throbber
Current Opinion in Molecular Therapeutics 2010 12(6):790-797
`
` Thomson Reuters (Scientific) Ltd ISSN 2040-3445
`
`DRUG PROFILE
`
`Dulaglutide, a long-acting GLP-1 analog fused with an Fc antibody
`fragment for the potential treatment of type 2 diabetes
`Espen Jimenez-Solem1, Mette H Rasmussen2, Mikkel Christensen3 & Filip K Knop3*
`
`Addresses
`1University of Copenhagen, Bispebjerg Hospital, Department of Clinical Pharmacology,
`Tagensvej 20, 2200 Copenhagen, Denmark
`
`2University of Copenhagen, Bispebjerg Hospital, Department of General Practice,
`Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
`
`3University of Copenhagen, Gentofte Hospital, Department of Internal Medicine F,
`Niels Andersens Vej 65, 2900 Hellerup, Denmark
`Email: filipknop@dadlnet.dk
`
`*To whom correspondence should be addressed
`
`Dulaglutide (LY-2189265) is a novel, long-acting glucagon-like peptide 1 (GLP-1) analog being developed by Eli Lilly for the treatment
`of type 2 diabetes mellitus (T2DM). Dulaglutide consists of GLP-1(7-37) covalently linked to an Fc fragment of human IgG4, thereby
`protecting the GLP-1 moiety from inactivation by dipeptidyl peptidase 4. In vitro and in vivo studies on T2DM models demonstrated
`glucose-dependent insulin secretion stimulation. Pharmacokinetic studies demonstrated a t1/2 in humans of up to 90 h, making
`dulaglutide an ideal candidate for once-weekly dosing. Clinical trials suggest that dulaglutide reduces plasma glucose, and has an
`insulinotropic effect increasing insulin and C-peptide levels. Two phase II clinical trials demonstrated a dose-dependent reduction in
`glycated hemoglobin (HbA1c) of up to 1.52% compared with placebo. Side effects associated with dulaglutide administration were
`mainly gastrointestinal. To date, there have been no reports on the formation of antibodies against dulaglutide, but, clearly, long-term
`data will be needed to asses this and other possible side effects. The results of several phase III clinical trials are awaited for
`clarification of the expected effects on HbA1c and body weight. If dulaglutide possesses similar efficacy to other GLP-1 analogs, the
`once-weekly treatment will most likely be welcomed by patients with T2DM.
`
`Introduction
`In 2000, an estimated 171 million people worldwide
`had diabetes (all types). This igure is expected to
`increase by 144% to 366 million by 2030, as a result of
`increasing
`longevity, obesity and sedentary
`lifestyle
`[880516]. The most common
`form of diabetes
`is
`type 2 diabetes mellitus (T2DM), which is characterized
`by two main physiological defects: β-cell dysfunction
`and insulin resistance [995537], [995560]. Additionally,
`T2DM
`is characterized by
`fasting and postprandial
`hyperglucagonemia contributing
`to
`the hyperglycemic
`state of
`the patients via glucagon-induced hepatic
`glucose production. It
`is evident
`that T2DM
`is a
`progressive disease with an almost linear decline in β-cell
`function over time [995565]. Consequently, the current
`treatment paradigm aims to counteract the progressive
`deterioration in blood glucose homeostasis.
`include
`The classical oral antidiabetic drugs (OADs)
`insulin sensitizers, such as biguanides (eg, metformin)
`and thiazolidinediones (TZDs; eg, pioglitazone), insulin
`secretagogs, such as sulfonylureas (SUs; eg, glibenclamide)
`and meglitinides (eg, repaglinide), or drugs that delay
`glucose absorption
`through gut enzyme
`inhibition,
`
`Therapeutic Dulaglutide
`
`Originator Eli Lilly
`
`Status Phase III Clinical
`
`Indication Type 2 diabetes mellitus
`
`Actions Glucagon-like peptide-1 analog, Glucose-lowering agent
`
`Technologies Antibody fragment, Biological therapeutic, Protein
`fusion, Subcutaneous formulation
`
`Synonym LY-2189265
`
`such as acarbose. These medications enable modest
`reductions of glycated hemoglobin (HbA1c), usually
`between 0.5 and 1.5%, but are not able to correct either
`the impairment or the progressive decline of β-cell function
`[995569]. Therefore, regardless of optimal treatment, the
`endogenous insulin response becomes attenuated, and,
`in later stages of disease, lost, necessitating exogenous
`insulin replacement [931356]. It is currently believed
`that treatments that preserve or improve β-cell function
`may halt or delay disease progression in T2DM, thereby
`
`MPI EXHIBIT 1029 PAGE 1
`
`MPI EXHIBIT 1029 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1029-0001
`
`

`

`insulin
`for
`the need
`delaying or even preventing
`therapy [995571]. Medications that are able to address
`cardiovascular risk factors, as well as provide glycemic
`control and preserve or improve β-cell function, thereby
`possibly halting or delaying disease progression, are
`highly anticipated [995571].
`The scientiic knowledge regarding optimal HbA1c levels
`has been recently complicated by the publication of
`mega trials demonstrating limitations to the paradigm
`of an overzealous glucose lowering strategy [1048568],
`[1147074],
`[1147078]. However,
`because
`of
`the
`long-standing evidence
`for an association between
`poor glycemic control and long-term diabetes-related
`complications, the overall scientiic consensus and major
`guidelines still advocate intensiication of treatment to
`maintain a HbA1c level of < 7.0% [996028]. The majority
`of patients fail to achieve this target, as exempliied in a
`recent trial in the US, which demonstrated that more
`than half of patients with diabetes did not meet a target
`HbA1c level of < 7.0% [996031]. This may, in part,
`be a result of reluctance on the part of physicians to
`prescribe
`therapies with
`unwanted
`side
`effects;
`hypoglycemia, a common side effect of insulin and also
`frequently associated with SUs, may be of particular
`concern. Diabetes is a complex disease to manage and
`primary care physicians rate diabetes as harder to treat
`than other common diseases [996035]. Another limiting
`factor to treatment may be the patient's own acceptance
`of a prescribed regimen. Common barriers to patient
`adherence
`include concern about unwanted weight
`gain [996037],
`fear of hypoglycemia and perceived
`inconvenience
`[996040],
`[996603], which may all
`indirectly undermine glycemic control if the prescribed
`therapy is not followed.
`is
`insulin secretion
`Under physiological conditions,
`including
`the
`incretin
`controlled by several
`factors
`hormone glucagon-like peptide-1 (GLP-1). This hormone,
`secreted by endocrine cells in the intestine, stimulates
`glucose-dependent insulin secretion and accounts for a
`considerable part of the insulin response to glucose
`[827063], [996043], [1147079]. The physiological effects
`of GLP-1 are mediated by a G-protein-coupled receptor
`[996045], which is widely distributed across different
`tissues. As a result, the peripheral and central nervous
`systems, heart, stomach,
`lungs,
`intestines, pituitary,
`endothelium, kidneys and pancreas are all affected by
`GLP-1 [827063], [996047]. In addition to its incretin
`effect, GLP-1 exerts several effects
`that are also
`potentially beneicial in the treatment of T2DM. These
`include preservation of β-cell mass [439499], [475556],
`[996049], potentiation of glucose-induced suppression
`from pancreatic a-cell, a
`of glucagon
`secretion
`protective effect on the cardiovascular system [580626],
`[996071], delayed gastric emptying and an increase in
`the feeling of satiety [996074]; the latter two factors
`can contribute to a decreased energy intake.
`The early established role of GLP-1 in glucose homeostasis
`prompted research into its potential for therapeutic use
`in T2DM [874585], [875837]. It is noteworthy that in
`
`Dulaglutide Jimenez-Solem et al 791
`
`patients with T2DM, despite a frequently impaired secretion
`of GLP-1 [875837], [996163], [996169], the insulinotropic
`effect is preserved [931364]. This established GLP-1 as
`a therapeutic candidate for the treatment of T2DM, and
`subsequent trials have demonstrated that exogenous
`native GLP-1 normalizes the insulin response in individuals
`with impaired glucose tolerance [996079] and decreases
`blood glucose levels in patients with T2DM [475562],
`[760698].
`While native GLP-1 offers signiicant beneits for patients
`with T2DM, its rapid breakdown by the enzyme dipeptidyl
`peptidase 4 (DPP-4) incurs the critical disadvantage of a
`short half-life
`(1
`to 2 min)
`following
`intravenous
`administration [996173]; thus, native GLP-1 would need
`to be continually infused for effective therapy. To circumvent
`this physiological degradation, DPP-4-resistant GLP-1
`analogs have been developed. Two such compounds are
`currently on the market: exenatide, a synthetic version
`of exendin-4(1-39), which is a hormone isolated from
`the saliva of the Gila monster lizard [996232], and
`liraglutide, an acylated GLP-1 analog with 97% amino acid
`sequence homology to endogenous human GLP-1(7-37)
`[1070808]. However, many more agents are in phase II
`or III clinical development [1147081].
`The focus of this review is dulaglutide (LY-2189265), a
`novel, long-acting GLP-1 analog, being developed by
`Eli Lilly for the treatment of T2DM. Dulaglutide consists
`of a DPP-4–protected GLP-1 analog covalently linked to
`an Fc fragment of human IgG4, thereby increasing its
`duration of pharmacological activity. The irst phase III
`clinical trials of dulaglutide were initiated in early 2008
`[970293].
`
`Synthesis and SAR
`Dulaglutide comprises a DPP-4-protected GLP-1(7-37)
`analog fused to a modiied IgG4 Fc fragment [1100363].
`Initial studies using DPP-4-protected GLP-1 fused to
`IgG1 demonstrated signiicantly reduced in vitro activity
`compared with
`free DPP-4-protected GLP-1. Linker
`sequences between the C-terminus of the GLP-1 analog
`and the N-terminus of the Ig molecule were assessed
`in an attempt to
`improve activity, and IgG1 was
`replaced with a modiied IgG4 (Phe234Ala and Leu235Ala to
`reduce interaction with high-afinity Fc receptors, and
`Ser228Pro to eliminate half-antibody formation) to reduce
`the potential of complement- and/or antibody-dependent
`cell-mediated cytotoxicity. An Arg36Gly mutation
`in
`GLP-1 was introduced to de-immunize the fusion protein
`and the C-terminal lysine of the IgG4-Fc was removed.
`Dulaglutide exhibited 4-fold greater
`in vitro activity
`than the free DPP-4-protected GLP-1. The GLP-1-Fc fusion
`proteins used in these studies were expressed from
`HEK293-EBNA cells [1100363].
`
`Preclinical development
`In vitro
`Isolated rat islets were used to measure the effect
`of dulaglutide on glucose-induced
`insulin secretion
`[1100363]. At high glucose levels (16.8 mM) the insulin
`
`MPI EXHIBIT 1029 PAGE 2
`
`MPI EXHIBIT 1029 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1029-0002
`
`

`

`792 Current Opinion in Molecular Therapeutics 2010 Vol 12 No 6
`
`secretion was enhanced 2.5- to 3-fold by the addition
`of 3 or 30 nM dulaglutide to the extracellular medium.
`No enhancement was observed at low glucose levels
`(2.8 mM). In comparison, human GLP-1 (3 nM) caused a
`4-fold increase in insulin secretion at 16.8 mM glucose.
`At 2.7 nM dulaglutide, half-maximal stimulation of
`insulin secretion was observed, and the maximal 4-fold
`stimulation was observed with 300 nM dulaglutide. The
`addition of the GLP-1 receptor antagonist exendin(9-39)
`(1 µM) to the medium reversed the effects of dulaglutide
`on insulin secretion. In islets isolated from cynomolgus
`monkeys, dulaglutide increased insulin secretion in a
`concentration-dependent manner at high concentrations
`of glucose [1100363].
`
`In vivo
`
`Dose-dependent increases in glucose-dependent insulin
`secretion were demonstrated 24 h after a single dose
`of dulaglutide (0.3, 1, 3 or 30 nmol/kg sc) in conscious
`rats receiving a graded glucose infusion. However, only
`the 3- and 30-nmol/kg doses achieved statistically
`signiicant (p < 0.05) increases in insulin secretion (up to
`4-fold) compared with vehicle [1100363].
`Insulin secretion in response to graded glucose infusion
`was evaluated 1, 5 and 7 days after a single dose of
`dulaglutide (1.7 nmol/kg sc) to cynomolgus monkeys
`[1100363]. Plasma concentrations of dulaglutide remained
`detectable
`throughout
`the 7 days.
`Insulin
`(2-fold
`versus vehicle; p < 0.0001) and C-peptide levels were
`signiicantly enhanced during the 7-day period, whereas
`glucose, GLP-1 and glucagon levels were unaffected.
`Multiple dosing of dulaglutide (1.7 nmol/kg sc, qw for
`4 weeks) before graded glucose infusion (4 days after the
`last dose) was also assessed in cynomolgus monkeys.
`Progressively
`increasing
`glucose-stimulated
`insulin
`release (2-fold versus vehicle; p < 0.0002) and C-peptide
`levels were observed following the glucose infusion.
`Triglyceride
`levels were
`reduced, but glucose and
`glucagon levels remained unaltered [1100363].
`In 5-week-old diabetic (db/db) mice treated with dulaglutide
`(10 nmol/kg sc, biw for 4 weeks), plasma glucose levels
`were consistently decreased
`throughout
`the study
`period (p < 0.001 versus vehicle); treated mice also
`exhibited a small, but signiicant, reduction in body
`mass versus vehicle (body mass = 35.5 versus 38.5 g;
`p < 0.02) [1100363].
`
`Toxicity
`At the time of publication, toxicity data were not available.
`
`Metabolism and pharmacokinetics
`The pharmacokinetic proile of single-dose dulaglutide
`(0.1 mg/kg sc) was assessed in rats and cynomolgus
`monkeys. In rats and monkeys respectively, Cmax values
`were 179.7 and 292.2 ng/ml, Tmax values were 24.0 and
`16.7 h, AUC0-∞ values were 10,537 and 15,207 ng/ml·h,
`t1/2 values were 38.2 and 51.6 h, clearance was 9.6 and
`
`7.3 ml/h/kg, and the volume of distribution was 525.0
`and 557.5 ml/kg [1100363].
`The pharmacokinetics of dulaglutide (0.1 to 12 mg sc;
`administered ≥ 3 weeks apart) were analyzed in healthy
`volunteers (n = 20). The t1/2 value of dulaglutide was
`~ 90 h and the Tmax value was between 24 and 48 h.
`When the dose was doubled, Cmax and AUC0-∞ of plasma
`dulaglutide increased by 1.84- and 1.88-fold, respectively,
`suggesting
`linear kinetics
`[1013502],
`[1087432].
`In
`patients (n = 43) with T2DM, dulaglutide (0.05 to
`8 mg sc, qw) had a t1/2 value of ~ 95 h [1087431],
`[1087438].
`
`Clinical development
`Phase I
`In a randomized, double-blind, placebo-controlled, three-
`period, crossover, single-center clinical trial, the safety,
`tolerability, pharmacokinetics and insulinotropic activity
`of escalating single doses of dulaglutide (0.1 to 12 mg sc;
`administered ≥ 3 weeks apart) were analyzed in healthy
`volunteers (n = 20). Compared with placebo, there
`was a glucose-dependent increase in insulin secretion
`after graded glucose infusion, and a suppression of
`plasma glucose after an oral glucose tolerance test at
`all doses [1013502], [1087432].
`In an open-label, parallel clinical trial, the effects of
`dulaglutide (1 or 3 mg sc, qw for 4 weeks) on gastric
`emptying were assessed in healthy volunteers (n = 30)
`using paracetamol (1 g po) pharmacokinetics as a probe.
`Both the 1- and 3-mg doses of dulaglutide caused a
`reduction in paracetamol Cmax (36 and 50%, respectively)
`and a delay in Tmax (1 and 2 h, respectively), indicating a
`delay in gastric emptying; this was only observed after
`the irst dose of dulaglutide and not at steady-state. The
`body mass of volunteers was also signiicantly decreased
`by 1.4 and 2.4 kg from baseline in the 1- and 3-mg dose
`groups, respectively [1109660].
`The effects of dulaglutide (1.5 mg sc, qw for 4 weeks) on
`gastric emptying would also be assessed by scintigraphy
`in a phase I, randomized, double-blind, placebo-controlled
`clinical trial (ClinicalTrials.gov identiier: NCT01215968).
`At the time of publication, patients (estimated n = 40)
`with T2DM were being recruited to this trial.
`
`Phase II
`
`A double-blind, placebo-controlled, parallel group, 5-week
`clinical trial assessed the effects of dulaglutide (0.05 to
`8 mg sc, qw) in patients (n = 43) with T2DM. Statistically
`signiicant decreases in HbA1c (-0.69 to -1.34%) were
`observed at all dose levels. Increases in insulin and
`C-peptide AUC values were also observed, indicating an
`insulinotropic effect. Fasting and postprandial plasma
`glucose excursions were signiicantly
`reduced with
`dulaglutide doses ≥ 1 mg. At dulaglutide doses ≥ 5 mg,
`statistically signiicant effects on gastric emptying after
`the irst dose and weight loss after the last dose were
`observed [1087431], [1087438].
`
`MPI EXHIBIT 1029 PAGE 3
`
`MPI EXHIBIT 1029 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1029-0003
`
`

`

`randomized, double-blinded, placebo-
`II,
`A phase
`controlled, parallel assignment, multicenter clinical trial
`(EGO; NCT00630825) assessed the safety and eficacy of
`dulaglutide
`in patients (n = 262) with uncontrolled
`T2DM (baseline HbA1c = 8.2%) and a BMI of 33.9 kg/m2.
`Patients were randomized into three dulaglutide groups
`or a matching placebo. The dulaglutide groups were:
`0.5 mg (sc, qw) for 4 weeks titrated to 1.0 mg (sc, qw)
`for 12 weeks, 1.0 mg (sc, qw) for 16 weeks, or 1.0 mg
`(sc, qw) for 4 weeks titrated to 2.0 mg (sc, qw) for
`12 weeks [1016672], [1087434], [1087444], [1109617],
`[1122363], [1140475]. At 16 weeks, the mean reduction
`from baseline in HbA1c was -1.28, -1.29 and -1.52%
`in the 0.5/1.0-, 1.0/1.0- and 1.0/2.0-mg dose groups,
`respectively,
`compared with
`-0.27%
`for placebo
`(p < 0.001). Reductions were also observed in fasting
`plasma glucose (-2.09, -2.04 and -2.64 versus -0.49 mm;
`p < 0.001), solid mixed meal test glucose excursions
`(30.71, 32.21 and 28.24 versus 36.36 mM∙h; p < 0.001)
`and body mass (-1.58, -1.40 and -2.51 versus -0.07 kg;
`p < 0.05) [1016672], [1087444]. β-cell function was
`also signiicantly increased compared with placebo, as
`assessed by
`the homeostatic model assessment of
`β-cell function (HOMA%B: 39.20, 44.26 and 45.61 versus
`1.04%; p < 0.05) [1087444].
`In addition, the impact of Hispanic ethnicity was assessed
`[1109617], [1122363], [1140475]. At baseline, HbA1c
`levels were signiicantly greater (p = 0.006) in Hispanic
`patients (8.4%) compared with non-Hispanic Caucasian
`patients (8.1%); the decrease in HbA1c at week 16 was
`also greater in Hispanic patients (-1.5 versus -1.1%;
`p = 0.02). Furthermore, the reduction in postprandial
`glucose excursion was signiicantly greater in Hispanic
`patients (-2.8 versus -0.5 mM∙h; p = 0.003), although fasting
`plasma glucose, insulin levels and β-cell function were
`not affected by ethnicity [1109617], [1122363], [1140475].
`Further to these trials, additional phase II, randomized,
`double-blind,
`placebo-controlled, multicenter
`clinical
`trials of dulaglutide were listed in the NIH clinical trial
`registry at the time of publication. Trial NCT00791479
`assessed
`the dose-dependent effects of dulaglutide
`(0.1, 0.5, 1.0 and 1.5 mg sc, qw for up to 12 weeks)
`on glycemic control in patients (estimated n = 168)
`with T2DM;
`this
`trial had been completed. Trial
`NCT01001104 was
`assessing
`the
`dose-response
`characteristics of dulaglutide (0.25, 0.50 and 0.75 mg sc,
`qw for up to 12 weeks) on HbA1c in Japanese patients
`(estimated n = 144) with inadequately controlled T2DM
`who were taking no more than one OAD (not DPP-4
`inhibitors); patient recruitment to this trial was complete,
`although the trial was still ongoing. Data were not
`available from either trial.
`
`Phase III
`
`At the time of publication, a phase II/III, randomized,
`double-blind,
`placebo-controlled, multicenter
`clinical
`trial (NCT00734474) comparing dulaglutide (0.25, 0.5,
`0.75, 1.0, 1.5, 2.0 and 3.00 mg sc, qw) with the DPP-4
`inhibitor sitagliptin (100 mg po, qd) was ongoing in
`
`Dulaglutide Jimenez-Solem et al 793
`
`patients (estimated n = 1566) with T2DM. Treatment
`would last for 2 years and the primary outcome was
`HbA1c change from baseline to 12 months.
`Additionally, Eli Lilly has
`initiated a program of
`phase III, randomized, multicenter clinical trials known as
`AWARD. The placebo-controlled, double-blind AWARD-1
`(NCT01064687) clinical
`trial was
`recruiting patients
`(estimated n = 980) with T2DM inadequately controlled
`with metformin and pioglitazone. This
`trial would
`determine the eficacy of dulaglutide (0.75 and 1.5 mg sc, qw),
`compared with
`placebo
`or
`exenatide,
`as
`an
`add-on to baseline therapy. The open-label AWARD-2
`(NCT01075282) clinical
`trial was
`recruiting patients
`(estimated n = 837) with T2DM inadequately controlled
`with metformin and the SU agent glimepiride. The
`trial would compare
`the eficacy and safety of
`dulaglutide (0.75 and 1.5 mg sc, qw), compared with
`insulin glargine, as an add-on to baseline therapy. The
`placebo-controlled, double-blind AWARD-3 (NCT01126580)
`clinical trial was recruiting patients (estimated n = 753)
`with T2DM. The trial would compare the eficacy and
`safety of dulaglutide (0.75 and 1.5 mg/kg sc, qw) with
`metformin (1500 to 2000 mg/day po). The open-label
`AWARD-4 (NCT01191268) clinical
`trial would
`recruit
`patients (estimated n = 837) with T2DM. The trial would
`compare the eficacy and safety of dulaglutide (0.75
`and 1.5 mg/kg sc, qw) with insulin glargine, both in
`combination with insulin lispro.
`
`Side effects and contraindications
`In
`the
`trial evaluating single escalating doses of
`dulaglutide (0.1 to 12 mg sc) in healthy volunteers, the
`most frequent adverse event was dyspepsia. Statistically
`signiicant
`increases
`in supine heart rate at doses
`≥ 0.3 mg and in supine diastolic blood pressure at
`doses ≥ 1 mg compared with placebo were noted.
`Dose-dependent increases in the rates of headache,
`injection-site irritation, nausea and vomiting were also
`reported. Importantly, hypoglycemia or antibodies against
`dulaglutide were not reported [1013502], [1087432].
`In the trial evaluating the effects of dulaglutide (1 or 3 mg)
`on gastric emptying
`in healthy volunteers, safety
`and
`tolerability were also assessed. Gastrointestinal
`disturbances were the most frequent adverse events.
`Furthermore, signiicant
`increases
`from baseline
`in
`pulse rate (7.8 and 9.6 bpm at 1 and 3 mg, respectively)
`and decreases from baseline in systolic blood pressure
`(4.1 and 7.7 mmHg at 1 and 3 mg, respectively) were
`also observed [1109660].
`In the 5-week trial of dulaglutide (0.05 to 8 mg sc, qw)
`in patients with T2DM, dulaglutide was generally well
`tolerated. Nausea, vomiting, headache and diarrhea
`were the most common adverse events. At the 5-mg
`dose, a statistically signiicant increase in heart rate was
`reported. Antibodies against dulaglutide were not
`detected [1087431], [1087438].
`
`MPI EXHIBIT 1029 PAGE 4
`
`MPI EXHIBIT 1029 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1029-0004
`
`

`

`794 Current Opinion in Molecular Therapeutics 2010 Vol 12 No 6
`
`In the EGO trial in patients with T2DM, dulaglutide
`(1 mg for 16 weeks, 0.5 mg for 4 weeks titrated to
`1 mg for 12 weeks, or 1 mg for 4 weeks titrated to
`2 mg for 12 weeks; all sc qw) was generally well tolerated.
`The most common adverse events were nausea (13%),
`diarrhea
`(8.8%) and abdominal distension
`(8.0%).
`The frequency of hypoglycemic episodes did not differ
`among treatment groups [1016672], [1087444].
`Because of the effects on heart rate and blood pressure
`observed
`in
`the
`initial clinical
`trials, a phase
`II,
`randomized, double-blind, placebo-controlled, multicenter
`clinical trial (NCT01149421) was initiated to assess these
`parameters. Patients (estimated n = 693) with T2DM
`treated with one or more OAD were treated with
`dulaglutide (0.75 and 1.5 mg sc, qw) for up to 26 weeks.
`Inclusion criteria included HbA1c between 7 and 9.5%,
`blood pressure > 90/60 and < 140/90 mmHg, and
`BMI ≥ 23 kg/m2.
`
`Patent summary
`The genesis of dulaglutide appears to have come from
`the GLP-1
`fusion proteins
`claimed
`in Eli Lilly's
`WO-00246227, most
`likely protecting
`forms of
`the
`company's GLP-1 analog LY-548806 and involving some
`of the same inventors as listed on the product patent
`for dulaglutide. Although WO-00246227 has generic
`claims covering the GLP-1 analog and the peptide linker
`present in dulaglutide, the IgG4 Fc component does
`not have the amino acid substitutions present in the
`dulaglutide moiety that stabilize heavy chain dimer
`formation and eliminate their natural effector function.
`The irst
`two claims of WO-2005000892 describe
`dulaglutide, including its peptide linker. WO-2005000892
`is part of the WO-2004110472 family covering a whole
`range of Fc fragment fusion proteins including one based
`on FGF-21. The granted equivalents relating speciically
`to GLP-1
`fusion proteins are EA-00008831 and
`EP-01641483 both expiring in June 2024, and US-07452966
`expiring in July 2024 after a short patent term extension.
`Further patenting by Eli Lilly has concentrated on
`providing more stable
`formulations
`for dulaglutide.
`WO-2006068910 claims a buffer solution
`for
`the
`protein, which gives greater resistance to protease, and
`WO-2009009562 claims a solution giving greater physical
`stability. Only the earlier of the two applications has had
`any granted applications, by the European and Eurasian
`authorities. There
`is also WO-2009020802 claiming
`combinations of dulaglutide or LY-548806 with FGF-21
`for the potential treatment of obesity.
`The only patenting on dulaglutide by third parties has
`been by Amylin Pharmaceuticals, Eli Lilly's partner for
`the development and
`launch of exenatide. Amylin's
`WO-2009143014 claims a cell-based assay for measuring
`the activity of multiple GLP-1 agonists,
`including
`dulaglutide and exenatide.
`
`Current opinion
`Incretin-based therapies represent an important addition
`to the conventional antidiabetic treatments by targeting
`several key metabolic defects in T2DM. Even if the
`promising effects on β-cell proliferation and preservation
`observed in preclinical studies are not replicated in the
`clinic, the GLP-1 analogs as a class still represent strong
`drug candidates. Firstly,
`they represent a versatile
`physiological approach, targeting both pancreatic a- and β-cell
`dysfunction and obesity, combined with improvements
`in glycemic indices that are comparable with traditional
`OADs. Secondly, severe
`limitations associated with
`existing therapies, including the risk of hypoglycemia
`(as observed with insulin and SUs) and unsuitability for
`patients with renal impairment (metformin) or signiicant
`cardiovascular disease
`(TZDs),
`creates opportunity
`for incretin-based therapies in an expanding diabetic
`drug market. Thirdly, there are several potential future
`indications that may prove worthwhile to pursue, such
`as management of obesity, postmyocardial
`infarction
`size reduction [996601], adjunction to insulin therapy in
`patients with type 1 diabetes mellitus [996602] and,
`depending on the eventual trophic effects on β-cells,
`pancreatic islet transplantation patients.
`the GLP-1
`Regarding
`the eficacy and safety of
`analogs, there appears to be a class effect. The main
`difference between compounds appears to rely in the
`pharmacokinetic proile. In this regard, the relatively long
`t1/2 value of dulaglutide (~ 90 h) could prove to be a
`signiicant advantage. Clinical data suggests that a more
`stable pharmacokinetic proile (ie, a longer t1/2) leads
`to higher eficacy, and lower frequencies of nausea and
`vomiting [953142]. Furthermore, the beneits to patient
`convenience and compliance are obvious.
`There are also limitations to consider. Firstly, there is a
`general problem concerning gastrointestinal side effects,
`especially nausea. Even if the nausea and vomiting
`were more infrequent with dulaglutide (because of an
`increased t1/2) compared with some other GLP-1 analogs,
`its use will still be limited to patients who are able to
`manage these side effects and, for example, use them
`as a positive reinforcement strategy for weight loss.
`Ultimately,
`it appears that ~ 5% of patients with
`T2DM do not tolerate GLP-1 analogs currently on the
`market [976021], [996244], [996601]. Secondly,
`the
`consequences of antibody formation are not yet fully
`known. Evidence
`from clinical
`trials of exenatide
`suggests some signiicance with regard to glycemic
`control especially in patients exhibiting high antibody
`titers [996596]. Dulaglutide consists of GLP-1(7-37)
`covalently linked to an Fc fragment of human IgG4,
`thereby protecting it from DPP-4 cleavage and increasing
`its duration of pharmacological activity. One concern is
`that
`treating patients with engineered bioproducts
`could lead to the formation of antibodies against foreign
`epitopes. From the published data, it is not possible to
`ignore the potential formation of antidrug antibodies,
`which could lead to decreased eficacy of dulaglutide in
`
`MPI EXHIBIT 1029 PAGE 5
`
`MPI EXHIBIT 1029 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1029-0005
`
`

`

`Dulaglutide Jimenez-Solem et al 795
`
`is the
`isotope, which
`patient subgroups. The IgG4
`carrier protein for dulaglutide, has so far been unable
`to demonstrate that it activates complement in vitro
`[1147082]. However, further evaluation and long-term
`data,
`including measurements of anti-GLP-1-Fc titers
`in humans, are required. Thirdly, it is evident from
`studies of barriers to
`insulin therapy that
`injection
`therapy remains a signiicant concern for many patients
`and diabetes care providers [996040], [996603], which
`may
`limit
`the acceptance of dulaglutide. However,
`as hypoglycemia is also a major concern, it can be
`speculated that the
`low
`frequency of hypoglycemia,
`combined with weight loss and only once weekly dosing,
`could make dulaglutide more acceptable than insulin
`
`therapy.
`
`Demonstration of comparable eficacy (not only on
`HbA1c, but also on mortality and/or hard micro
`and/or macrovascular endpoints) and,
`in particular,
`long-term safety compared with other GLP-1 analogs,
`incretin enhancers (ie, DPP-4 inhibitors) or even other
`OADs will be of crucial importance in determining the
`role of dulaglutide. The
`incretin enhancers, which
`also
`increase
`levels of GLP-1 agonism,
`lack
`the
`weight-reducing capacity (albeit weight neutral) observed
`with GLP-1 analogs, but the lower incidence of nausea
`
`and the oral route of administration clearly favors the
`use of these agents as initial therapy.
`In conclusion, the GLP-1-Fc fusion protein dulaglutide is a
`new GLP-1 analog intended for once-weekly dosing with
`longer plasma residence than the native hormone. The
`pharmacokinetic proile is the main difference between
`dulaglutide and the other GLP-1 analogs. Conirmation
`that dulaglutide and other GLP-1 analogs can alter the
`natural course of diabetes is still awaited. With the
`present knowledge,
`the use
`in clinical practice of
`GLP-1 analogs is justiied by the evidence of improved
`postprandial and throughout-the-day glycemic control,
`resulting
`in placebo-corrected
`lowering of HbA1c by
`0.5 to 1.5%. The main limitations of the clinical use of
`GLP-1 analogs are the insuficient long-term safety and
`eficacy data, subcutaneous route of administration and
`the high treatment cost compared with the classical
`therapeutic agents. Consequently, a place for GLP-1
`analogs in endocrinologists' drug armamentaria appears
`certain, but with only minor differences in potency
`between the different GLP-1 analogs, head-to-head trials
`will be needed to determine the drug of choice. Lastly,
`the potential of GLP-1 analogs to reduce cardiovascular
`disease and
`increase
`lifespan among T2DM patients
`still needs to be conirmed in long-term clinical trials.
`
`Development status
`
`Developer
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Eli Lilly
`
`Country
`
`Canada
`
`Europe
`
`India
`
`South Korea
`
`Taiwan
`
`US
`
`South America
`
`Australia
`
`South Africa
`
`Puerto Rico
`
`Japan
`
`Status
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase III
`
`Phase II
`
`Indication
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Type 2 diabetes mellitus
`
`Date
`
`28-AUG-08
`
`28-AUG-08
`
`28-AUG-08
`
`28-AUG-08
`
`28-AUG-08
`
`28-AUG-08
`
`28-AUG-08
`
`23-FEB-10
`
`18-MAY-10
`
`18-MAY-10
`
`22-OCT-09
`
`Reference
`
`942857
`
`942857
`
`942857
`
`942857
`
`942857
`
`942857
`
`942857
`
`1088280
`
`1146159
`
`1146159
`
`1088285
`
`Associated pate

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