulin/(22.5 ⫻ elnglucose) (18).
`
`Meal test
`A venflon catheter was inserted in an an-
`tecubital arm vein. After a 10-min rest,
`fasting blood samples were taken. Imme-
`diately thereafter the subjects took a sin-
`gle dose of either liraglutide or placebo,
`then the standard breakfast meal consist-
`ing of 2.5 MJ (carbohydrate 50 E%, pro-
`tein 13 E%, and fat 37 E%) was served
`and consumed within 30 min. Paraceta-
`mol (15 mg/kg) dissolved in 150 ml of
`water was administered to the subjects at
`initiation of the breakfast meal for assess-
`ment of the gastric emptying rate. Blood
`samples were collected regularly over the
`240 min after starting the meal for analy-
`sis of plasma paracetamol, glucose, insu-
`lin, glucagon, C-peptide, and lipid
`profile. Ratings for appetite were made on
`100-mm VASs, with the text expressing
`the most positive and the most negative
`rating anchored at each end. VAS was
`used to assess satiety, hunger, fullness,
`prospective food consumption, and
`thirst. The ratings were recorded immedi-
`ately before the meal and throughout a
`4-h period after the meal.
`An ad libitum lunch meal consisting
`of carbohydrate 49.5 E%, protein 13.3
`E%, and fat 37.2 E% was served ⬃4.5 h
`after completion of the breakfast meal.
`The lunch meal should be completed
`within 30 min. To reduce participants’
`awareness of the amount of food being
`consumed, food was served in excess. The
`quantity of food and fluid consumed was
`measured, and the total energy intake was
`calculated.
`
`DEXA scan
`Body composition was assessed before
`treatment by DEXA scan in all subjects
`and again after the 8-week intervention
`period. A dual-energy X-ray absorptiom-
`eter (Lunar Radiation, Madison, WI) was
`used. All scans were performed in the
`
`1916
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`MPI EXHIBIT 1085 PAGE 2
`
`

`

`Table 1—Baseline characteristics of patients
`
`n
`Demographics
`Men/women
`Age (years)
`Anthropometrics
`BMI (kg/m2)
`Diabetes
`Duration (years)
`HbA1c (%)
`Fasting plasma glucose (mmol/l)
`Previous diabetes treatment
`Diet
`OHA
`Data are means ⫾ SD or n (%).
`
`Liraglutide
`
`21
`
`11/10
`59.9 ⫾ 11.0
`
`36.8 ⫾ 4.6
`
`4.5 ⫾ 6.4
`7.4 ⫾ 1.0
`10.7 ⫾ 2.4
`
`4 (19)
`17 (81)
`
`Placebo
`
`12
`
`1/11
`60.1 ⫾ 6.7
`
`36.1 ⫾ 3.4
`
`3.3 ⫾ 3.4
`7.7 ⫾ 1.6
`10.4 ⫾ 2.8
`
`7 (58)
`5 (42)
`
`slow mode on fasting subjects. The fol-
`lowing parameters were assessed: lean tis-
`sue mass, fat mass, and total body weight.
`
`Respiratory chamber
`The 24-h energy expenditure was mea-
`sured in an open-circuit indirect calorim-
`etry respiratory chamber as described
`elsewhere (19). The measurement started
`at 9:00 A.M. and continued for 24 h. Basic
`metabolic rate was measured during the
`last hour of the stay. Subjects arrived at
`the department at 10:00 P.M. the evening
`before the examination and stayed in the
`chamber overnight. This procedure was
`chosen for the subject to adapt to the
`chamber, thereby minimizing stress dur-
`ing the 24-h measurement period. During
`the 24-h stay, subjects followed a stan-
`dard protocol, including two periods of
`10 min of bicycle exercise (75 watts) and
`two periods of walking back and forth in
`the chamber (with a total distance of
`182.5 m each time).
`
`Safety assessments
`Adverse events, hematology, blood chem-
`istry, and urine values were monitored
`throughout the study period. In addition,
`electrocardiograms and blood pressure
`were measured. Subjects were also sup-
`plied with a diary and asked to record
`information on home measurements of
`blood glucose, any changes in concomi-
`tant medication, concomitant illness, and
`hypoglycemic episodes.
`
`Statistical analysis
`All statistical tests were two sided at a 5%
`significance level. The model used was an
`
`ANOVA with treatment as a fixed factor
`and baseline value as a covariate. The
`main comparison was after 8 weeks of
`treatment (long-term assessment). In ad-
`dition, the end points were assessed after
`2– 4 days of treatment (short-term assess-
`ment) and for the period between the long
`and short terms. The latter was only done
`for the within-group comparison. This
`model was applied to all efficacy end
`points. Safety end points were summa-
`rized by descriptive statistics (number of
`observations, mean, SE, minimum, and
`maximum) or listed.
`
`RESULTS — Thirty-five patients were
`randomized. Thirty-three patients com-
`pleted the intervention, 21 assigned to the
`liraglutide group and 12 to the placebo
`group. One subject was withdrawn from
`the trial due to claustrophobia and one
`subject due to difficulties with blood
`drawing, both of whom withdrew before
`the trial drug was administered. There
`was an unbalanced distribution of sex in
`the two treatment groups. However, this
`uneven distribution has been analyzed
`and no statistical significant effect was
`seen. Clinical characteristics of the en-
`rolled patients are shown in Table 1.
`
`Total body weight and body
`composition
`After 8 weeks, no significant difference in
`total body weight change was observed
`between liraglutide and placebo treat-
`ments (liraglutide, ⫺0.7 kg, and placebo,
`⫺0.9 kg; P ⫽ 0.756) (Fig. 1 and Table 2).
`Although nonsignificant, the results from
`the DEXA scan indicated that fat mass (as
`
`Harder and Associates
`
`a percentage) decreased (liraglutide,
`⫺1.0%, and placebo, ⫺0.1%; P ⫽ 0.088)
`and that lean tissue mass increased after
`treatment with liraglutide compared with
`placebo (liraglutide, 1.0%, and placebo,
`0.2%; P ⫽ 0.118) (Table 2).
`
`Glycemic control and lipids
`Table 2 shows changes from baseline
`within each group and a comparison of
`changes from baseline between groups.
`Liraglutide improved glycemic control,
`and this effect was significant after the
`first week and persisted throughout the
`study period (difference in fasting serum
`glucose between liraglutide and placebo
`after 8 weeks: 2.17 mmol/l [95% CI
`⫺3.50 to ⫺0.83], P ⫽ 0.002). At the end
`of the study, liraglutide showed a signifi-
`cant decrease in HbA1c level compared
`with placebo (⫺ 0.80% [⫺ 1.50 to
`⫺0.09], P ⫽ 0.028). Liraglutide treat-
`ment suggested an improved ␤-cell func-
`tion compared with placebo as indicated
`by increased HOMA-S (liraglutide,
`17.2%, and placebo, ⫺15.2%; P ⫽
`0.015), whereas insulin resistance
`(HOMA-R) was not affected (liraglutide,
`⫺1.9, and placebo, ⫺0.8, P ⫽ 0.530).
`Liraglutide had no effect on lipid param-
`eters compared with placebo (data not
`shown).
`
`Meal tests
`The absolute area under the curve (AUC)
`for serum glucose was significantly sup-
`pressed in the liraglutide group compared
`with placebo both after 3 days (short
`term) (liraglutide, ⫺530.6 mmol 䡠 l⫺1 䡠
`min⫺1, and placebo, ⫺113.9; P ⫽ 0.002)
`and after 8 weeks (long term) of treatment
`(liraglutide, ⫺554.7, and placebo, 11.5;
`P ⫽ 0.004). No difference was detected
`for the incremental AUC for serum glu-
`cose in either the short term (liraglutide,
`0.23 mmol/l, and placebo, ⫺0.20
`mmol/l; P ⫽ 0.441) or the long term (li-
`raglutide, ⫺0.05 mmol/l, and placebo,
`⫺0.84 mmol/l; P ⫽ 0.128). Gastric emp-
`tying was unaffected as measured by
`mean change in postprandial plasma
`paracetamol concentrations after both 3
`days’ and 8 weeks’ treatment. Neither
`short- nor long-term treatment with lira-
`glutide changed the subjective appetite
`sensations during the meal test according
`to the VAS. Food intake during the ad
`libitum lunch meal after 3 days and 8
`weeks of treatment was not affected by
`liraglutide compared with placebo (short
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`1917
`
`MPI EXHIBIT 1085 PAGE 3
`
`

`

`Effects of liraglutide in type 2 diabetes
`
`Figure 1—Mean body weight (A), fasting serum
`glucose (B), and HbA1c (C) over 8 weeks in type 2
`diabetic patients treated with liraglutide 0.6 mg/kg
`versus placebo for 8 weeks.
`
`CONCLUSIONS — This clinical trial
`of 8 weeks’ treatment with subcutaneous
`liraglutide in patients with type 2 diabetes
`shows that 0.65 mg liraglutide, self-
`administered once daily before breakfast
`for 8 weeks, compared with placebo im-
`proves glycemic control by decreasing
`fasting glucose by 2.2 mmol/l and HbA1c
`by 0.8% in this group of relatively well-
`controlled type 2 diabetic patients, with-
`out inducing weight gain. Weight gain
`associated with improved glycemic con-
`trol is common in treatment with sulfo-
`nylureas, insulin, or thiazolidinediones
`and often occurs when HbA1c levels are
`significantly reduced. This effect has been
`reported in the intensively treated cohorts
`of the Diabetes Control and Complica-
`tions Trial (20) and the U.K. Prospective
`Diabetes Study (21). Weight gain is an
`undesirable effect as it is often associated
`with increased insulin resistance, espe-
`cially if the additional fat is deposited in
`the abdominal region (22). In the present
`study, we failed to see any weight loss
`produced by liraglutide, and in accor-
`dance with this observation there were no
`detectable effects on appetite sensation,
`gastric emptying, or energy intake at the
`ad libitum test meals in connection with
`administration of liraglutide. However,
`there was a trend to a reduction in fat
`mass, which results from a daily negative
`energy balance of a magnitude that would
`not be picked up by the meal tests or the
`appetite sensations. The trend in reduc-
`tion of fat mass suggests that liraglutide
`actually may have the potential to induce
`weight loss, and we speculate that larger
`doses and a longer study period might be
`required to produce an effect on appetite
`and weight loss (23). Further studies are
`needed to fully elucidate the weight pro-
`file of liraglutide. Preclinical studies in
`nondiabetic animals have shown a poten-
`tial for lowering food intake and body
`weight with liraglutide. In one study, a
`dose of 0.2 mg/kg has been shown to
`markedly decrease food intake and body
`weight in normal-weight and obese rats
`(24). Doses of liraglutide needed in rats
`are larger than in humans, also for blood
`glucose lowering (25).
`In addition, treatment with lira-
`
`term: liraglutide, 5 g, and placebo, ⫺1 g,
`P ⫽ 0.883; long term: liraglutide, 21 g,
`and placebo, 7 g, P ⫽ 0.766).
`
`treatment did not differ significantly be-
`tween the groups (P ⫽ 0.548).
`
`Twenty-four-hour energy
`expenditure
`Liraglutide had no short-term effect on
`energy balance as the mean change in
`24-h energy expenditure from before to
`after 3 days’ treatment did not differ sig-
`nificantly between the groups (lira-
`glutide, ⫺12.6 kJ/h, and placebo, ⫺13.7
`kJ/h; P ⫽ 0.799). The substrate oxidation
`was not affected by treatment with lira-
`glutide as the change in 24-h nonprotein
`respiratory quotient from before to after
`
`Safety assessments
`Eight weeks’ treatment with the subcuta-
`neous injection of liraglutide (single daily
`dose of 0.65 mg) was well tolerated. No
`hypoglycemic episodes occurred during
`the study. In the liraglutide group, 76%
`(16 of 21) of the subjects reported adverse
`events, whereas 58% (7 of 12) of the pla-
`cebo-treated subjects reported adverse
`events. Nausea and diarrhea were the
`most frequent events; however, these
`were transient episodes.
`
`1918
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`MPI EXHIBIT 1085 PAGE 4
`
`

`

`Table 2—Body weight, body composition, and glycemic control parameters before and after 8 weeks’ intervention
`
`Variable
`
`Liraglutide
`
`Placebo
`
`P
`
`Harder and Associates
`
`n
`Weight (kg)
`Before
`After
`Change
`Waist circumference (cm)
`Before
`After
`Change
`Fat mass (kg)
`Before
`After
`Change
`Lean mass (kg)
`Before
`After
`Change
`Fat mass (%)
`Before
`After
`Change
`Lean mass (%)
`Before
`After
`Change
`Fasting serum glucose (mmol/l)
`Before
`After
`Change
`HbA1c (%)
`Before
`After
`Change
`Fructosamine (␮mol/l)
`Before
`After
`Change
`Insulin (pmol/l)
`Before
`After
`Change
`C-peptide (nmol/l)
`1.64 ⫾ 0.39
`1.48 ⫾ 0.13
`Before
`1.53 ⫾ 0.35
`1.52 ⫾ 0.13
`After
`⫺0.09 (⫺0.41 to 0.23)
`0.02 (⫺0.22 to 0.26)
`Change
`Data are means ⫾ SE or mean change values (95% CI). All change values are adjusted for differences in baseline values between groups.
`
`21
`
`106.9 ⫾ 2.9
`104.8 ⫾ 3.0
`⫺0.7 (⫺1.6 to 0.3)
`
`114.6 ⫾ 2.0
`114.0 ⫾ 2.0
`⫺0.5 (⫺1.6 to 0.6)
`
`41.4 ⫾ 2.3
`39.8 ⫾ 2.1
`⫺1.0 (⫺1.8 to ⫺0.3)
`
`59.6 ⫾ 2.5
`60.2 ⫾ 2.6
`0.7 (⫺0.1 to 1.6)
`
`39.5 ⫾ 1.8
`38.5 ⫾ 1.7
`⫺1.0 (⫺1.6 to ⫺0.4)
`
`57.0 ⫾ 1.8
`58.1 ⫾ 1.6
`1.0 (0.5 to 1.6)
`
`10.7 ⫾ 0.5
`8.8 ⫾ 0.5
`⫺1.9 (⫺2.7 to ⫺1.1)
`
`7.37 ⫾ 0.21
`7.07 ⫾ 0.24
`⫺0.33 (⫺0.75 to 0.09)
`
`298.5 ⫾ 10.9
`272.8 ⫾ 9.0
`⫺25.0 (⫺41.2 to ⫺8.9)
`
`124.8 ⫾ 14.2
`118.7 ⫾ 16.7
`⫺7.8 (⫺38.1 to 22.5)
`
`12
`
`98.0 ⫾ 3.8
`96.0 ⫾ 4.0
`⫺0.9 (⫺2.2 to 0.4)
`
`108.6 ⫾ 2.0
`107.8 ⫾ 2.4
`⫺1.1 (⫺2.6 to 0.4)
`
`39.3 ⫾ 1.9
`38.6 ⫾ 2.0
`⫺0.7 (⫺1.7 to 0.4)
`
`49.7 ⫾ 1.9
`49.5 ⫾ 2.1
`0.2 (⫺1.4 to 1.1)
`
`42.7 ⫾ 1.2
`42.4 ⫾ 1.0
`⫺0.1 (⫺0.9 to 0.7)
`
`54.1 ⫾ 1.2
`54.5 ⫾ 1.1
`0.2 (⫺0.6 to 1.0)
`
`10.4 ⫾ 0.8
`10.7 ⫾ 1.0
`0.3 (⫺0.8 to 1.3)
`
`7.68 ⫾ 0.47
`8.10 ⫾ 0.47
`0.47 (⫺0.09 to 1.03)
`
`293.8 ⫾ 18.6
`313.4 ⫾ 16.1
`18.3 (⫺3.1 to 39.6)
`
`147.2 ⫾ 53.4
`122.8 ⫾ 48.7
`⫺21.5 (⫺61.6 to 18.5)
`
`0.756
`
`0.505
`
`0.590
`
`0.259
`
`0.088
`
`0.118
`
`0.002
`
`0.028
`
`0.003
`
`0.581
`
`0.592
`
`glutide did not result in any hypoglyce-
`mic episodes. Therapy with sulfonylureas
`and insulin is associated with the risk of
`hypoglycemia (26), whereas the insulino-
`tropic action of GLP-1 is attenuated as
`ambient glucose levels fall. This glucose-
`dependent mode of action is mediated by
`intracellular signaling mechanisms de-
`
`pendent on a high ATP-to-ADP ratio,
`which is generated by the glucose-
`glycolysis signaling pathway (27,28).
`Thus, treatment with the long-acting
`GLP-1 derivative potentially conveys a
`low risk of drug-related hypoglycemia.
`This glucose-dependent mechanism has
`been confirmed for liraglutide in a glu-
`
`cose-ramp study (29), where liraglutide
`only enhanced insulin secretory response
`under graded glucose infusion when
`plasma glucose was ⬎6 mmol/l. In the
`present study, the placebo-corrected fast-
`ing plasma glucose was reduced by 2.2
`mmol/l to an average of 8.8 mmol/l, and a
`corresponding signifi cant placebo-
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`1919
`
`MPI EXHIBIT 1085 PAGE 5
`
`

`

`Effects of liraglutide in type 2 diabetes
`
`corrected reduction in HbA1c of 0.8% was
`seen. In the liraglutide group, a relatively
`modest decrease of 0.33% in HbA1c from
`baseline was detected, which is most
`likely due to one or more of the following
`circumstances. First, the effect on HbA1c
`is most likely underestimated because af-
`ter only 8 weeks’ treatment steady-state
`levels on HbA1c are not achieved, together
`with the short prerandomization washout
`period. Second, the subjects in the lira-
`glutide group were relatively well con-
`trolled at baseline, with an HbA1c of
`7.4%, limiting the potential for observing
`further substantial absolute reductions in
`HbA1c. Third, the preponderance of
`OHA-treated subjects in the liraglutide
`group might also have led to an underes-
`timation of the effect of liraglutide on gly-
`cemic control; compared with oral
`antidiabetic drug–treated patients who
`have their treatment substituted, diet-
`treated patients would be expected to im-
`prove their glycemic control more when
`commencing drug treatment. Finally, the
`above-mentioned circumstances, to-
`gether with the potentially suboptimal
`dose of liraglutide, might have led to an
`underestimation of liraglutide’s true effi-
`cacy on glycemic control in the present
`trial.
`In addition to the overall improve-
`ment in glycemic control noted with lira-
`glutide in both sulfonylurea-treated and
`oral hypoglycemic agent–naive patients,
`liraglutide lowered the absolute AUC for
`glucose significantly in the postprandial
`period, which was maintained over the 8
`weeks. This is in accordance with previ-
`ous long-term studies (15,30), which
`have shown that administration of subcu-
`taneous injections of native GLP-1 before
`meals through periods of up to several
`weeks can reduce the mean blood glucose
`levels in patients with type 2 diabetes with
`suboptimal blood glucose control. How-
`ever, the incremental AUC for glucose
`was unaffected by liraglutide treatment
`compared with placebo. This is not in
`agreement with another liraglutide study,
`in which a single injection of liraglutide
`(10 ␮g/kg) was administered at 11:00 P.M.
`and a standardized mixed meal served at
`11:30 A.M. the next day. In this study,
`both absolute and incremental AUCs for
`glucose were markedly and significantly
`reduced by 23 and 27%, respectively, by
`liraglutide treatment (31). However, the
`dose was higher than in the present study,
`which might explain this discrepancy.
`
`In the present study, the liraglutide
`did not increase the postprandial insulin
`response significantly (data not shown)
`and did not improve insulin sensitivity as
`indicated by unchanged HOMA-R, but
`suppressed the postprandial glucagon re-
`sponse. ␤-Cell function seemed to im-
`prove after liraglutide treatment because
`HOMA-S increased significantly com-
`pared with placebo. Early and progress-
`ing decline in ␤-cell function is a key
`feature of type 2 diabetes and will even-
`tually lead to insulin deficiency. A study
`(15) with native GLP-1 found similar ef-
`fects on ␤-cell function after 6 weeks’
`treatment, which suggests that GLP-1 has
`a sustained ␤-cell–improving effect.
`This trial clearly shows that 8 weeks’
`treatment with the long-acting GLP-1 de-
`rivative liraglutide improves glycemic
`control in subjects with type 2 diabetes.
`Despite significant improved glycemic
`control, body weight was maintained
`during treatment with liraglutide. Appe-
`tite and food intake were unaffected, but a
`tendency toward a favorable change in
`body composition was observed. There-
`fore, we speculate that a higher dose
`might produce significant loss of fat mass.
`Treatment with liraglutide had no ther-
`mogenic effect, as 24-h energy expendi-
`ture was unaffected after 3 days’ treatment
`(short term). Adverse events were mainly
`mild and related to the gastrointestinal
`system. No episodes of hypoglycemia
`were observed.
`
`Acknowledgments — This study was finan-
`cially supported by Novo Nordisk.
`
`References
`1. Nauck MA, Holst JJ, Willms B, Schmiegel
`W: Glucagon-like peptide 1 (GLP-1) as a
`new therapeutic approach for type 2-dia-
`betes. Exp Clin Endocrinol Diabetes 105:
`187–195, 1997
`2. Holst JJ: Glucagonlike peptide 1: a newly
`discovered
`gastrointestinal
`hormone.
`Gastroenterology 107:1848 –1855, 1994
`3. Kreymann B, Williams G, Ghatei MA,
`Bloom SR: Glucagon-like peptide-1 7-36:
`a physiological incretin in man. Lancet
`2:1300 –1304, 1987
`4. Unger RH, Eisentraut AM: Entero-insular
`axis. Arch Intern Med 123:261–266, 1969
`5. Larsson H, Holst JJ, Ahren B: Glucagon-
`like peptide-1 reduces hepatic glucose
`production indirectly through insulin and
`glucagon in humans. Acta Physiol Scand
`160:413– 422, 1997
`
`6. Flint A, Raben A, Astrup A, Holst JJ: Glu-
`cagon-like peptide 1 promotes satiety and
`suppresses energy intake in humans.
`J Clin Invest 101:515–520, 1998
`7. Drucker DJ: Glucagon-like peptides. Dia-
`betes 47:159 –169, 1998
`8. Naslund E, Barkeling B, King N, Gutniak
`M, Blundell JE, Holst JJ, Rossner S, Hell-
`strom PM: Energy intake and appetite are
`suppressed by glucagon-like peptide-1
`(GLP-1) in obese men. Int J Obes Relat
`Metab Disord 23:304 –311, 1999
`9. Naslund E, Gutniak M, Skogar S, Rossner
`S, Hellstrom PM: Glucagon-like peptide 1
`increases the period of postprandial sati-
`ety and slows gastric emptying in obese
`men. Am J Clin Nutr 68:525–530, 1998
`10. Ranganath LR, Beety JM, Morgan LM,
`Wright JW, Howland R, Marks V: Atten-
`uated GLP-1 secretion in obesity: cause or
`consequence? Gut 38:916 –919, 1996
`11. Naslund E, Gryback P, Backman L, Jacob-
`sson H, Holst JJ, Theodorsson E, Hell-
`strom PM: Distal small bowel hormones:
`correlation with fasting antroduodenal
`motility and gastric emptying. Dig Dis Sci
`43:945–952, 1998
`12. Gutzwiller JP, Drewe J, Goke B, Schmidt
`H, Rohrer B, Lareida J, Beglinger C: Glu-
`cagon-like peptide-1 promotes satiety
`and reduces food intake in patients with
`diabetes mellitus type 2. Am J Physiol 276:
`R1541–R1544, 1999
`13. Toft-Nielsen MB, Madsbad S, Holst JJ:
`Continuous subcutaneous infusion of
`glucagon-like peptide 1 lowers plasma
`glucose and reduces appetite in type 2 di-
`abetic patients. Diabetes Care 22:1137–
`1143, 1999
`14. Nauck MA, Sauerwald A, Ritzel R, Holst
`JJ, Schmiegel W: Influence of glucagon-
`like peptide 1 on fasting glycemia in type
`2 diabetic patients treated with insulin af-
`ter sulfonylurea secondary failure. Diabe-
`tes Care 21:1925–1931, 1998
`15. Zander M, Madsbad S, Madsen JL, Holst
`JJ: Effect of 6-week course of glucagon-
`like peptide 1 on glycaemic control, insu-
`lin sensitivity, and beta-cell function in
`type 2 diabetes: a parallel-group study.
`Lancet 359:824 – 830, 2002
`16. Knudsen LB, Agerso H, Bjenning S, Bre-
`genholt S, Carr RD, Godtfredsen C, Holst
`JJ, Huusfeldt PO, Larsen MO, Larsen PJ,
`Nielsen PF, Ribel U, Rolin B, Romer J,
`Sturis J, Wilken M, Kristensen P: GLP-1
`derivatives as novel compounds for the
`treatment of type 2 diabetes: selection of
`NN2211 for clinical development. Drugs
`of the Future 26:677– 685, 2001
`17. Flint A, Raben A, Blundell JE, Astrup A:
`Reproducibility, power and validity of vi-
`sual analogue scales in assessment of ap-
`petite sensations in single test meal
`studies. Int J Obes Relat Metab Disord 24:
`38 – 48, 2000
`
`1920
`
`DIABETES CARE, VOLUME 27, NUMBER 8, AUGUST 2004
`
`MPI EXHIBIT 1085 PAGE 6
`
`

`

`Harder and Associates
`
`18. Matthews DR, Hosker JP, Rudenski AS,
`Naylor BA, Treacher DF, Turner RC: In-
`sulin resistance and beta-cell
`function
`from fasting plasma glucose and insulin
`concentrations in man. Diabetologia 28:
`412– 419, 1985
`19. Toubro S, Sorensen TI, Ronn B, Chris-
`tensen NJ, Astrup A: Twenty-four-hour
`energy expenditure: the role of body com-
`position, thyroid status, sympathetic ac-
`tivity, and family membership.
`J Clin
`Endocrinol Metab 81:2670 –2674, 1996
`20. Diabetes Control and Complications Trial
`Research Group: The effect of intensive
`treatment of diabetes on the development
`and progression of long-term complica-
`tions in insulin-dependent diabetes mel-
`litus. N Engl J Med 329:977–986, 1993
`21. United Kingdom Prospective Diabetes
`Study Group: United Kingdom Pros-
`pective Diabetes Study 24: a 6-year,
`randomized, controlled trial comparing
`sulfonylurea,
`insulin, and metformin
`therapy in patients with newly diagnosed
`type 2 diabetes that coul

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