throbber
I
`
`review article
`
`Diabetes, Obesity and Metabolism 14 (Suppl. 2): 20–32, 2012.
`© 2012 Blackwell Publishing Ltd
`
`Comparison of liraglutide versus other incretin-related
`anti-hyperglycaemic agents
`L. Blonde1 & E. Montanya2
`1OchsnerDiabetesClinicalResearchUnit,DepartmentofEndocrinology,DiabetesandMetabolism,OchsnerMedicalCenter,NewOrleans,LA,USA
`2HospitalUniversitariBellvitge-IDIBELL,UniversityofBarcelona,CIBERdeDiabetesyEnfermedadesMetab´olicasAsociadas(CIBERDEM),Barcelona,Spain
`
`The two classes of incretin-related therapies, dipeptidyl peptidase-4 (DPP-4) inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1
`RAs), have become important treatment options for patients with type 2 diabetes. Sitagliptin, saxagliptin, vildagliptin and linagliptin, the
`available DPP-4 inhibitors, are oral medications, whereas the GLP-1 RAs—twice-daily exenatide, once-weekly exenatide and once-daily
`liraglutide—are administered subcutaneously. By influencing levels of GLP-1 receptor stimulation, these medications lower plasma glucose
`levels in a glucose-dependent manner with low risk of hypoglycaemia, affecting postprandial plasma glucose more than most other anti-
`hyperglycaemic medications. Use of GLP-1 RAs has been shown to result in greater glycaemic improvements than DPP-4 inhibitors, probably
`because of higher levels of GLP-1 receptor activation. GLP-1 RAs can also produce significant weight loss and may reduce blood pressure and
`have beneficial effects on other cardiovascular risk factors. Although both classes are well tolerated, DPP-4 inhibitors may be associated with
`infections and headaches, whereas GLP-1 RAs are often associated with gastrointestinal disorders, primarily nausea. Pancreatitis has been
`reported with both DPP-4 inhibitors and GLP-1 RAs, but a causal relationship between use of incretin-based therapies and pancreatitis has not
`been established. In clinical trials, liraglutide has shown efficacy and tolerability and resulted in certain significant benefits when compared with
`exenatide and sitagliptin.
`Keywords: DPP-4 inhibitor, GLP-1, linagliptin, liraglutide, saxagliptin, sitagliptin, vildagliptin
`
`Date submitted 1 January 2012; date of final acceptance 11 January 2012
`
`article
`review
`
`Introduction
`Incretin-related therapies have become established as impor-
`tant treatment options for patients with type 2 diabetes
`(T2D) [1] and the number of available options will increase in
`the near future. Knowledge about how the current incretin-
`related anti-hyperglycaemic therapies compare to each other
`can help health care professionals make informed treatment
`choices for individual patients.
`Patients with T2D have an impaired incretin effect, which
`appears to be the result of reductions in the insulinotropic
`and glucagon-suppressive actions of
`the incretin hor-
`mones glucagon-like peptide (GLP-1) and glucose-dependent
`insulinotropic peptide (GIP) [2,3], although declines in the
`release of these hormones have also been reported among
`patients with T2D [2,4]. When these patients receive infusions
`of GLP-1 to supraphysiological levels, the insulin secretory
`response improves, glucagon secretion is suppressed and
`plasma glucose levels can be significantly improved. These
`effects occur in a glucose-dependent manner, that is, only
`when glucose levels are elevated, resulting in a low risk of
`hypoglycaemia [2,5,6].
`
`Correspondenceto: Dr Lawrence Blonde, Ochsner Diabetes Clinical Research Unit, 1514
`Jefferson Highway, New Orleans, LA 70121, USA.
`E-mail: lawrence.blonde@gmail.com
`
`Re-use of this article is permitted in accordance with the Terms and Conditions set out at
`http://wileyonlinelibrary.com/onlineopen#OnlineOpen_Terms
`
`In addition to improving glucose control, raising GLP-1
`levels can provide additional benefits such as slowed gastric
`emptying, decreased acid secretion, increased feeling of satiety
`and reduced energy intake [5,7]. In humans, endothelial
`dysfunction, cardiovascular function and β-cell function also
`appear to improve with a GLP-1 infusion [8–11], while animal
`studies suggest that GLP-1 can stimulate expansion of β-cell
`mass [12], and reduce high blood pressure [13].
`As native GLP-1 is degraded rapidly by the enzyme dipeptidyl
`peptidase-4 (DPP-4), resulting in a half-life of approximately
`2 min following intravenous administration [14], its therapeu-
`tic use is impractical. Two strategies have been employed to
`produce incretin-related therapies. One approach is to inhibit
`the DPP-4 enzyme, resulting in an extended half-life and
`an increase in circulating endogenous GLP-1 and GIP [3].
`The other approach involves the use of agents resistant to the
`breakdown of DPP-4 that bind to and activate the GLP-1 recep-
`tor, thus producing glucoregulatory effects similar to those of
`GLP-1. This article discusses the clinical profiles and compares
`the available agents in these two classes.
`
`Pharmacological Differences Between
`the Incretin-Related Therapies
`The GLP-1 receptor agonists (GLP-1 RAs) exenatide, exenatide
`once weekly (currently only approved in Europe) and
`liraglutide are peptides, and so they must be given by
`
`MPI EXHIBIT 1058 PAGE 1
`
`MPI EXHIBIT 1058 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0001
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`subcutaneous injection: twice daily (BID) for exenatide, within
`60 min before the two main meals and at least 6 h apart; once
`a week on the same day for exenatide once weekly, with or
`without meals; and once a day for liraglutide, independent
`of meals. Although both exenatide and liraglutide are GLP-1
`RAs, exenatide is a mimetic, discovered in the saliva of the
`Gila monster (Heloderma suspectum), with 53% amino acid
`sequence identity to the chemical structure of native GLP-
`1, while liraglutide is an analogue of human GLP-1 with
`97% sequence identity [15,16]. Exenatide once weekly is a
`long-acting formulation of exenatide in which exenatide is
`encapsulated in microspheres of poly(d,l lactic-co-glycolic
`acid) for gradual drug delivery [17]. Liraglutide differs from
`human GLP-1 by the attachment of a palmitic acid via a
`glutamic acid spacer to lysine at position 26 and by the
`replacement of lysine at position 34 with arginine [18]. It
`is thought that the addition of a fatty acid chain to liraglutide’s
`structure allows it to form heptamers when injected, which
`delay its absorption and binding to albumin, increasing its
`resistance to DPP-4 degradation and allowing the maximum
`concentration to be reached at 8–12 h after dosing [19,20].
`In contrast, exenatide reaches its median peak concentration
`in 2 h [21]. Exenatide once weekly takes much longer than
`either liraglutide or exenatide BID to reach maximum
`concentrations. After 2 weeks of administering exenatide
`once weekly, serum concentrations exceed minimal efficacy
`levels and continue to increase over the next 4–5 weeks if
`treatment is maintained [22]. Clinical studies have confirmed
`that administration of exenatide and liraglutide results in dose-
`dependent decreases in hyperglycaemia through insulinotropic
`activity and suppression of glucagon secretion, both occurring
`in a glucose-dependent manner [23–25]. It is probable that
`there is a greater degree of GLP-1 receptor stimulation with
`GLP-1 RAs than that resulting from the two- to threefold
`increase in GLP-1 levels with DPP-4 inhibitors [3,26].
`DPP-4 inhibitors are small molecule oral medications. There
`are presently three available in the USA: sitagliptin (also
`produced in a single pill combination tablet with metformin),
`saxagliptin (also produced in a single pill combination tablet
`with metformin extended release) and linagliptin. All are
`administered once a day at any time, except for the sitagliptin
`combination tablet with metformin, which should be taken
`BID with meals. In Europe, another twice-daily DPP-4
`inhibitor can be prescribed: vildagliptin (also produced in
`a single pill combination tablet with metformin). Sitagliptin
`is a phenethylamine type of DPP-4 inhibitor, saxagliptin
`and vildagliptin are cyanopyrrolidines [27] and linagliptin is
`xanthine-derived [28]. As saxagliptin has been observed in
`the laboratory to have strong interactions with the DPP-4
`residues Ser630, Glu205 and Glu206, which are essential to the
`enzyme’s catalytic activity, its potency for inhibiting DPP-4
`activity is considered more robust than that of both sitagliptin
`and vildagliptin [29]. However, linagliptin has demonstrated
`more potent inhibition of DPP-4 when compared to the three
`other DPP-4 inhibitors under identical in vitro conditions [28].
`Despite the variable selectivity of the currently approved DPP-4
`inhibitors, it is not clear that there is much clinical difference
`between them. When the recommended dosages for each agent
`
`review article
`
`are administered, the median time to maximum concentration
`(Tmax) ranges from 1.7 h for vildagliptin, 2 h for saxagliptin,
`1–4 h for sitagliptin and 1.5 h for linagliptin [30–33]. The
`duration of DPP-4 inhibition is claimed to be 24 h with each
`agent; however, the terminal half-life (t1/2) for saxagliptin is
`2.5 h, vildagliptin 3 h and sitagliptin 12.4 h [30–32]. Although
`the t1/2 for linagliptin is more than 100 h, the effective half-life
`is approximately 12 h [33]. Only saxagliptin appears to have a
`pharmacologically active metabolite, 5-hydroxy saxagliptin,
`which has a t1/2 of 3.1 h [31]. Following an oral glucose
`load or meal in patients receiving DPP-4 inhibitors, there
`is an increase in the circulating levels of GLP-1, a reduction
`in glucagon concentration and an enhancement of glucose-
`dependent insulin secretion [30–33].
`In the USA, all FDA-approved incretin-related therapies
`can be used as monotherapy (although liraglutide is not rec-
`ommended as first-line therapy) [33–37], whereas in Europe,
`only sitagliptin and linagliptin are approved as monother-
`apy [32,33]. In the USA, all incretin therapies can also be used in
`dual- or triple-combination therapy with metformin, sulpho-
`nylureas (SUs) and/or thiazolidinediones (TZDs) [33–37].
`However, the approved combined uses for these medications
`are slightly more restrictive in Europe, where liraglutide is
`approved for use in dual combination with metformin or SUs,
`and exenatide, exenatide once weekly, saxagliptin, sitagliptin
`and vildagliptin can be used in dual combination with met-
`formin, SUs or TZDs [20–22,30–32]. Exenatide, exenatide
`once weekly, liraglutide and sitagliptin are the only medica-
`tions approved for triple combination with metformin and
`a SU or metformin and a TZD in Europe [20–22,32]. In
`both the USA and Europe, sitagliptin is approved for use
`with insulin [32,34]. The efficacy and safety of exenatide and
`liraglutide in conjunction with insulin have been studied, but
`only limited approval has been obtained for any combined use
`[38,39]. In one 52-week trial, the addition of insulin detemir to
`liraglutide 1.8 mg and metformin in patients not achieving gly-
`caemic targets led to decreases in HbA1c, sustained weight loss
`and a small increase in minor hypoglycaemic events [40,41]. In
`Europe, insulin detemir may be used as add-on therapy with
`liraglutide [41]. The addition of liraglutide in patients already
`treated with insulin has not been evaluated. The addition of
`exenatide following insulin optimisation led to reduction in
`HbA1c, modest weight decrease, and no change in hypogly-
`caemic rates [38], and in the USA, exenatide may be added
`on to therapy with insulin glargine [37]. No clinical data exist
`regarding the combination of GLP-1 RAs with DPP-4 inhibitors
`in treatment, and it is not currently recommended, although
`data in minipigs have suggested pharmacokinetics are unlikely
`to be altered by the combination [42].
`As expected from the glucose-dependent pharmacoki-
`netic/pharmacodynamic profiles of incretin-related therapies,
`these treatments lead to improvements in controlling postpran-
`dial glucose (PPG) excursions [43,44]. Twice-daily exenatide
`is dosed to peak with PPG concentrations, unlike the other
`incretin-related therapies, which can be administered without
`regard for meals, and provides control of postprandial excur-
`sions. In head-to-head trials, exenatide taken BID lowered PPG
`to a greater degree than did sitagliptin and liraglutide [15,45].
`
`Volume 14 No. (Suppl. 2) April 2012
`
`doi:10.1111/j.1463-1326.2012.01575.x 21
`
`I
`
`MPI EXHIBIT 1058 PAGE 2
`
`MPI EXHIBIT 1058 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0002
`
`

`

`review article
`
`Improvements in HbA1c demonstrated by incretin-related
`therapies when added to metformin are shown in Table 1.
`
`Effect on Gastric Emptying and Weight Loss
`Patients with T2D often have accelerated gastric emptying,
`which may contribute to postprandial hyperglycaemia [51,52].
`Native GLP-1 can slow accelerated emptying of the stomach,
`and slow acid secretion, contributing to its effectiveness at
`lowering postprandial hyperglycaemia [5,7]. Clinical studies
`have shown that GLP-1 RAs produce the same effect as
`native GLP-1 [23,25], while DPP-4 inhibitors do not [3,53].
`The slowed gastric emptying observed with GLP-1 RAs
`may contribute
`to the most
`common gastrointestinal
`adverse event
`reported for
`these therapies
`in clinical
`trials—nausea [15,26,43,54–58]. Gastrointestinal problems
`are infrequent with DPP-4 inhibitors [43].
`In clinical studies, GLP-1 RAs have been associated with
`dose-dependent weight loss [49,59], which has generally not
`been seen with DPP-4 inhibitors as the latter appear to be
`weight neutral [60,61]. The higher levels of GLP-1 receptor
`stimulation achieved with GLP-1 RAs compared to DPP-4
`inhibitors are probably the most important factor responsible
`for the difference in weight effect observed between the
`two kinds of incretin-related therapies. The weight loss is
`not primarily related to gastrointestinal symptoms such as
`nausea, as many patients using GLP-1 RAs lose weight
`without experiencing any nausea, and the nausea is typically
`transient [3]. Preclinical and clinical studies have shown that
`these agents can dose-dependently increase the feeling of satiety,
`reduce meal size and lower energy intake in a manner similar to
`that of native GLP-1 [7,62–65]. The slowed gastric emptying
`by GLP-1 RAs may also contribute to increased satiety, reduced
`food intake and the resultant clinically significant weight loss
`shown in many studies with these agents [3,15,49,55–59].
`Another possible explanation for the lack of DPP-4 inhibitor
`effect on weight may be that they inhibit cleavage of the gut
`hormone peptide YY (PYY). Thus, levels of intact PYY1-36,
`which stimulates food intake, may be increased while levels
`of the active form PYY3-36, which reduces food intake, may
`be reduced [66]. In a recent 12-week, randomised, placebo-
`controlled clinical study, sitagliptin decreased PYY3-36 while
`increasing intact PYY1-36. The depression of PYY3-36 levels
`with DPP-4 inhibitor treatment may thus contribute to the
`difference in weight response between the two classes of incretin
`therapies [67]. Lastly, in animal studies, GIP has been linked
`with obesity through over-nutrition [68]. Although T2D is a
`GIP-resistant state [2], DPP-4 inhibitors raise GIP, as well as
`GLP-1 levels, by blocking the activity of the DPP-4 enzyme.
`This effect might also have a role in the weight neutrality (rather
`than weight loss) that is seen with DPP-4 inhibitor therapy.
`
`Blood Pressure, Lipids and Other
`Cardiovascular Risk Factors
`GLP-1 RAs have been shown to potentially improve multiple
`cardiovascular risk factors, but the mechanisms for these
`additional benefits are not yet clear. Reductions in systolic
`
`DIABETES, OBESITY AND METABOLISM
`
`blood pressure (SBP) that range from 2 to 7 mmHg over 26
`weeks [15,54–58] have been shown to precede any significant
`weight loss [69,70]. Nevertheless, weight loss due to GLP-1 RAs
`may be responsible for some of the observed improvements
`in blood pressure and lipids. After 3.5 years of exenatide
`twice-daily treatment in an open-label study, the quarter of
`patients who experienced the largest mean weight loss (12.8 kg)
`also had the greatest mean changes in SBP (−8.1 mmHg),
`high-density lipoprotein cholesterol (HDL-C) (+10.6 mg/dl)
`and triglycerides (−104.2 mg/dl) [71], despite a minimal
`correlation in the overall results between weight loss and lipid
`changes [71]. Further research is necessary to determine the
`actual mechanism for the improvements in blood pressure and
`the modest but significant reductions in triglycerides, free fatty
`acids and low-density lipoprotein cholesterol (LDL-C) levels
`that result with GLP-1 RA treatments [54,72]. Furthermore, a
`significant increase in HDL-C has also been observed in some
`studies [71,73]. In addition, liraglutide treatment has been
`associated with significant decreases in levels of plasminogen
`activator inhibitor-1 (PAI-1) and B-type natriuretic peptide
`(BNP), both of which are considered as biomarkers for
`cardiovascular risk [74]. The mechanisms for these effects also
`remain to be shown.
`Although blood pressure reductions and improved lipid
`profiles similar to those experienced with GLP-1 RAs have
`not been seen in clinical trials of DPP-4 inhibitors, a modest
`reduction in blood pressure has been reported for sitagliptin
`and vildagliptin in some studies [75–77]. A retrospective study
`of a large cohort database that found an association between
`sitagliptin treatment, slight weight loss and a small decrease
`in blood pressure suggests that the improvement in blood
`pressure is connected to weight loss [75], but more study
`of the underlying mechanism is warranted because DPP-4
`inhibitors are generally considered weight neutral [60,61]. A
`few studies have also recorded modest beneficial effects on lipid
`profiles with sitagliptin and vildagliptin [75,78,79]. One clinical
`trial concluded that vildagliptin may counteract postprandial
`hyperlipidaemia by either decreasing chylomicron production,
`increasing chylomicron clearance or both [79]. However, more
`study is needed to confirm whether DPP-4 inhibitors have any
`clinically significant effect on lipid levels and, if so, what the
`possible mechanisms would be.
`
`Metabolism and Tolerability
`Sitagliptin and saxagliptin are eliminated from the body primar-
`ily through renal excretion; in Europe, sitagliptin is therefore
`not recommended for patients with moderate and severe renal
`insufficiency, while saxagliptin can be used in these populations
`with dose reductions. In the USA, lower dosages should be used
`with both treatments in these populations [31,32,34,35]. Exe-
`natide is also eliminated by the kidneys, and both exenatide
`twice daily and once weekly are contraindicated in patients
`with severe renal impairment or end-stage renal disease (CrCl
`<30 ml/min) [21,22,37]. Caution should also be applied when
`initiating or escalating doses of exenatide BID in patients
`with moderate renal impairment (CrCl 30–50 ml/min) and
`in patients with renal transplantation [21,37]. Exenatide once
`
`22 Blonde and Montanya
`
`Volume 14 No. (Suppl. 2) April 2012
`
`I
`
`MPI EXHIBIT 1058 PAGE 3
`
`MPI EXHIBIT 1058 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0003
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`review article
`
`NS
`
`diarrhoea
`
`Nausea,URTI,
`
`vomiting
`
`Nausea,diarrhoea,
`
`diarrhoea
`
`Nausea,URTI,
`
`hypoglycaemia
`hyperhidrosis,
`
`Tremor,
`
`headache,dizziness
`
`Pio
`
`Nasopharyngitis,
`
`nausea/diarrhoea
`headache,
`
`5.3¶
`
`5.3¶
`
`4.5¶
`
`assistance)
`requiring
`incidents
`hypoglycaemic
`are10severe
`16.2†(included
`
`1.7§
`
`NS
`
`Nasopharyngitis,
`
`5%minor§
`
`open-labelmet
`medicationplus
`blindedstudy
`inadditionto
`upwardto45mg)
`couldbetitrated
`pio15mg(which
`receivedopen-label
`extensionand
`enrolledintrial
`
`Patientsimmediately
`
`headache/influenza
`nasopharyngitis,
`
`Diarrhoea,
`
`headache,URTI
`
`Nasopharyngitis,
`
`headache
`diarrhoea,
`
`Nasopharyngitis,
`
`headache
`nasopharyngitis,
`
`Diarrhoea,
`increased
`UTI/headache/BG
`nasopharyngitis,
`
`Hyperglycaemia,
`
`influenza
`nasopharyngitis,
`
`SU
`
`Hyperglycaemia,
`
`allowed
`medicationif
`Rescue
`
`therapyreported
`commonAEsper
`Threemost
`
`5.0†
`
`3.9†
`
`5.2†
`
`7.8†
`
`∗
`
`2.3
`
`(%)
`hypoglycaemia
`moderate
`mildor
`Frequencyof
`
`∗
`
`0.4
`
`−0.3
`−2.8
`−1.6
`
`+1.6
`−0.2
`
`−1.0
`
`effect
`placebo
`similarto
`Allchanges
`
`effect
`placebo
`similarto
`Allchanges
`
`(kg)
`baseline
`from
` weight
`
`+0.1
`−0.8
`−0.4
`
`−0.5
`−0.4
`
`−0.9‡
`
`+0.1
`−0.6
`
`−0.7
`
`−0.6
`
`+0.2
`
`34
`
`34
`
`34
`
`31.7
`
`31.8
`
`32.6
`
`31.6
`
`31.1
`
`31.2
`
`31.7
`
`30.1
`
`−0.5
`(%)
`frombaseline
` HbA1c
`
`29.9
`
`(kg/m2)
`BMI
`Baseline
`
`8.2
`
`8.2
`
`8.3
`
`7.3
`
`7.3
`
`8.5
`
`8.1
`
`8.0
`
`8.1
`
`8.1
`
`8.0
`
`8.1
`
`(%)
`HbA1c
`Baseline
`
`6.6
`
`4.9
`
`6.2
`
`5.8
`
`5.7
`
`6.3
`
`6.7
`
`6.3
`
`6.4
`
`6.7
`
`(53%)
`n=93
`>5years
`
`(56%)
`n=285
`>5years
`
`(years)
`ofdiabetes
`Duration
`
`PlaceboBID113
`
`113
`
`110
`
`10μgBID
`
`5μgBID
`
`1393
`
`≤6mg/day
`Glimepiride
`
`1396
`
`50mgBID
`
`219
`
`100mgOD
`
`179
`
`PlaceboOD
`
`181
`
`10mgOD
`
`191
`
`5mgOD
`
`≥1500
`
`≥1500
`
`≥1500
`
`[49]
`
`30
`
`Exenatide
`
`[48]
`
`52
`
`Vildagliptin
`
`26
`
`192
`
`2.5mgOD
`
`≥1500
`
`[47]
`
`24
`
`Saxagliptin
`
`177
`
`PlaceboOD
`
`523
`
`5mgOD
`
`(n)
`Participants
`
`addition
`Therapy
`
`≥1500
`(mg/day)
`dosage
`Met
`
`[46]
`
`24
`
`Linagliptin
`
`(weeks)
`duration
`Trial
`
`comefromseparatetrialswithdifferentpatientpopulationcharacteristicsanduniquestudydesigns,anddonotsupportdirectcomparison.
`Table1.Studyresultsofincretin-relatedtherapiesamongpatientsinadequatelycontrolledwithmetformininrandomisedtrials.ExceptforthePratleystudycomparingsitagliptinandliraglutide,resultsshown
`
`Volume 14 No. (Suppl. 2) April 2012
`
`doi:10.1111/j.1463-1326.2012.01575.x 23
`
`I
`
`MPI EXHIBIT 1058 PAGE 4
`
`MPI EXHIBIT 1058 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0004
`
`

`

`review article
`
`DIABETES, OBESITY AND METABOLISM
`
`headache/diarrhoea
`nasopharyngitis,
`
`Nausea,
`
`headache
`nasopharyngitis,
`
`NS
`
`Nausea,
`
`oedema,diarrhoea
`
`URTI,peripheral
`
`headache
`
`Nausea,diarrhoea,
`
`vomiting
`
`NS
`
`Nausea,diarrhoea,
`
`allowed
`medicationif
`Rescue
`
`therapyreported
`commonAEsper
`Threemost
`
`5%minor§
`occurred)
`assistance
`eventrequiring
`hypoglycaemic
`major
`additionone
`5%minor§(in
`
`1
`
`3
`
`1
`
`(%)
`hypoglycaemia
`moderate
`mildor
`Frequencyof
`
`−2.9
`2.8
`
`−0.8
`
`−1.2
`−1.2
`−0.9
`
`−2.3
`(kg)
`baseline
`from
` weight
`
`−1.5
`(%)
`baseline
`from
` HbA1c
`
`¶Symptomsreportedconsistentwithpossiblyconfirmedplasmaglucose<3.3mmol/l.
`§Plasmaglucose<3.1mmol/l.
`‡TherelativehighefficacyofsitagliptinincomparisontotheefficacyoftheotherDDP-4inhibitorsoccurredinpatientswhowerenottreatmentna¨ıve.
`†Mildormoderatereportedhypoglycaemiadidnotrequiretreatmentormedicalintervention.
`Plasmaglucose≤3.9mmol/l.
`respiratorytractinfection.
`AE,adverseevent;BG,bloodglucose;BID,twicedaily;BMI,bodymassindex;met,metformin;NS,notspecified;OD,oncedaily,pio,pioglitazone;SU,sulphonylurea;UTI,urinarytractinfection;URTI,upper
`
`∗
`
`−3.4
`
`−1.5
`
`33.1
`
`8.4
`
`6.4
`
`221
`
`1.8mgOD
`
`32.6
`
`32
`
`32
`
`32
`
`8.4
`
`8.5
`
`8.5
`
`8.6
`
`6
`
`5
`
`6
`
`165
`
`166
`
`160
`
`(kg/m2)
`BMI
`Baseline
`
`(%)
`HbA1c
`Baseline
`
`(years)
`diabetes
`Durationof
`
`(n)
`Participants
`
`45mgpio
`
`sitagliptin
`
`100mg
`
`weekly
`2mgonce
`
`addition
`Therapy
`
`study
`throughout
`
`weekly[50]
`once
`
`Stabledoses
`
`26
`
`Exenatide
`
`(mg/day)
`dosage
`Met
`
`(weeks)
`duration
`Trial
`
`Table1.Continued.
`
`6.0
`
`225
`
`1.2mgOD
`
`≥1500
`
`[26]
`
`26
`
`Liraglutide
`
`24 Blonde and Montanya
`
`Volume 14 No. (Suppl. 2) April 2012
`
`I
`
`MPI EXHIBIT 1058 PAGE 5
`
`MPI EXHIBIT 1058 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0005
`
`

`

`DIABETES, OBESITY AND METABOLISM
`
`weekly is not recommended in patients with moderate renal
`impairment [22]. Vildagliptin, which is eliminated primarily
`by the kidneys, is not recommended in patients with moderate,
`severe or end-stage renal disease [30], nor should it be used in
`patients with hepatic impairment, including those who have
`alanine aminotransferase (ALT) or aspartate aminotransferase
`(AST) levels three times the normal upper limit. Liver function
`tests for ALT and AST should be performed before treatment,
`and periodically thereafter [30].
`In contrast to the other incretin-related therapies, linagliptin
`is mainly eliminated unchanged via faeces, so no dose
`reduction is recommended in the case of renal or hepatic
`impairment [33,80]. Liraglutide degrades in the body by
`enzymatic activity like GLP-1, only much more slowly [81],
`and should therefore pose a lower risk of accumulation in the
`case of renal disease.
`In a study of 30 patients who were given a single dose of
`liraglutide 0.75 mg, no clear change in pharmacokinetics was
`evident among the 24 patients with varying degrees of renal
`dysfunction compared with the 6 patients with normal renal
`function [82]. In a meta-analysis of 2783 patients, of whom
`486 receiving liraglutide treatment had mild renal impairment,
`similar decreases in HbA1c occurred in patients with mild
`renal impairment (60 ml/min ≤ CrCl ≤ 89 ml/min), compared
`with patients with normal renal function (CrCl > 89 ml/min)
`who received either liraglutide 1.2 or 1.8 mg as monotherapy
`or in combination with an oral anti-hyperglycaemic for 26
`weeks [83]. Changes in serum creatinine were not significantly
`different
`from baseline for either treatment groups [83];
`however, there is limited experience with liraglutide in patients
`with moderate renal impairment and no experience in patients
`with severe renal impairment. Consequently, liraglutide is not
`recommended for these two patient groups in Europe [20].
`In the USA, there have been post-marketing reports of acute
`renal failure and worsening of chronic renal failure, usually in
`association with nausea, vomiting, diarrhoea or dehydration,
`which may sometimes require haemodialysis, so caution is
`warranted when initiating or escalating doses in patients with
`renal impairment [36].
`Very few drug interactions have been noted with incretin
`therapies [84]. When used with an SU, a reduction in the dosage
`of the SU should be considered to reduce the risk of hypogly-
`caemia with most incretin-related therapies [20–22,31–33].
`In the case of vildagliptin, dosage should be reduced when
`combined with a SU [30]. When saxagliptin is used concur-
`rently with a strong cytochrome P450 3A4/5 (CYP3A4/5)
`inhibitor, for example, ketoconazole, plasma concentrations of
`saxagliptin will increase, so a dosage reduction of saxagliptin
`is recommended in the USA, although none is advised in
`Europe [31,35]. Linagliptin’s efficacy may be reduced by a
`strong P-gp or CYP3A4 inducer, such as rifampicin, so
`an alternative treatment is advised [33]. Unlike some other
`pharmacologic treatments for diabetes, particularly SUs and
`insulin, hypoglycaemia is less common with incretin-related
`therapies [40,85]. Use of DPP-4 inhibitors may be associated
`with an increased risk of infections, such as urinary tract
`infections, nasopharyngitis, upper respiratory tract infections
`and headaches [33–35,43]. With exenatide and liraglutide a
`
`review article
`
`few cases of angiooedema have been reported [20,21]. More
`common with GLP-1 RAs are gastrointestinal adverse events
`(primarily nausea), which are usually transient and occur
`mostly during the first 4 weeks of treatment [54,56]. A phase
`2 study that used a validated scale system, the Gastrointesti-
`nal System Rating Scale (GSRS), to evaluate the quantity and
`intensity of gastrointestinal adverse events from the patients’
`perspective confirmed that such disorders are usually tran-
`sient and of mild-to-moderate severity [86]. Titrating the
`dosage of GLP-1 RAs according to instructions can help
`to reduce the incidence of gastrointestinal effects. Pancre-
`atitis has been reported with both DPP-4 inhibitors and
`GLP-1 RAs [20–22,32,33]; however, patients with T2D have
`a nearly three-times greater risk of developing pancreatitis
`than individuals without diabetes [87], and a causal rela-
`tionship between use of incretin-based therapies and pan-
`creatitis has not been established. The evidence regarding the
`risk for pancreatitis with incretin-related therapies compared
`with non-incretin-related therapies is, at present, conflict-
`ing [88–91].
`In rodent studies, thyroid C-cell tumours have resulted from
`exposure to high doses of exenatide and liraglutide [20–22,92],
`but the relevance of these findings for humans is not yet
`known [84]. The ability of GLP-1 RAs to stimulate calcitonin
`release appears to be species-dependent. In studies with non-
`human primates exposed for up to 87 weeks with doses 60-fold
`greater than recommended for humans and in clinical trials
`with up to 2 years’ exposure,
`increased calcitonin release
`did not occur with liraglutide treatment [92]. In a meta-
`analysis of clinical trials lasting no more than 2 years with
`over 5000 patients receiving either liraglutide or control
`therapy, 3-month measurements of serum calcitonin showed
`that mean serum calcitonin concentrations were at the low
`end of the normal range in all treatment groups at baseline
`and remained low throughout the trials [93]. The European
`label for liraglutide cautions patients against possible thyroid
`adverse events [20], while the US prescribing instructions state
`that liraglutide is contraindicated in patients who have a
`personal or family history of medullary thyroid carcinoma
`or of Multiple Endocrine Neoplasia syndrome type 2 (MEN
`2) [36]. Monitoring with serum calcitonin tests or thyroid
`ultrasounds is not advised by the American Association of
`Clinical Endocrinologists (AACE) because it may increase
`unnecessary procedures, because of low test specificity for
`serum calcitonin and a high background incidence of thyroid
`disease. AACE also does not recommend obtaining baseline
`serum calcitonin levels or thyroid ultrasounds unless thyroid
`nodules are detected on initial physical examination, or
`if nodules occur during administration of liraglutide [94].
`Thyroid nodules in liraglutide-treated patients should be
`managed in the usual manner [94].
`
`Clinical Trials Comparing DPP-4 Inhibitors
`and GLP-1 RAs
`So far, few trials have been conducted directly comparing the
`two classes of incretin-related therapies. The available data are
`included here.
`
`Volume 14 No. (Suppl. 2) April 2012
`
`doi:10.1111/j.1463-1326.2012.01575.x 25
`
`I
`
`MPI EXHIBIT 1058 PAGE 6
`
`MPI EXHIBIT 1058 PAGE 6
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1058-0006
`
`

`

`review article
`
`Exenatide taken BID was compared with sitagliptin in a
`4-week, randomised, crossover study in 61 patients with T2D
`with an average HbA1c of 8.5 ± 1.2% and an average 2-h
`PPG of 245 ± 65 mg/dl [45]. Patients received exenatide (5 μg
`BID for 1 week, then 10 μg BID for 1 week) or sitagliptin
`[100 mg once every morning (QAM)] for 2 weeks. After
`2 weeks, patients crossed over to the alternate therapy. At
`the end of the study, the change in fasting plasma glucose
`(FPG) was similar with the two therapies (−15 ± 4 mg/dl
`for exenatide vs. −19 ± 4 mg/dl for sitagliptin; p = 0.3234),
`but
`the 2-h PPG levels were significantly lower with
`exenatide (−133 ± 6 mg/dl vs. −208 ± 6 mg/dl, respectively;
`p < 0.0001). When patients switched from exenatide to
`sitagliptin, 2-h PPG rose by 73 ± 11 mg/dl, and when
`patients switched from sitagliptin to exenatide, 2-h PPG
`decreased by 76 ± 10 mg/dl. Compared to sitagliptin, exenatide
`showed greater improvement in the insulinogenic index of
`insulin secretion (ratio 1.50 ± 0.26; p = 0.0239), produced
`a greater reduction in postprandial glucagon [AUC0 – 240 min
`(pg min/ml) geometric LS mean ratio 0.88 ± 0.03; p = 0.0011]
`and total caloric intake (−134 ± 97 kcal vs. +130 ± 97 kcal;
`p = 0.0227), and slowed gastric emptying to a greater extent
`[acetaminophen AUC0 – 240 min (mg min/dl) LS mean ratio
`0.56 ± 0.05; p < 0.0001]. With both treatments, mild-to-
`moderate gastrointestinal symptoms were the most common
`adverse events reported.
`In a double-blind comparison of exenatide once weekly with
`sitagliptin or the TZD treatment pioglitazone, patients were
`randomised to 2 mg exenatide as a once-weekly injection plus
`oral placebo once daily (n = 160), 100 mg sitagliptin once
`daily plus once-weekly placebo injection (n = 166), or 45 mg
`pioglitazone once daily plus once-weekly placebo injection
`(n = 165) for 26 weeks [50]. Baseline HbA1c levels were 8.6, 8.5
`and 8.5%, respectively. At trial end, the reduction in HbA1c was
`−1.5% with exenatide once weekly, −0.9% with sitagliptin and
`−1.2% with pioglitazone with treatment differences of −0.6%
`(95% CI [−0.9; −0.4]; adjusted p < 0.0001) for exenatide
`once weekly vs. sitagliptin, and −0.3% (95% CI [−0.6; −0.1];
`adjusted p = 0.0165) vs. pioglitazone. Weight change with
`exenatide once weekly was −2.3 kg, compared to −0.8 kg with
`sitagliptin (p = 0.0002) or +2.8 kg with pioglitazone (p <
`0.0001) [50]. There was also a significant difference in reduction
`of SBP between exenatide once weekly and sitagliptin (−4
`mmHg; p = 0.0055), but none between exenatide once weekly
`and pioglitazone [50]. The most common adverse events
`patients experienced for exenatide once weekly and sitagliptin
`were nausea (24 and 10%, respectively)

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