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
`
`Improved Glycemic Control With No
`Weight Increase in Patients With Type 2
`Diabetes After Once-Daily Treatment
`With the Long-Acting Glucagon-Like
`Peptide 1 Analog Liraglutide (NN2211)
`A 12-week, double-blind, randomized, controlled trial
`
`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`CONCLUSIONS — A once-daily dose of
`liraglutide provides efficacious glycemic con-
`trol and is not associated with weight gain.
`Adverse events with the drug are mild and
`transient, and the risk of hypoglycemia is
`negligible.
`
`Diabetes Care 27:1335–1342, 2004
`
`T ype 2 diabetes is characterized by
`
`insulin resistance and defective
`␤-cell function and is associated
`with hyperglucagonemia, increased he-
`patic glucose production, and obesity (1).
`In addition, patients with type 2 diabetes
`experience a subnormal secretion of the
`incretin hormone glucagon-like peptide 1
`(GLP-1) during meals (2,3). Sulfonyl-
`ureas, although efficient in stimulating
`insulin secretion and lowering blood glu-
`cose, pose the disadvantages of weight
`gain and risk of hypoglycemia (4). Studies
`have demonstrated that GLP-1 stimulates
`insulin secretion, inhibits glucagon secre-
`tion in a glucose-dependent manner, and
`delays gastric emptying (2,5–7). In addi-
`tion, several studies have shown GLP-1 to
`have an appetite-reducing effect (8 –10),
`and one study has demonstrated weight
`loss after treatment with GLP-1 (11).
`These mechanisms make this hormone an
`attractive candidate for the treatment of
`type 2 diabetes. However, native GLP-1
`has a very short half-life (1 min) (12), be-
`ing rapidly metabolized by the enzyme
`dipeptidyl peptidase IV (13). It has been
`shown that GLP-1 must be present con-
`tinuously in the blood stream to exert its
`actions (14).
`Liraglutide is a long-acting, acylated
`GLP-1 analog, acting as a full agonist to-
`ward the GLP-1 receptor (15). Studies in
`animals and humans have demonstrated
`promising blood glucose⫺lowering ef-
`
`1
`
`STEN MADSBAD, MD, DMSC
`2
`OLE SCHMITZ, MD, DMSC
`3
`JONAS RANSTAM, PHD
`GRETHE JAKOBSEN, MSCPHARM
`
`3
`
`4
`DAVID R. MATTHEWS, DPHIL, FRCP
`ON BEHALF OF THE NN2211-1310
`INTERNATIONAL STUDY GROUP
`
`OBJECTIVE — Liraglutide is a long-acting glucagon-like peptide 1 analog designed for once
`daily injection. This study assessed the efficacy and safety of liraglutide after 12 weeks of
`treatment in type 2 diabetic patients.
`
`RESEARCH DESIGN AND METHODS — A double-blind, randomized, parallel-
`group, placebo-controlled trial with an open-label comparator arm was conducted among 193
`outpatients with type 2 diabetes. The mean age was 56.6 years and the mean HbA1c was 7.6%
`across the treatment groups. Patients were randomly assigned to one of five fixed-dosage groups
`of liraglutide (0.045, 0.225, 0.45, 0.60, or 0.75 mg), placebo, or open-label sulfonylurea
`(glimepiride, 1– 4 mg). The primary end point was HbA1c after 12 weeks; secondary end points
`were fasting serum glucose, fasting C-peptide, fasting glucagon, fasting insulin, ␤-cell function,
`body weight, adverse events, and hypoglycemic episodes.
`
`RESULTS — A total of 190 patients were included in the intention-to-treat (ITT) analysis.
`HbA1c decreased in all but the lowest liraglutide dosage group. In the 0.75-mg liraglutide group,
`HbA1c decreased by 0.75 percentage points (P ⬍ 0.0001) and fasting glucose decreased by 1.8
`mmol/l (P ⫽ 0.0003) compared with placebo. Improvement in glycemic control was evident
`after 1 week. Body weight decreased by 1.2 kg in the 0.45-mg liraglutide group (P ⫽ 0.0184)
`compared with placebo. The proinsulin-to-insulin ratio decreased in the 0.75-mg liraglutide
`group (⫺0.18; P ⫽ 0.0244) compared with placebo. Patients treated with glimepiride had
`decreased HbA1c and fasting glucose, but slightly increased body weight. No safety issues were
`raised for liraglutide; observed adverse events were mild and transient.
`
`● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ●
`
`From the 1Hvidovre Hospital, University of Copenhagen, Denmark; the 2University Hospital of Aarhus,
`Aarhus, Denmark; 3Novo Nordisk A/S, Bagsvaerd, Denmark; and the 4Oxford Centre for Diabetes, Endo-
`crinology & Metabolism, University of Oxford, Oxford, U.K.
`Address correspondence and reprint requests to Dr. Sten Madsbad, Department of Endocrinology,
`Hvidovre Hospital, Kettegaards Alle´ 30, DK-2650, Hvidovre, Denmark. E-mail: sten.madsbad@hh.hosp.dk.
`Received for publication 21 October 2003 and accepted in revised form 7 March 2004.
`D.R.M. has received consulting fees from Novo Nordisk, and the Oxford Centre for Diabetes, Endocri-
`nology & Metabolism is supported by Novo Nordisk in an academic partnership.
`Abbreviations: GLP-1, glucagon-like peptide 1; HOMA, homeostasis model assessment; ITT, intention
`to treat; OHA, oral hypoglycemic agent.
`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.
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`1335
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00001
`
`

`

`Improved glycemic control with liraglutide (NN2211)
`
`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`fects as well as a favorable safety profile.
`The half-life of liraglutide is ⬃12 h in
`both healthy subjects and type 2 diabetic
`patients after single and multiple dosing
`(16 –18). The dosing regimen is a once
`daily injection. This trial investigated the
`efficacy and safety of liraglutide after 12
`weeks of treatment in type 2 diabetic pa-
`tients.
`
`RESEARCH DESIGN AND
`METHODS —This trial was a 12-week,
`multicenter, double-blind, randomized,
`parallel-group, dosage-response trial in
`six treatment arms. In addition, an open-
`label sulfonylurea (glimepiride) arm was
`included as a reference group. Patients
`were instructed to maintain their diet, ex-
`ercise program, and daily routines during
`the course of the trial. After a 4-week
`wash-out period during which current
`oral hypoglycemic agent (OHA) treat-
`ment was discontinued, patients were
`equally randomized to double-blind
`treatment of one of six arms (one of five
`dosages of liraglutide or placebo) or to the
`open-label reference group (glimepiride).
`The trial was conducted between Decem-
`ber 2000 and October 2001 in Scandina-
`via and the U.K. The trial was conducted
`in accordance with the Helsinki Declara-
`tion (19), and the protocol was approved
`by each institution’s independent ethics
`committee. The trial was explained to all
`patients, and their written informed con-
`sent was obtained before any trial-related
`procedures were initiated.
`Eligible patients were men and
`women age ⱖ30 years who had a type 2
`diabetes diagnosis (according to Ameri-
`can Diabetes Association criteria) (20),
`had BMI ⱕ40 kg/m2, were being treated
`ⱕ
`with diet or an OHA, and had an HbA1c
`9.5% (OHA) or 7.5–10.0% (diet). For pa-
`tient safety, an upper limit for HbA1c was
`defined as a 4-week wash-out period and
`a placebo arm was included in the trial.
`Patients were excluded if any of the fol-
`lowing were present: liver or renal dis-
`ease, heart failure (New York Heart
`Association class III and IV) (21), unsta-
`ble angina pectoris, myocardial infarction
`within the previous 12 months, concom-
`itant treatment with thiazolidinediones or
`other investigational drugs, or other sig-
`nificant conditions likely to affect a pa-
`tient’s diabetes and/or ability to complete
`the trial. Women who were pregnant,
`breast-feeding, or not using an adequate
`method of contraception were also ex-
`
`cluded. At randomization, fasting blood
`glucose had to be 6 –13 mmol/l. Patients
`were withdrawn from the trial if fasting
`plasma glucose was ⬎15 mmol/l.
`In total, 311 patients were screened
`and 193 patients were randomized to a
`treatment group (Table 1). The reasons
`for failing screening were not meeting any
`inclusion criteria or meeting any exclu-
`sion criteria (n ⫽ 86), not meeting ran-
`domization criteria (n ⫽ 7), withdrawn
`consent (n ⫽ 6), and other (n ⫽ 19). The
`majority of patients (n ⫽ 158) were being
`treated with OHA(s) and the remaining
`35 patients were being treated with diet
`only. Most patients were on monotherapy
`with either metformin (65 patients) or
`sulfonylureas (55 patients); 22 patients
`received a combination of metformin and
`sulfonylureas, 14 patients received repa-
`glinide, and 2 patients received acarbose
`treatment. In all, 190 patients were ex-
`posed to the experimental protocol; 3 pa-
`tients withdrew consent before receiving
`randomized treatment.
`Patients were recruited from the par-
`ticipating investigators’ outpatient clinics
`or by local advertisements. Each patient
`was seen on seven occasions: screening;
`baseline; after 1, 4, 8, and 12 weeks of
`treatment; and follow-up. Current treat-
`ment with oral antidiabetic medication
`was discontinued at screening. Patients
`were supplied with a blood glucose meter
`(One Touch Profile Glucometer) and in-
`structed in its use. Fasting blood glucose
`was measured every morning. HbA1c,
`fasting serum glucose, insulin, C-peptide,
`and glucagon were measured every 4
`weeks. Fasting serum glucose was also
`measured after the first week of treatment.
`Proinsulin was assessed in a subset of pa-
`tients (n ⫽ 74, equally distributed among
`the groups). Fasting samples were ob-
`tained before administration of the trial
`drug. Safety parameters (adverse events,
`hypoglycemic episodes, weight, standard
`hematology and biochemistry profile, vi-
`tal signs, and electrocardiogram) were
`assessed at each visit. Liraglutide anti-
`bodies were measured before and after
`treatment.
`The trial was double blind for the five
`dosage levels of liraglutide and placebo,
`and open label for glimepiride. The blind-
`ing was kept until database release.
`Liraglutide and placebo (Novo Nor-
`disk A/S, Bagsvaerd, Denmark) were ad-
`ministered as a once-daily injection
`(subcutaneously) in the morning before
`
`breakfast. The five dosages of liraglutide
`administered were 0.045, 0.225, 0.45,
`0.60, and 0.75 mg. These dosages were
`chosen based on observations from previ-
`ous trials (16 –18). In one of those trials, a
`single dose of 10 ␮g/kg (corresponding to
`⬃0.80 mg) showed significant effects on
`glycemia, but 2 of 11 patients reported
`nausea (18). Further, once-daily dosing
`for 7 days showed a 40% increase in Cmax
`in the steady state (17). Therefore, to
`avoid causing unacceptable side effects,
`the highest dosage included in this trial
`was 0.75 mg, corresponding to 7.5 ␮g/kg
`(equivalent to 10.5 ␮g/kg in steady state)
`for a person weighing 100 kg. The lowest
`dosage of 0.045 mg was considered too
`low to have any significant effect on gly-
`cemic control. Glimepiride (Amaryl;
`Aventis Pharma, Frankfurt, Germany)
`was supplied as 1- and 2-mg tablets for
`oral use, with the dosage adjusted accord-
`ing to glycemic control during the first 4
`weeks, with the aim to achieve a fasting
`plasma glucose level ⬍7 mmol/l. The
`mean glimepiride dosage during the trial
`was 2.7 mg. Compliance was assessed by
`drug accountability and plasma concen-
`tration measurements.
`
`Analytic methods
`HbA1c was analyzed with a Unimate
`HbA1c assay (Roche Diagnostics; normal
`range 4.5–5.7%). Liraglutide antibodies
`were determined by a radioimmunoassay
`developed by the Department of Immu-
`nochemistry at Novo Nordisk A/S. The
`serum concentrations of insulin, C-
`peptide, and proinsulin were analyzed by
`enzyme-linked immunosorbent assay
`methods. Plasma glucagon was analyzed
`by MDS Pharma Services (Wangen, Swit-
`zerland) using a radioimmunoassay
`(Linco Research, St. Charles, MO). Stan-
`dard laboratory analyses were performed
`by a central laboratory (Novo Medical
`Medi-Lab, Clinical Trials Lab, Copenha-
`gen, Denmark, if not otherwise stated).
`
`Statistical analysis
`Pretrial calculation showed that a two-
`sided test of the highest dosage versus the
`placebo group required 30 patients per
`group to detect a difference in mean
`HbA1c of at least 1% unit with 5% signif-
`icance and 95% power.
`The primary end point HbA1c, as well
`as secondary end points (fasting serum
`glucose, fasting C-peptide, glucagon, in-
`sulin, homeostasis model assessment
`
`1336
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00002
`
`

`

`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`Table 1—Patient disposition and characteristics
`
`Placebo
`
`0.045 mg
`
`0.225 mg
`
`0.45 mg
`
`0.60 mg
`
`0.75 mg
`
`Glimepiride Total
`
`Liraglutide
`
`Madsbad and Associates
`
`—
`—
`—
`
`—
`
`—
`—
`29
`29
`5
`0
`3
`2
`24
`29
`
`—
`—
`—
`
`—
`
`—
`—
`26
`26
`3
`0
`2
`1
`23
`26
`
`—
`—
`—
`
`—
`
`—
`—
`25
`24
`3
`1
`1
`1
`21
`24
`
`—
`—
`—
`
`—
`
`—
`—
`27
`27
`7
`0
`4
`3
`20
`27
`
`—
`—
`—
`
`—
`
`—
`—
`30
`30
`2
`1
`1
`0
`28
`30
`
`—
`—
`—
`
`—
`
`—
`—
`29
`28
`2
`1
`1
`0
`26
`28
`
`—
`—
`—
`
`—
`
`—
`—
`27
`26
`0
`0
`0
`0
`26
`26
`
`311
`118
`86
`
`7
`
`6
`19
`193
`190
`22
`3
`12
`7
`168
`190
`
`Patient disposition
`Screened
`Screening failures
`Nonfulfillment of any
`inclusion/exclusion
`criteria
`Not meeting randomization
`criterion
`Withdrew consent
`Other
`Randomized
`Exposed
`Withdrawals
`Adverse events
`Ineffective therapy
`Other
`Completed
`Included in ITT population
`Baseline characteristics
`Age (years)
`Sex
`Male
`Female
`BMI (kg/m2)
`Previous diabetes treatment
`Diet
`OHA
`Duration of diabetes (years)
`HbA1c (%)
`Fructosamine (mmol/l)
`Fasting serum glucose
`(mmol/l)
`0.9 ⫾ 0.5
`1.0 ⫾ 0.4
`1.1 ⫾ 0.6
`1.1 ⫾ 0.5
`1.0 ⫾ 0.5
`1.0 ⫾ 0.5
`1.1 ⫾ 0.4
`C-peptide (mmol/l)
`72.5 ⫾ 55.3 71.5 ⫾ 42.0 64.1 ⫾ 40.4
`73.8 ⫾ 51.0
`84.7 ⫾ 43.6 74.1 ⫾ 40.1 81.2 ⫾ 73.0
`Insulin (mmol/l)
`99 ⫾ 31.1
`91 ⫾ 23.4
`101 ⫾ 23.6
`110 ⫾ 33.0
`110 ⫾ 37.7
`111 ⫾ 47.6
`131 ⫾ 140.0
`Glucagon (mmol/l)
`0.21 ⫾ 0.11 0.25 ⫾ 0.15 0.28 ⫾ 0.19
`0.30 ⫾ 0.20
`0.23 ⫾ 0.15 0.23 ⫾ 0.16 0.26 ⫾ 0.17
`Proinsulin-to-insulin ratio
`44.06 ⫾ 26.3 34.65 ⫾ 23.1 31.83 ⫾ 22.6 31.95 ⫾ 31.7 34.40 ⫾ 37.3 33.75 ⫾ 21.7 26.78 ⫾ 18.2
`␤-Cell function (%)
`Data are means ⫾ SD. Demographics were obtained at visit 1 (screening) and diabetes characteristics at visit 2 (baseline). Of the 193 patients randomized, 3 were
`never exposed. Preferred terms for adverse event withdrawals include sinusitis and fever (0.225 mg), aggressive reaction (0.60 mg), and abnormal hepatic function
`(0.75 mg). Pro-insulin was measured only in a subset of patients (n ⫽ 74).
`
`57 ⫾ 9.4
`
`53 ⫾ 9.0
`
`58 ⫾ 7.5
`
`57 ⫾ 11.3
`
`57 ⫾ 7.7
`
`58 ⫾ 9.7
`
`57 ⫾ 9.2
`
`20
`9
`30.3 ⫾ 4.2
`
`22
`4
`30.2 ⫾ 5.4
`
`15
`9
`32.0 ⫾ 5.3
`
`18
`9
`30.1 ⫾ 5.0
`
`20
`10
`30.4 ⫾ 4.8
`
`16
`12
`31.9 ⫾ 4.3
`
`16
`10
`30.2 ⫾ 4.6
`
`4
`25
`3.4 ⫾ 2.9
`7.4 ⫾ 1.2
`335 ⫾ 72.5
`9.7 ⫾ 2.9
`
`2
`24
`4.1 ⫾ 3.7
`7.4 ⫾ 0.8
`344 ⫾ 77.9
`10.2 ⫾ 2.2
`
`11
`13
`4.4 ⫾ 4.0
`7.9 ⫾ 0.8
`358 ⫾ 57.7
`10.9 ⫾ 3.6
`
`2
`25
`4.5 ⫾ 4.6
`7.7 ⫾ 1.0
`374 ⫾ 104.9
`11.2 ⫾ 2.8
`
`4
`26
`4.6 ⫾ 4.6
`7.4 ⫾ 1.2
`352 ⫾ 74.2
`10.8 ⫾ 2.8
`
`4
`24
`6.1 ⫾ 7.9
`7.4 ⫾ 0.9
`344 ⫾ 73.7
`9.9 ⫾ 2.3
`
`7
`19
`3.8 ⫾ 3.4
`7.8 ⫾ 0.9
`364 ⫾ 69.5
`10.6 ⫾ 2.4
`
`[HOMA], proinsulin-to-insulin ratio, and
`weight) were analyzed in a mixed-effects
`model with treatment, visit, and center as
`fixed effects and patient as the random
`effect. The interaction term, baseline
`HbA1c by visit, was included in the model
`as a covariate. Adjusted end point levels at
`12-week follow-up were calculated for
`each treatment group by means of this
`model.
`The HOMA(S) (␤-cell function) was
`determined as ␤-cell function (%) ⫽ 20 ⫻
`insulin/(glucose ⫺ 3.5). HOMA(R) (insu-
`lin resistance) was determined as resis-
`tance ⫽ insulin/(22.5e⫺ln[glucose]) (22).
`All analyses were performed for the
`
`intention-to-treat (ITT) population (i.e.,
`all patients who received at least one dose
`of a trial drug). Statistical analyses were
`performed using SAS software (version 8;
`SAS Institute, Cary, NC).
`
`RESULTS
`
`Enrollment
`The 193 patients randomized in this trial
`were evenly distributed across treatment
`groups (Table 1). Baseline clinical charac-
`teristics of the 190 patients exposed to the
`experimental protocol are given in Table
`1. No apparent differences were seen
`
`among the treatment groups with regard
`to baseline characteristics.
`
`Effect on glycemic control
`After 12 weeks of treatment, HbA1c was
`decreased in all groups except the lowest
`liraglutide dosage group (Fig. 1A). Treat-
`ment with the two highest dosages re-
`duced HbA1c significantly more than
`placebo (Table 2). At these dosage levels,
`the effect of liraglutide was comparable
`with that of glimepiride with respect to
`effect on HbA1c. The effect of liraglutide
`increased with duration of treatment. The
`largest decreases in HbA1c levels were ob-
`served at the end of the 12-week treat-
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`1337
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00003
`
`

`

`Improved glycemic control with liraglutide (NN2211)
`
`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`Figure 1—Glycemic control. Mean difference in HbA1c compared with placebo (95% CI) after 12
`weeks of treatment (A), mean HbA1c (%) during the 12-week treatment period (B), mean differ-
`ence in fasting serum glucose compared with placebo (95% CI) after 12 weeks of treatment (C),
`and mean fasting serum glucose (mmol/l) during the 12-week treatment period in the ITT popu-
`lation (D). A and C: *P ⬍ 0.05, **P ⬍ 0.01, ***P ⬍ 0.001 vs. placebo. Glim, glimepiride. B and
`D: F, 0.045 mg liraglutide; f, 0.225 mg liraglutide; Œ, 0.45 mg liraglutide; , 0.60 mg lira-
`glutide; }, 0.75 mg liraglutide; [gray circles], glimepiride; 䡺, placebo.
`
`protocol as blood glucose ⬍2.8 mmol/l)
`and 7 reported symptoms of hypoglyce-
`mia only. These incidences seemed lower
`than in the glimepiride group (n ⫽ 26),
`where four patients experienced minor
`hypoglycemia and five patients reported
`symptoms of hypoglycemia.
`The number of patients with adverse
`events (based on spontaneous adverse
`event reporting) was comparable across
`the liraglutide groups and the placebo
`
`group (60% [81 of 135]) vs. 55% [16 of
`29] of patients, respectively) and was
`lower in the open-label reference group
`(35% [9 of 26] of patients). For gastroin-
`testinal events, the incidence seemed to
`increase with increasing doses of lira-
`glutide; nausea was reported by 1–2 pa-
`tients in each of the lowest dose groups
`and by 5 of 28 patients in the highest dose
`group (in total, reported by 10 of 135 pa-
`tients exposed to liraglutide) compared
`
`ment period for the highest dosages (Fig.
`1B), and it seemed that HbA1c levels were
`still decreasing at the end of the treatment
`period at these dosages. We observed that
`59% of patients completing the trial in the
`two highest liraglutide dosage groups
`ⱕ7% after 12 weeks.
`achieved HbA1c
`As was seen with HbA1c, fasting se-
`rum glucose levels decreased in most
`treatment groups during the trial, with
`the decreases being statistically significant
`for the 0.225-, 0.60-, and 0.75-mg lira-
`glutide dosage groups compared with
`placebo (Table 2). In accordance with the
`observations for HbA1c, the effect of the
`highest dosages of liraglutide was compa-
`rable with that of glimepiride with respect
`to fasting serum glucose (Fig. 1C). In the
`liraglutide groups, maximal effect on fast-
`ing serum glucose was evident after the
`first week of treatment (Fig. 1D).
`
`Effect on body weight
`Treatment with liraglutide did not in-
`crease body weight (Fig. 2). Furthermore,
`for the 0.45-mg liraglutide dosage group
`(Table 2), a statistically significant de-
`crease, compared with placebo, was ob-
`served.
`
`Effect on islet cell function
`HOMA (22) was used to assess ␤-cell
`function and insulin resistance after 12
`weeks of liraglutide treatment. Mean
`␤-cell function (derived from fasting in-
`sulin and glucose) was significantly
`higher in the 0.75-mg liraglutide group
`after 12 weeks than in the placebo group
`(Table 2). The effect obtained with the
`highest liraglutide dosage was similar to
`that observed with glimepiride. For insu-
`lin resistance, no differences were seen
`among the three treatments (liraglutide,
`glimepiride, and placebo; data not
`shown). Further, the proinsulin-to-
`insulin ratio decrease was statistically sig-
`nificantly after treatment with 0.75 mg
`liraglutide compared with placebo (Table
`2). No change in the proinsulin-to-insulin
`ratio was demonstrated after treatment
`with glimepiride. There were no statisti-
`cally significant differences between the
`liraglutide groups and the placebo group
`for fasting insulin, C-peptide, and gluca-
`gon (Table 2).
`
`Safety evaluation
`Of the 135 patients exposed to liraglutide,
`1 (in the 0.60-mg group) experienced mi-
`nor hypoglycemia (defined in the study
`
`1338
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00004
`
`

`

`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`Madsbad and Associates
`
`with 1 of 29 patients in the placebo group.
`Other gastrointestinal events included di-
`arrhea (5 of 135 liraglutide-treated pa-
`tients), vomiting (3 of 135 patients), and
`constipation (3 of 135 patients). None of
`these events were reported in the placebo
`or glimepiride groups, with the exception
`of one patient experiencing vomiting in
`the glimepiride group. Approximately
`two-thirds of the gastrointestinal events
`reported during treatment with lira-
`glutide were resolved within 1–3 days.
`None of the patients withdrew due to gas-
`trointestinal events (Table 1). Overall, the
`most frequent adverse events were head-
`ache and nausea, which were also the
`events most often assessed as related to
`the trial product. Generally, adverse
`events were mild or moderate and com-
`pletely resolved. Only 1 of the 135 pa-
`tients exposed to liraglutide experienced
`two mild injection site reactions, de-
`scribed as an “urticarial reaction at injec-
`tion place.” No antibody formation
`against liraglutide could be detected in
`this trial.
`No safety issues with regard to vital
`signs, electrocardiogram, or laboratory
`analysis were raised by this trial.
`
`CONCLUSIONS — This trial was the
`first to demonstrate a sustained improve-
`ment in glycemic control after long-term
`treatment with the GLP-1 analog lira-
`glutide, administered once daily. After 12
`weeks of treatment with 0.60 or 0.75 mg
`liraglutide, both HbA1c and fasting serum
`glucose levels were significantly lower
`than with placebo. Glycemic control was
`maintained throughout the treatment pe-
`riod and was as effective at the highest
`liraglutide dosages as that provided by the
`open-label reference therapy glimepiride
`
`Figure 1—Continued
`
`Table 2—Repeated measures analysis after 12 weeks, comparison with placebo in ITT population
`
`Liraglutide (mg)
`
`0.045
`
`0.225
`
`0.45
`
`0.60
`
`0.75
`
`Glimepiride
`
`26
`28
`30
`27
`24
`26
`0.25 (0.1905) ⫺0.34 (0.0877) ⫺0.30 (0.1131) ⫺0.70 (0.0002) ⫺0.75 (⬍0.0001) ⫺0.74 (0.0001)
`0.74 (0.1499) ⫺1.37 (0.0090) ⫺0.35 (0.4976) ⫺2.14 (⬍0.0001) ⫺1.82 (0.0003)
`⫺2.60 (⬍0.0001)
`⫺0.03 (0.9602) ⫺0.74 (0.1544) ⫺1.20 (0.0184)
`0.27 (0.5838) ⫺0.39 (0.4391)
`0.94 (0.0622)
`⫺0.04 (0.6468) ⫺0.12 (0.1314) ⫺0.04 (0.6502) ⫺0.12 (0.1775) ⫺0.18 (0.0244) ⫺0.003 (0.9635)
`⫺6.07 (0.3546)
`7.55 (0.2514)
`8.55 (0.1978)
`7.33 (0.2409)
`23.56 (0.0002)
`24.57 (0.0002)
`⫺2.44 (0.8425) ⫺4.15 (0.7381) ⫺0.50 (0.9679) ⫺7.47 (0.5269)
`8.57 (0.4708)
`16.79 (0.1597)
`⫺0.09 (0.4033) ⫺0.01 (0.9449)
`0.05 (0.6374) ⫺0.08 (0.4308)
`0.11 (0.2921)
`0.08 (0.4636)
`12.74 (0.3619) ⫺15.84 (0.2710) ⫺6.77 (0.6277) ⫺7.68 (0.5625) ⫺9.33 (0.4980)
`⫺7.21 (0.5970)
`
`n
`HbAlc (%)
`Fasting serum glucose (mmol/l)
`Weight (kg)
`Proinsulin-to-insulin ratio
`␤-Cell function (%)
`Fasting insulin (mmol/l)
`Fasting C-peptide (mmol/l)
`Fasting glucagon (mmol/l)
`P value given in parentheses.
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`1339
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00005
`
`

`

`Improved glycemic control with liraglutide (NN2211)
`
`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`tected after 12 weeks of treatment. The
`sensitivity of the assay has been found to
`be 30 ng/ml (data on file) using a mono-
`clonal antibody. The liraglutide antibody
`assay is essentially similar to a previously
`published assay used to assess insulin and
`insulin aspart antibodies with a similar
`and acceptable sensitivity (33).
`Another approach for investigating
`the potential of GLP-1 includes adminis-
`tering native GLP-1 by means of continu-
`ous infusion (11,14). Continuous
`intravenous infusion of GLP-1 for 1 week
`resulted in an instant decrease in blood
`glucose levels in patients in whom sulfo-
`nylurea therapy failed to be effective (14).
`A 6-week trial with GLP-1 administered
`as continuous subcutaneous infusion via
`an insulin pump demonstrated significant
`effects on fasting and 8-h plasma glucose
`and HbA1c, as well as an inhibition of gas-
`tric emptying and body weight loss (11).
`The latter trial was conducted in patients
`with poorly controlled HbA1c levels, indi-
`cating that the GLP-1 treatment concept
`may be applied to a wide range of glyce-
`mia in type 2 diabetes. Also, the peptide
`exenatide (synthetic exendin-4) has dem-
`onstrated effects similar to those of GLP-1
`(34). In this 28-day study, adding ex-
`enatide to the existing treatment two or
`three times daily showed a significant ef-
`fect in controlling patients’ HbA1c levels,
`which were previously not satisfactorily
`controlled with diet and/or OHAs.
`In conclusion, the long-acting GLP-1
`analog liraglutide shows considerable
`promise as a once-daily therapy in type 2
`diabetes for lowering blood glucose with-
`out weight gain or substantial risk of
`hypoglycemia.
`
`Acknowledgments — This trial was spon-
`sored by Novo Nordisk A/S, which established
`the protocol in conjunction with an academic
`committee that was responsible for data re-
`view and this article. The decision to submit
`the manuscript was that of the named authors.
`We are indebted to the following principal
`investigators and their staff, who participated
`in the NN2211-1310 international study
`group. Norway: H. Ulrichsen, Harstad; A.
`Tandberg, Bekkestua; M. Wangestad, Oslo; S.
`Madsbu, Elverum. Sweden: B. Eliasson, Gote-
`borg; A. Gamstedt, Orebro; E. Bjork, Uppsala;
`B. Polhem, Goteborg. Denmark: S. Madsbad,
`Hvidovre; O. Schmitz, Aarhus; K. Koelendorf,
`Koege; H. Perrild, Copenhagen; A. Prange,
`Kolding; S. Nistrup Holmegaard, Thisted.
`
`Figure 2—Mean weight change from baseline (kg), ITT population. F, 0.045 mg liraglutide; f,
`0.225 mg liraglutide; Œ, 0.45 mg liraglutide; , 0.60 mg liraglutide; }, 0.75 mg liraglutide; [gray
`circles], glimepiride; 䡺, placebo.
`
`at an average dosage of 2.7 mg (a near-
`maximal dosage) (23), with trends toward
`less hypoglycemia and weight loss. At the
`highest dosage levels, there was a trend
`toward improved HbA1c with duration of
`treatment. This suggests that further im-
`provements in HbA1c levels may be seen
`with extended (⬎12 weeks) treatment.
`More than half the patients in the two
`highest dosage liraglutide groups had
`ⱕ 7% after 12 weeks of treatment.
`HbA1c
`Furthermore, treatment with liraglutide
`caused no weight increase, and, at the
`highest dosage, the difference with
`glimepiride was statistically significant.
`This observation was in accordance with
`findings that GLP-1 suppresses energy in-
`take in humans (8).
`The full dosage-response curve for li-
`raglutide remains to be elucidated, and
`future trials will investigate higher dos-
`ages of drug, but the current trial con-
`firmed the proposed once-daily dosing
`regimen for liraglutide.
`The proinsulin-to-insulin ratio de-
`creased during 12 weeks’ treatment with
`liraglutide. This ratio is known to be ele-
`vated in type 2 diabetes and is considered
`an index of insulin secretory dysfunction
`(24,25). It is believed that the hyperpro-
`insulinemia is caused by an increased
`demand placed on the ␤-cell by hypergly-
`cemia and insulin resistance, which in
`turn leads to secretion of an increased
`amount of incompletely processed gran-
`ules containing proinsulin. This is also
`
`known as the “overworked ␤-cell” hy-
`pothesis (26). An alternative hypothesis is
`that the increased proinsulin concentra-
`tion is related to an intrinsic ␤-cell defect
`in type 2 diabetes (24). In both cases, im-
`provement of the proinsulin-to-insulin
`ratio, and thus of ␤-cell function, would
`be a desirable effect of a potential new
`treatment for type 2 diabetes. ␤-Cell func-
`tion, as determined by HOMA analysis,
`was significantly improved at the highest
`liraglutide dosage compared with pla-
`cebo. There was no effect on the ␣-cells in
`the fasting state, in so far as the glucagon
`concentrations were unaltered.
`The safety profile of liraglutide was
`favorable, with the expected very low risk
`of hypoglycemic episodes as anticipated
`with the glucose-dependent mode of ac-
`tion of native GLP-1 (27). Adverse events
`were isolated, with the most frequent be-
`ing headache and nausea. Usually the
`events were transient and of mild or mod-
`erate severity and resolved without inter-
`vention. Although both nausea and
`vomiting were reported, neither of these
`was of a severity that caused the patient to
`withdraw. Most gastrointestinal events
`were transient (i.e., they resolved within
`1–3 days). Side effects within the gastro-
`intestinal system are known to occur after
`GLP-1 treatment and seem to be dosage
`related (14,28,29). These effects probably
`result from the inhibition of gastric emp-
`tying by GLP-1 (9,30 –32). No formation
`of antibodies toward liraglutide was de-
`
`1340
`
`DIABETES CARE, VOLUME 27, NUMBER 6, JUNE 2004
`
`Novo Nordisk Exhibit 2075
`Mylan Pharms. Inc. v. Novo Nordisk A/S
`IPR2023-00724
`Page 00006
`
`

`

`Downloaded from http://diabetesjournals.org/care/article-pdf/27/6/1335/645887/zdc00604001335.pdf by guest on 13 January 2024
`
`Madsbad and Associates
`
`prandial glycemia in type 2 diabetes. Di-
`abetes 51:424 – 429, 2002
`19. World Medical Association Declaration of
`Helsinki: recommendations guiding phy-
`sicians in biomedical research involving
`human subjects.
`JAMA 277:925–926,
`1997
`20. American Diabetes Association: Guide to
`diagnosis and classification of diabetes
`mellitus and other categories of glucose
`intolerance (Position Statement). Diabetes
`Care 20 (Suppl. 1):S21, 1997
`21. Criteria Committee of the New York Heart
`Association: Nomenclature and Criteria for
`Diagnosis of Diseases of the Heart and Great
`Vessels. Boston, Little, Brown, 1994
`22. Matthews DR, Hosker JP, Rudenski AS,
`Naylor BA, Treacher DF, Turner RC:
`Homeostasis model assessment: insulin
`resistance and beta-cell
`function from
`fasting plasma glucose and insulin con-
`centrations in man. Diabetologia 28:412–
`419, 1985
`23. Goldberg RB, Holvey SM, Schneider J: A
`dose-response study of glimepiride in pa-
`tients with NIDDM who have previously
`received sulfonylurea agents: the Glime-
`piride Protocol #201 Study Group. Diabe-
`tes Care 19:849 – 856, 1996
`24. Porte D Jr, Kahn SE: Hyperproinsuline-
`mia and amyloid in NIDDM: clues to eti-
`ology of islet ␤-cell dysfunction? Diabetes
`38:1333–1336, 1989
`25. Clark A, Jones LC, de Koning E, Hansen
`BC, Matthews DR: Decreased insulin se-
`cretion in type 2 diabetes: a problem of
`cellular mass or function? Diabetes 50
`(Suppl. 1):S169 –S171, 2001
`function
`26. Leahy JL:
`Impaired beta-cell
`with chronic hyperglycemia: “overworked
`␤-cell” hypothesis. Diabetes Rev 4:298 –
`319, 1996
`27. Nauck MA, Heimesaat MM, Behle K,
`Holst JJ, Nauck MS, Ritzel R, Hufner M,
`Schmiegel WH: Effects of glucagon-like
`peptide 1 on counterregulatory hormone
`responses, cognitive functions, and insu-
`lin secretion during hyperinsulinemic,
`stepped hypoglycemic clamp experi-
`ments in healthy volunteers. J Clin Endo-
`crinol Metab 87:1239 –1246, 2002
`28. Ritzel R, Orskov C, Holst JJ, Nauck
`MA: Pharmacokinetic, insulinotropic, and
`glucagonostatic properties of GLP-1 [7-
`36 amide] after subcutaneous injection in
`healthy volunteers: dose-response-rela-
`tionships. Diabetologia 38:720–725, 1995
`29. Nauck MA, Wollschlager D, Werner J,
`Holst
`JJ, Orskov C, Creutzfeldt W,
`Willms B: Effects of subcutaneous gluca-
`gon-like peptide 1 (GLP-1 [7–36 amide])
`in patients with NIDDM. Diabetologia 39:
`1546 –1553, 1996
`30. Layer P, Holst JJ, Grandt D, Goebell H:
`Ileal release of glucagon-like peptide-1
`(GLP-1): association with inhibition of
`
`U.K.: R. Blandford, Worksop; C. Fox, North-
`hampton; B. Frier, Edinburgh; R. Gregory,
`Leicester; S. Heller, Sheffield; D. Hepburn,
`Hull; A. Jaap, Kilmarnock; P. Mansell, Not-
`tingham; D. Matthews, Oxford; B. Millward,
`Plymouth; P. O’Hare, Rugby; M. Page, Llant-
`risant; J. Vora, Liverpool; A. Ogilvie, Enfield.
`The study results were presented in an oral
`presentation at the American Diabetes Associ-
`ation 62nd Annu

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