throbber
I
`
`New Drug Class
`
`The incretin system: glucagon-like peptide-1 receptor
`agonists and dipeptidyl peptidase-4 inhibitors in type 2
`diabetes
`
`Daniel J Drucker, Michael A Nauck
`
`Glucagon-like peptide 1 (GLP-1) is a gut-derived incretin hormone that stimulates insulin and suppresses glucagon
`secretion, inhibits gastric emptying, and reduces appetite and food intake. Therapeutic approaches for enhancing
`incretin action include degradation-resistant GLP-1 receptor agonists (incretin mimetics), and inhibitors of dipeptidyl
`peptidase-4 (DPP-4) activity (incretin enhancers). Clinical trials with the incretin mimetic exenatide (two injections
`per day or long-acting release form once weekly) and liraglutide (one injection per day) show reductions in fasting
`and postprandial glucose concentrations, and haemoglobin A1c (HbA1c) (1–2%), associated with weight loss (2–5 kg).
`The most common adverse event associated with GLP-1 receptor agonists is mild nausea, which lessens over time.
`Orally administered DPP-4 inhibitors, such as sitagliptin and vildagliptin, reduce HbA1c by 0·5–1·0%, with few
`adverse events and no weight gain. These new classes of antidiabetic agents, and incretin mimetics and enhancers,
`also expand β-cell mass in preclinical studies. However, long-term clinical studies are needed to determine the benefi ts
`of targeting the incretin axis for the treatment of type 2 diabetes.
`
`Lancet 2006; 368: 1696–705
`Samuel Lunenfeld Research
`Institute, Mount Sinai
`Hospital, University of Toronto,
`Toronto, Ontario, Canada
`(Prof D J Drucker MD); and
`Diabeteszentrum Bad
`Lauterberg, Bad Lauterberg,
`Germany (Prof M A Nauck MD)
`Correspondence to:
`Prof Daniel J Drucker,
`Samuel Lunenfeld Research
`Institute, Mount Sinai Hospital,
`600 University Avenue, Toronto,
`Ontario, Canada M5G 1X5
`d.drucker@utoronto.ca
`
`Adipose tissue
`
`Glucose uptake ↑
`Glycogen synthesis ↑
`(? indirect actions)
`
`Liver
`
`Muscle
`
`Introduction
`Eating provokes the secretion of multiple gastrointestinal
`hormones involved in the regulation of gut motility,
`secretion of gastric acid and pancreatic enzymes, gall
`bladder contraction, and nutrient absorption. Gut
`hormones also facilitate the disposal of absorbed glucose
`through the stimulation of insulin secretion from the
`
`Brain/nervous system:
`Hypothalamus
` Appetite ↓, satiety ↑, food intake ↓, water intake ↓
`Nucleus tractus solitarii
` GLP–1 production
`Access
`
`CNS: circumventricular organs (circulating GLP-1)
`Autonomic nervous system
`Afferent vagus (GLP-1 from GI tract)
`“Hepatoportal” region
`
`
`
`Stomach
` Gastric emptying decelerated
` Acid secretion ↓
`
`that enteral
`endocrine pancreas. The observation
`nutrition provided a more potent insulinotropic stimulus
`compared with isoglycaemic intravenous challenge led to
`the development of the incretin concept.1 The fi rst
`incretin to be identifi ed, glucose-dependent insulinotropic
`polypeptide (GIP), was purifi ed from porcine intestinal
`extracts and had weak eff ects on gastric acid secretion
`but more potent insulinotropic actions in human beings.2
`GIP is a 42-aminoacid hormone synthesised in duodenal
`and jejunal enteroendocrine K cells in the proximal small
`bowel.
`A second incretin hormone, glucagon-like peptide-1
`(GLP-1) was identifi ed after the cloning of the cDNAs
`and genes encoding proglucagon (fi gure 1). GLP-1 exists
`in
`two circulating equipotent molecular
`forms,
`GLP-1(7-37) and GLP-1(7-36)amide, although GLP-1(7-
`36)amide is more abundant in the circulation after eating.
`Most GLP-1 is made in enteroendocrine L cells in the
`distal ileum and colon, but plasma levels of GLP-1, like
`GIP, also increase within minutes of eating. Hence a
`combination of endocrine and neural signals probably
`promote the rapid stimulation of GLP-1 secretion well
`before digested food transits through the gut to directly
`engage the L cell in the small bowel and colon. More
`proximally located L cells in the duodenum and jejunum
`have also been described; however,
`the precise
`
`Search strategy and selection criteria
`
`We searched the MEDLINE and PubMed databases
`(1987–2006) with the search terms “glp-1”, “glucagon”,
`“glucagon-like”, “gip”, “incretin”, “dipeptidyl peptidase-4”,
`and “diabetes”. We preferentially selected publications from
`the past 5 years, but did not exclude older publications that
`are commonly referenced or highly regarded. We also
`searched the reference lists of articles identifi ed by this search
`strategy and selected those we judged relevant.
`
`Endocrine pancreas:
`Secretion
`β cells: insulin secretion ↑
`α cells: glucagon secretion ↓
`δ cells: somatostatin secretion ↑
`Biosynthesis
`
`(Pro-) insulin↑
`β-cell mass
`Growth, regeneration, neogenesis ↑
` Apoptosis ↓
`
`Ileum
`Site of GLP-1 synthesis from proglucagon,
`GLP-1 secretion ↑ after meals (carbohydrate, fat)
`
`Figure 1: Physiology of GLP-1 secretion and action on GLP-1 receptors in diff erent organs and tissues
`
`1696
`
`www.thelancet.com Vol 368 November 11, 2006
`
`MPI EXHIBIT 1024 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0001
`
`

`

`I
`
`New Drug Class
`
`GLP-1 (amidated form)
`
`HisAlaGluGlyThrPheThrSerAspValSerSerTyrLeuGluGlyGlnAlaAlaLysGluPheIleAlaTrpLeuValLysGlyArgamide
`7
`10
`15
`20
`25
`30
`35 36
`Proteolytic inactivation (DPP-4)
`
`Exenatide
`
`· N98e ·
`~
`HisGlyGluGlyThrPheThrSerAspLeuSerLysGlnMetGluGluGluAlaValArgLeuPheIle GluTrpLeuLysAsnGlyGlyProSerSerGlyAlaProProProSeramide
`~
`'
`
`Liraglutide
`
`HisAlaGluGlyThrPheThrSerAspValSerSerTyrLeuGluGlyGlnAlaAlaLysGluPheIle AlaTrpLeuValArgGlyArgGly
`Glu
`
`~
`
`C-16 free fatty acid
`
`Albumin
`
`Sitagliptin
`
`F
`
`NH₂
`
`O
`
`F
`
`O
`
`N
`
`OH
`
`NC
`
`HN
`
`Vildagliptin
`
`A~0 ~CY-{
`
`N C
`
`F₃
`
`N
`
`N
`
`N
`
`F
`
`Figure 2: Structure of GLP-1, GLP-1R agonists exenatide and liraglutide, and DPP-4 inhibitors vildagliptin and sitagliptin
`
`contributions of the proximal and distal L cells to the
`early rapid increase in plasma GLP-1 remains unclear.
`Plasma levels of GLP-1 are low in the fasted state, in the
`range of 5–10 pmol/L, and increase rapidly after eating,
`reaching 15–50 pmol/L. The circulating levels of intact
`GLP-1 and GIP decrease rapidly because of enzymatic
`inactivation, mainly dipeptidyl peptidase-4 (DPP-4), and
`renal clearance.3 Whether additional proteases, such as
`human neutral endopeptidase 24·11, are also essential
`determinants of GLP-1 inactivation is being investigated.
`Both GIP and GLP-1 contain alanine at position 2, and
`hence are excellent substrates for DPP-4. Indeed, DPP-4
`is essential for incretin inactivation, and mice with
`targeted inactivation of the DPP-4 gene have raised levels
`of plasma GIP and GLP-1, increased insulin secretion,
`and reduced glucose excursion after glycaemic challenge.4
`As a result of DPP-4 activity, intact, biologically active
`GLP-1 represents only 10–20% of total plasma GLP-1.5
`Both GIP and GLP-1 exert their actions by the
`engagement of structurally distinct G-protein-coupled
`receptors (GPCRs). The GIP receptor is predominantly
`expressed on islet β cells, and to a lesser extent, in adipose
`tissue and in the central nervous system. By contrast, the
`GLP-1 receptor (GLP-1R) is expressed in islet α and β
`cells and in peripheral tissues, including the central and
`peripheral nervous systems, heart, kidney, lung, and
`gastrointestinal tract (fi gure 1). Activation of both incretin
`
`receptors on β cells leads to rapid increases in levels of
`cAMP and intracellular calcium, followed by insulin
`exocytosis,
`in a glucose-dependent manner.6 More
`sustained incretin receptor signalling is associated with
`activation of protein kinase A, induction of gene
`transcription, enhanced levels of insulin biosynthesis,
`and stimulation of β-cell proliferation.7 Both GLP-1R and
`GIP receptor activation also promote resistance to
`apoptosis and enhanced β-cell survival, in both rodent8
`and human islets.9 Consistent with the distribution of
`GLP-1R expression, GLP-1 also
`inhibits glucagon
`secretion, gastric emptying, and food ingestion, and
`promotes enhanced glucose disposal through neural
`mechanisms,10 actions that also contribute to the control
`of glucoregulation. Notably, eff ects on glucagon secretion,
`like those on insulin secretory responses, are glucose-
`dependent, whereas counter-regulatory
`release of
`glucagon in response to hypoglycaemia is fully preserved
`even in the presence of pharmacological concentrations
`of GLP-1.11
`The physiological importance of endogenous GIP and
`GLP-1 for glucose homoeostasis has been investigated in
`studies with receptor antagonists, or gene-knockout
`mice. Acute antagonism of GIP or GLP-1 lowers insulin
`secretion and increases plasma glucose after glycaemic
`challenge in rodents. Similarly, mice with inactivating
`mutations in the GIP or GLP-1 receptors also have
`
`www.thelancet.com Vol 368 November 11, 2006
`
`1697
`
`MPI EXHIBIT 1024 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0002
`
`

`

`I
`
`New Drug Class
`
`insulin secretion and
`defective glucose-stimulated
`impaired glucose tolerance.12,13 GLP-1, but not GIP, is also
`essential for control of fasting glycaemia, since acute
`antagonism or genetic disruption of GLP-1 action leads
`to increased levels of fasting glucose in rodents.13
`Furthermore, GLP-1 is essential for glucose control in
`human beings: studies with the antagonist exendin(9-39)
`show defective glucose-stimulated insulin secretion,
`reduced glucose clearance, increased levels of glucagon,
`and quicker gastric emptying after disruption of GLP-1
`action.14
`The pleiotropic actions of GLP-1 and GIP on the control
`of blood glucose have fostered considerable interest in
`the use of these agents for the treatment of type 2
`
`diabetes. Whereas in healthy human beings oral glucose
`elicits a considerably higher insulin secretory response
`than does intravenous glucose (even if leading to the
`same glycaemic increments), this incretin eff ect is
`substantially reduced or even lost in patients with type 2
`diabetes.15 As an explanation for the acquired incretin
`defect, GIP but not GLP-1 shows noticeably attenuated
`insulinotropic action in patients with type 2 diabetes.16
`Furthermore, those with type 2 diabetes show a small but
`signifi cant reduction in meal-stimulated levels of GLP-1.17
`Since GLP-1 action remains relatively preserved in
`diabetic patients, most pharmaceutical eff orts directed at
`potentiation of incretin action for the treatment of type 2
`diabetes have focused on GLP-1R agonists.
`
`Biological actions of
`incretin hormone in
`type 2 diabetes
`
`Native GLP-1*
`
`Incretin mimetics
`
`DPP-4 inhibitors
`(eg, vildagliptin,
`sitagliptin)
`
`Exenatide
`
`Liraglutide
`
`Lack of biphasic response†
`
`Slow insulin secretory response to meals50
`
`Reduction in or absence of incretin eff ect15
`
`Hyperglucagonaemia52
`
`Hypoglycaemia counter-regulation
`
`Reduced pancreatic β-cell insulin content
`
`Reduced endocrine pancreatic β-cell mass56,57
`
`Characteristic features of type 2 diabetes
`Defective glucose-stimulated insulin secretion Glucose-dependent
`stimulation of insulin
`secretion
`Restoration of biphasic
`responses
`More adequate insulin
`secretory response after
`meals‡
`Replacement of incretin
`activity, greater incretin
`eff ect§
`Suppression of glucagon
`secretion
`Glucagon secretion, when
`plasma glucose is low
`Increased synthesis of
`proinsulin
`Increase in pancreatic islet
`β-cell mass
`Diff erentiation of islet
`precursor cells into β cells
`Inhibition of
`toxin-induced|| β-cell
`apoptosis
`Deceleration in gastric
`emptying
`Suppression of
`appetite/induction of
`satiety
`Weight loss (fi gure 3)
`
`Abnormally high rate of β-cell apoptosis57
`
`Normal, decelerated or accelerated67 gastric
`emptying
`Hypercaloric energy intake/obesity72
`
`Yes16
`
`Yes48
`
`Yes21,51
`
`Yes‡
`
`Yes16
`
`Yes11
`
`Yes55
`
`Yes58
`
`Yes62
`
`Yes8,63
`
`Yes68
`
`Yes73
`
`Yes22
`
`Yes45
`
`Yes49
`
`Yes45
`
`Yes‡
`
`Yes45
`
`Yes53
`
`Yes
`
`Yes59
`
`Yes
`
`Yes64
`
`Yes69
`
`Yes74
`
`Yes25-27
`
`Yes46
`
`Yes47
`
`Not tested
`
`Not tested
`
`Yes46
`
`Yes‡
`
`Yes46
`
`Yes54
`
`Yes
`
`Yes60
`
`Yes
`
`Yes65
`
`Yes46
`
`Yes75
`
`Yes32
`
`Yes47
`
`Not tested, but probable
`
`Yes47
`
`Not tested
`
`Yes
`
`Yes61
`
`Unknown
`
`Probable66
`
`Marginal70,71
`
`No obvious eff ect
`
`No weight change34
`
`Pharmacological characteristics
`Mode of administration
`
`Frequency of administration
`Predominant adverse event
`
`Intravenous,
`subcutaneous
`Continuous
`Nausea
`
`Subcutaneous
`
`Subcutaneous
`
`Orally
`
`Twice daily
`Nausea
`
`Once daily
`Nausea
`
`Once (or twice) daily
`None noted
`
`*GLP-1 exists as glycine-extended (7–37) and amidated form (7–36)amide, with both forms having similar properties; †Biphasic response is only seen under artifi cial
`conditions leading to rapid rise in glucose concentrations (glucose bolus injection or “squarewave stimulus” when starting hyperglycaemic clamp); ‡As judged by
`improvement (normalisation) of postprandial glucose excursions; §By defi nition, GLP-1 and incretin mimetics replace incretin activity; ¶These actions have only been
`reported from animal or in vitro (eg, islet) studies; methods to assess human β-cell mass in vivo are not available; ||Hydrogen peroxide, free fatty acids, or streptozotocin.
`
`Table: Type 2 diabetes and biological actions of GLP-1, incretin mimetics, and DPP-4 inhibitors
`
`1698
`
`www.thelancet.com Vol 368 November 11, 2006
`
`MPI EXHIBIT 1024 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0003
`
`

`

`I
`
`New Drug Class
`
`Antidiabetic actions of GLP-1
`of GLP-1
`Short-term
`intravenous
`infusions
`(1–1·2 pmol kg–¹ min–¹, leading to pharmacological
`plasma concentrations of total GLP-1 of 70–150 pmol/L,
`and of intact biologically active GLP-1 of 10–20 pmol/L)
`lowers blood glucose in patients with type 2 diabetes
`through a transient glucose-dependent stimulation of
`insulin and suppression of glucagon secretion and
`gastric emptying.18–21 A 6-week subcutaneous infusion of
`GLP-1 in patients with type 2 diabetes, achieving plasma
`levels of GLP-1 in the 60–70 pmol/L range,22 produced
`substantial improvements in insulin secretory capacity,
`insulin sensitivity, a reduction in HbA1c of 1·2% and
`modest weight loss (1·9 kg).22 Although intravenous or
`subcutaneous GLP-1 infusions could be useful for the
`short-term control of hyperglycaemia,23,24 the long-term
`treatment of type 2 diabetes needs a more feasible
`approach to achieve sustained activation of GLP-1
`receptors. The effi cacy of injectable GLP-1 receptor
`agonists (degradation-resistant peptides or larger pro-
`teins with more suitable pharmacokinetic properties,
`fi gure 2) and DPP-4 inhibitors (small molecules with
`good oral bioavailability, webtable),25–42 has been assessed
`in clinical trials.
`
`GLP-1R agonists
`Exenatide
`Exenatide (synthetic exendin-4) was discovered in the
`search for biologically active peptides in lizard venom.43
`Exendin-4 shares roughly 50% of
`its aminoacid
`sequence with mammalian GLP-1 , is encoded by a
`unique gene in the lizard,44 and is a potent degradation-
`resistant agonist at the mammalian GLP-1R (fi gure 2).
`Exenatide has been developed for the treatment of
`type 2 diabetes (table).47–75 Exenatide has a circulating
`half-life of 60–90 min,69 with increases in plasma
`exenatide concentrations lasting 4–6 h after a single
`subcutaneous injection.76,77
`Phase III trials investigated the effi cacy of adding
`exenatide (5 or 10 μg by subcutaneous injection twice
`daily) to ongoing therapy in patients suboptimally
`controlled on oral antidiabetic agents (metformin,25
`sulphonylureas,26 a combination of both,27 or thiazo-
`lidinediones28). The starting dose of exenatide is 5 μg
`twice daily for 4 weeks, followed by an increase to 10 μg
`twice daily.78,79 Exenatide reduced HbA1c concentrations
`by 0·8–1·0 % (fi gure 3)25–42 over 30 weeks, with prevention
`of weight gain or modest weight loss of 1·5–3 kg.
`Patients continuing in an open-label extension lost more
`weight, with the total weight loss reaching 4–5 kg after
`80 weeks.80 The commonest adverse events with
`exenatide were gastrointestinal (nausea, or more rarely
`vomiting or diarrhoea25–27) (fi gure 3). However, exenatide
`was rarely discontinued because of side-eff ects, and the
`occurrence of nausea lessened the longer the duration
`of therapy.25–27 An increased number of mild to moderate
`hypoglycaemic events was noted in patients given
`
`exenatide and sulphonylureas,26,27 but not in those given
`exenatide and metformin,25 despite a similar reduction
`in glycaemia.
`40–50% of patients receiving exenatide develop
`antibodies with weak binding affi nity and low titres.25–27
`Antibody formation has not been associated with
`impaired antidiabetic eff ectiveness of exenatide in most
`of those treated. However, the drug might not be as
`eff ective in the few patients with high-titre antibodies.
`Exenatide has been compared with insulin glargine in
`an open-label study as additional treatment for diabetic
`patients not achieving eff ective glucose control on
`metformin and a sulphonylurea.29 Fasting glucose
`concentrations were reduced more in patients receiving
`insulin glargine, but postprandial glucose reduction was
`greater with exenatide, especially after breakfast and
`dinner. Both exenatide and insulin glargine reduced
`levels of HbA1c by 1·1% over 26 weeks.29 No signifi cant
`diff erences in overall rates of hypoglycaemia were seen
`in the diff erent treatment groups, although nocturnal
`hypoglycaemia was less frequent with exenatide and
`daytime hypoglycemia was less common in patients
`given insulin glargine. Gastrointestinal side-eff ects, such
`as nausea and vomiting, were more often reported with
`exenatide than with insulin glargine, and the dropout
`rate was also higher in the exenatide-treated cohort.
`However, patients receiving insulin glargine gained an
`average of 1·8 kg compared with a 2·3 kg weight loss in
`exenatide-treated patients.29 Exenatide was approved by
`the US Food and Drug Administration for the treatment
`of type 2 diabetes in April, 2005. In Europe, exenatide is
`expected to be approved by the end of 2006 or early 2007
`for use in patients with type 2 diabetes that is not well
`controlled on oral agents.
`
`Liraglutide
`Liraglutide, a partly DPP-4-resistant GLP-1 analogue,
`contains a Arg34Lys substitution, and a glutamic acid
`and 16-C free-fatty-acid addition to Lys26 (fi gure 2).81 The
`acyl moiety promotes non-covalent binding to albumin
`with 1–2% of liraglutide circulating as the non-albumin-
`bound free peptide.82 Liraglutide has a half-life of about
`10–14 h after subcutaneous administration in human
`beings,83,84 and can be given as a once daily injection.
`Early phase II studies were done with up to 0·75 mg per
`day of liraglutide,31,85 but more recent studies with weekly
`escalating dose-titration have investigated the effi cacy of
`doses up to 2·0 mg.32 Liraglutide reduces fasting and
`postprandial glucose, and levels of HbA1c by up to 1·75 %
`(fi gure 3),33 while preventing weight gain or inducing
`modest but signifi cant weight loss.32,33 Nausea, vomiting,
`and diarrhoea were the most prominent adverse events
`but were generally mild, transient, and rarely caused
`discontinuation of liraglutide treatment.32,33 So far, no
`studies of exposure to liraglutide have reported antibody
`formation, and phase III testing was started earlier this
`year.
`
`See Online for webtable
`
`www.thelancet.com Vol 368 November 11, 2006
`
`1699
`
`MPI EXHIBIT 1024 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0004
`
`

`

`-
`
`New Drug Class
`
`I I □□
`
`Exenatide (twice daily)
`Exenatide LAR (once weekly)
`Liraglutide (once daily)
`Placebo
`
`□□□
`
`Insulin glargine
`Vildagliptin
`Sitagliptin
`
`D I □
`
`E
`
`D
`
`C
`
`B
`
`A
`
`Nausea (%)
`
`Hypoglycaemia (%)
`
`Change in bodyweight (kg)
`
`Change in fasting glucose (mg/dL)
`
`HbA 1c (%)
`
`1·0
`
`0·5
`
`0·0
`
`–0·5
`
`–1·0
`
`–1·5
`–2·0
`
`20
`
`0
`
`–20
`
`–40
`
`–60
`
`–80
`
`0
`
`321
`
`–1
`–2
`–3
`–4
`–5
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`
`0
`
`Incretin mimetics
`
`Up to 10 μg
`twice daily
`
`Up to 2 mg
`
`weekly
`
`Up to
`
`Duration (weeks):
`
`30
`
`15 26 15 12
`
`5
`
`Diet ± O
`
`Metformin
`
`AD
`
`Figure 3: Clinical eff ects of
`GLP-1R agonists (incretin
`mimetics) and DPP-4
`inhibitors (incretin
`enhancers) on HbA1c, fasting
`glucose concentrations,
`bodyweight, hypoglycaemic
`episodes, and nausea
`Doses are indicated in top
`panels (A); concomitant
`medication is shown in lower
`panel (D). Results are from
`phase II or III studies on:
`exenatide;25–29 exenatide LAR;30
`liraglutide;31-33 vildagliptin;34-38
`and sitagliptin.39-42 *Signifi cant
`diff erences to placebo or
`respective comparator; if no
`comparator is shown, results
`are depicted as placebo-
`subtracted diff erences. Bars are
`mean and SE. OAD=oral
`antidiabetic agents.
`
`1700
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`
`0
`
`Zin
`
`DeFronzo et al, 2005
`Kendall et al, 2005
`Heine et al, 2005
`Buse et al, 2004
`Nauck et al, 2006
`man et al, 2006
`Madsbad et al, 2004
`
`Ki
`
`m et al, 2
`
`006
`
`Vilsb
`Øll et al, 2
`
`Metformin and/or sulfonylurea
`Metformin+sulfonylurea
`TZD (+metformin)
`Sulfonylurea
`Metformin
`
`Diet or O
`
`AD
`
`(washed out)
`
`006
`
`Åhren et al, 2005
`
`1·0
`
`0·5
`
`0·0
`
`–0·5
`
`–1·0
`
`–1·5
`–2·0
`
`20
`
`0
`
`–20
`
`–40
`
`–60
`
`–80
`
`0
`
`321
`
`–1
`–2
`–3
`–4
`–5
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`
`0
`
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`
`0
`
`Metformin
`Glimepiride
`Rosiglitazone
`
` 0·75, 2·0, 1·9 mg
`
`once daily
`
`14
`
`DPP-4 inhibitors
`
`50 mg
`
`100 mg
`
`25 mg Up to 100 mg
`
`Up to 200 mg
`
`100 mg
`
`Duration (weeks):
`
`12 52 12 52
`
`24
`
`18
`
`24
`
`Metformin
`
`Dru
`g-n
`aiv
`e p
`atie
`nts
`
`Metformin
`
`Diet or O
`
`(washed out)
`Metformin
`
`Pio
`glitaz
`
`one
`
`AD
`
`(1
`2 w
`(5
`2 w
`
`eeks)
`
`Pratle
`y et al, 2
`
`eeks)
`
`Karasik et al, 2006
`Rosenstock et al, 2006
`Aschner et al, 2006
`Raz et al, 2006
`Garber et al, 2006
`Rosenstock et al, 2006
`Dejager et al, 2006
`
`004
`
`www.thelancet.com Vol 368 November 11, 2006
`
`MPI EXHIBIT 1024 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0005
`
`

`

`I
`
`New Drug Class
`
`Long-acting GLP-1R agonists
`Because one subcutaneous injection of exenatide does
`not produce eff ective glucose control for more than
`6–8 h, there is considerable interest in the development
`of long-acting GLP-1R agonists that need less frequent
`parenteral administration. Exenatide long-acting release
`(LAR) is a polylactide-glycolide microsphere suspension
`containing 3% exendin-4 peptide that shows sustained
`dose-dependent glycaemic control in diabetic fatty
`Zucker rats for up to 28 days after one subcutaneous
`injection.86 Preliminary experience with exenatide LAR
`in 45 patients with type 2 diabetes indicates a much
`greater reduction in fasting glucose concentrations and
`HbA1c after once weekly administrations of exenatide
`LAR for 15 weeks compared with exenatide twice daily.30
`However, long-term experience with the drug in larger
`numbers of patients has not yet been reported. Exenatide
`LAR is currently being assessed in a phase III head-to-
`head trial against twice-daily exenatide.
`Additional strategies for development of long-acting
`GLP-1R agonists include the use of chemical linkers to
`form covalent bonds between GLP-1 (CJC-1131) or
`exendin-4 (CJC-1134).87 Similarly, recombinant albumin-
`GLP-1 proteins have been developed that mimic the full
`range of GLP-1 actions in preclinical studies.88 Although
`these drugs are expected
`to have an extended
`pharmacokinetic profi le suitable for once weekly dosing
`in diabetic patients, little clinical information is available
`about the effi cacy and safety of these albumin-based drugs
`in human beings.
`
`DPP-4 inhibitors
`The observation that GLP-1 is rapidly degraded by
`DPP-45,89,90 has fostered the development of specifi c
`protease inhibitors that prevent the rapid fall of GLP-1 in
`circulating plasma after eating. DPP-4 is a ubiquitous
`membrane-spanning cell-surface aminopeptidase widely
`expressed in many tissues, such as liver, lung, kidney,
`intestinal brush-border membranes, lymphocytes, and
`endothelial cells.91–93 The extracellular domain of DPP-4
`can also be cleaved from its membrane-anchored form
`and circulate in plasma, where it retains its full enzymatic
`activity. DPP-4 preferentially cleaves peptides with a
`proline or alanine residue in the second aminoterminal
`position. Many gastrointestinal hormones, neuropeptides,
`cytokines, and chemokines are substrates for DPP-4,91,93
`among them both GIP89,90,94 and GLP-1.5,89,90,95 In preclinical
`studies, DPP-4 inhibitors mimic many of the actions
`ascribed to GLP-1R agonists, including stimulation of
`insulin and
`inhibition of glucagon secretion, and
`preservation of β-cell mass through stimulation of cell
`proliferation and inhibition of apoptosis.7,96 By contrast,
`DPP-4 inhibitors are generally not associated with a
`deceleration of gastric emptying or weight loss, perhaps
`due to the modest stabilisation of postprandial levels of
`intact biologically active plasma GLP-1 (doubled to
`15–25 pmol/L) seen after DPP-4 inhibition (table).
`
`Many small-molecule DPP-4 inhibitors have been
`developed that specifi cally and potently inhibit DPP-4
`activity after oral administration. Typically, these agents
`reduce serum DPP-4 activity by more than 80%, with
`some inhibition maintained for 24 h after one dose or with
`once daily treatment.47,97 DPP-4 inhibition is accompanied
`by a rise in postprandial levels of intact GLP-1.47,97,98 Most
`published studies used vildagliptin.99
`
`Vildagliptin and sitagliptin
`At a dose of 100 mg once daily, fasting and postprandial
`glucose concentrations were reduced after 4 weeks of
`vildagliptin treatment.47 Plasma glucagon concentrations
`were suppressed after vildagliptin treatment, together
`with an increase in the ratio of insulin to glucose.47 In
`clinical studies of longer duration, the addition of
`vildagliptin to patients already given metformin reduced
`HbA1c by 0·8% after 12 weeks, compared with placebo,34
`and this diff erence was maintained during an open-label
`extension for 52 weeks34 (fi gure 3). Indirect evidence
`from modelling experiments suggests that β-cell function
`is improved with vildagliptin treatment over 1 year in
`patients with type 2 diabetes.100 Preliminary reports of
`longer phase III clinical studies with vildagliptin
`monotherapy, either 50 mg twice daily or 100 mg once
`daily, showed sustained effi cacy but slight non-inferiority
`compared with metformin36 after 1 year of therapy,
`although vildagliptin was better tolerated than metformin.
`Similarly, vildagliptin was as eff ective as rosiglitazone in
`direct comparison monotherapy study37 and also produced
`signifi cant reductions in HbA1c when used in combination
`with metformin38 (fi gure 3).
`Clinical studies have also been reported for sitagliptin97
`(fi gure 3). Phase III clinical trial data presented at the
`American Diabetes Association meeting in June, 2006,
`indicated that sitagliptin is well-tolerated at doses of
`100 mg once daily, either as monotherapy, or in
`combination with metformin or pioglitazone, without
`signifi cant hypoglycaemia or weight gain.40–42 Fewer data
`are available for other DPP-4 inhibitors in development
`such as saxagliptin101 or denagliptin.102 Thus whether
`various chemically distinct DPP-4 inhibitors will show
`signifi cant diff erences in pharmacokinetic profi les, side-
`eff ects, or clinical activity cannot be predicted. Sitagliptin
`was approved for the treatment of type 2 diabetes in the
`USA in October, 2006.
`No characteristic pattern of adverse events has been
`associated with the use of vildagliptin34,47 or other DPP-4
`inhibitors,103-105 despite the large number of potential
`substrates for DPP-4.91,93 In view of the widespread
`expression of DPP-4 on many cell types, including
`lymphocytes, there is considerable interest in the long-
`term safety profi le of DPP-4 inhibitors. Although highly
`selective DPP-4 inhibition seems to be well tolerated in
`preclinical studies and DPP-4
`inhibitors do not
`substantially inhibit cell proliferation in experiments
`with human
`lymphocytes
`in vitro,106 considerable
`
`www.thelancet.com Vol 368 November 11, 2006
`
`1701
`
`MPI EXHIBIT 1024 PAGE 6
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1024-0006
`
`

`

`I
`
`New Drug Class
`
`additional clinical experience with these agents will be
`needed before any theoretical safety concerns emerge.
`Furthermore, given the large number of chemically
`distinct DPP-4 inhibitors under clinical development, it
`seems likely that one or more of these agents could be
`associated with adverse events arising as a result of unique
`properties attributable to the individual chemical structure,
`as opposed to a class eff ect arising as a consequence of
`inhibition of DPP-4 activity. Non-selectivity for actions on
`the related enzymes DPP-8, DPP-9, or both, could be of
`particular importance.106
`
`Contrasting properties of GLP-1R agonists and
`DPP-4 inhibitors
`Twice daily exenatide through subcutaneous injection is
`indicated for the treatment of patients with type 2 diabetes
`mellitus in whom one or more oral agents do not work,
`often as an alternative to insulin treatment. By contrast,
`once daily DPP-4 inhibitors could be used as fi rst-line
`therapy, or as add-on therapy to patients failing one or
`more oral agents. While there does not seem to be a great
`diff erence in the HbA1c-lowering capacity of GLP-1R
`agonists compared with DPP-4 inhibitors, the obvious
`diff erence between these classes of drugs is their eff ect on
`bodyweight. Weight loss is a common outcome of therapy
`with native GLP-1,22 exenatide,25–27 and liraglutide,32,75
`whereas treatment with DPP-4 inhibitors is associated
`with prevention of weight gain34,47,103,104 (fi gure 3). By
`contrast, gastrointestinal side-eff ects, predominantly
`nausea, are often reported after treatment with injectable
`GLP-1R agonists but have not been described with DPP-4
`inhibition.34,47,102–105 These diff erences might be explained in
`part by the relatively modest stabilisation of postprandial
`GLP-1 seen after DPP-4 inhibition, compared with the
`pharmacological increases in circulating levels of GLP-1R
`agonists exemplifi ed by exenatide. Hence therapy with
`DPP-4 inhibitors might not be associated with weight loss
`perhaps partly because of the relative levels of GLP-1
`achieved after treatment with these agents. Although
`nausea is a common side-eff ect of exenatide therapy, many
`patients lose weight independently of nausea. Consistent
`with the above diff erences in circulating levels of GLP-1,
`GLP-1R agonists, but not DPP-4 inhibitors, greatly
`decelerate gastric emptying.69,107,108
`
`Future developments
`Liraglutide and exenatide are fi rst-generation GLP-1
`receptor agonists, requiring once or twice daily parenteral
`administration, respectively. Much eff ort continues to be
`directed towards improvement of the pharmacokinetic
`profi le of GLP-1R agonists, to minimise peak levels of the
`drug and thus reduce the extent of nausea. Longer-acting
`GLP-1R agonists should ideally provide more uniform and
`sustained GLP-1R activation over a 24-h period, but require
`less frequent administration.
`Furthermore, there is great interest in determining
`whether chronic therapy with GLP-1R agonists will be
`
`associated with sustained long-term control of HbA1c and
`improvement in β-cell function beyond that achievable
`with existing agents. Similar questions pertain to the
`DPP-4 inhibitors, which also indirectly target β cells;
`however, long-term clinical data assessing the durability
`and effi cacy of these agents in the treatment of type 2
`diabetes are not yet available. Because patients with
`type 2 diabetes have increased risks of cardiovascular
`morbidity and mortality, the observation that GLP-1R
`agonists improve myocardial function in human patients
`after myocardial infarction109 highlights the need for
`studies that assess cardiovascular endpoints in patients
`treated with DPP-4 inhibitors or GLP-1R agonists.
`Overall, agents that enhance incretin action show great
`promise for the treatment of type 2 diabetes by
`recruitment of new, often physiologically based
`mechanisms of action for glucoregulation, in the context
`of a currently favourable safety profi le. Never theless,
`long-term clinical studies are needed to compare these
`agents with existing oral therapies or insulin, or both, to
`permit a greater understanding of the true benefi ts and
`role of these drugs for the treatment of diabetes
`mellitus.
`Confl ict of interest statement
`D J Drucker is an inventor or co-inventor on patents related to the fi eld
`of type 2 diabetes that are licensed to Amylin Pharmaceuticals Inc or
`Arisaph Pharmaceuticals Inc. He has served as a consultant or adviser
`within the past 12 months to Abbott Laboratories, Amgen Inc, Amylin
`Pharmaceuticals, Bayer Inc, Chugai Inc, Conjuchem Inc, Eli Lilly Inc,
`GlaxoSmithKline, Glenmark Inc, Johnson &

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