throbber
Incretin-Based Therapies for Type 2 Diabetes Mellitus:
`Current Status and Future Prospects
`
`Scott R. Drab, Pharm.D.
`
`Incretin-based therapies encompass two new classes of antidiabetic drugs:
`glucagon-like peptide-1 (GLP-1) receptor agonists (e.g., liraglutide, exenatide,
`and exenatide long-acting release), which are structurally related to GLP-1,
`and the dipeptidyl peptidase-4 (DPP-4) inhibitors (e.g., sitagliptin and
`saxagliptin), which limit the breakdown of endogenous GLP-1. To evaluate
`the safety and effectiveness of incretin-based therapies for the treatment of
`type 2 diabetes mellitus and the role of these therapies in clinical practice, a
`MEDLINE search (January 1985–November 2009) was conducted. Relevant
`references from the publications identified were also reviewed. Of 28 studies
`identified, 22 were randomized controlled trials. Data show that these
`therapies affect insulin secretion in a glucose-dependent manner, achieving
`clinically meaningful reductions in hemoglobin A1c levels, with very low rates
`of hypoglycemia. In addition, reductions in body weight have been observed
`with GLP-1 receptor agonists, which also exert a pronounced effect on
`systolic blood pressure. Various human and animal studies show that GLP-1
`improves ␤-cell function and increases ␤-cell proliferation in vitro, which may
`slow disease progression. Thus, incretin-based therapies represent a
`promising addition to the available treatments for type 2 diabetes.
`type 2 diabetes, incretin-based therapies, glucagon-like peptide-
`Key Words:
`1, GLP-1, dipeptidyl peptidase-4, DPP-4.
`(Pharmacotherapy 2010;30(6):609–624)
`
`OUTLINE
`Treatment Potential for Type 2 Diabetes Mellitus
`Goals of Therapy and Limitations of Current
`Treatments
`Incretin-Based Therapies
`Physiologic Effects of Glucagon-Like Peptide-1
`Two Alternative Approaches
`Pharmacology
`Efficacy and Safety: Clinical Studies
`Benefits
`Dosing and Administration
`Place in Therapy
`Conclusion
`
`From the University of Pittsburgh School of Pharmacy,
`Pittsburgh, Pennsylvania, and the University Diabetes Care
`Associates, Jeannette, Pennsylvania.
`For reprints, visit http://www.atypon-link.com/PPI/loi/phco.
`For questions or comments, contact Scott R. Drab,
`Pharm.D., University of Pittsburgh School of Pharmacy, 922
`Salk Hall, Pittsburgh, PA 15261; e-mail: drab@pitt.edu.
`
`The increasing prevalence of diabetes mellitus
`is an acknowledged world health crisis that is
`both a major contributor to patient morbidity
`and mortality and a huge economic burden.1 The
`complex pathophysiology of type 2 diabetes
`makes effective treatment problematic. Hyper-
`glycemia is associated with an increased risk of
`microvascular complications, sensory neuropathy,
`myocardial infarction, stroke, macrovascular
`mortality, and all-cause mortality.2 Type 2 diabetes
`is also linked causally with obesity—in itself a
`global health crisis—which independently increases
`the risk of serious cardiovascular comorbidities.1
`Hypertension, also often associated with type 2
`diabetes, can further increase cardiovascular risk.
`The global economic impact of diabetes, due to
`premature death and complications from the
`disease, is considerable. At least $232 billion were
`spent on treatment and prevention of diabetes
`worldwide in 2007, with three quarters of that
`
`MPI EXHIBIT 1022 PAGE 1
`
`MPI EXHIBIT 1022 PAGE 1
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0001
`
`

`

`610
`
`PHARMACOTHERAPY Volume 30, Number 6, 2010
`
`amount spent in industrialized countries on the
`treatment of long-term complications and on
`general care, such as efforts to prevent micro-
`and macrovascular complications.3 The indirect
`costs of the disease must also be included in any
`discussion of its burden. In 2007, it was estimated
`that disability, lost productivity, and premature
`death due to diabetes cost the United States
`economy alone $58 billion.1
`
`Treatment Potential for Type 2 Diabetes
`Mellitus
`Insulin resistance and impaired insulin secretion
`both contribute to the development of type 2
`diabetes, with the shift from normal to impaired
`glucose tolerance and then to diabetes, accom-
`panied by a 40% decrease in insulin sensitivity
`and a 3–4-fold deterioration in insulin secretion
`due to loss of ␤-cell capacity (50–80%).4, 5 However,
`if impaired glucose tolerance is diagnosed early
`enough, overt type 2 diabetes can be prevented or
`at least delayed, with lifestyle interventions
`aimed at reducing calorie intake and body fat6, 7
`or through drug treatment7 to normalize glycemic
`control. Once type 2 diabetes has been diagnosed,
`weight loss alone can greatly improve both
`glycemic control and patients’ cardiovascular risk
`profiles.8 The effects are interrelated: glycemic
`control, coupled with effective management of
`additional risk factors, benefits cardiovascular
`prognosis.9
`Despite treatment guidelines that recommend
`early, aggressive intervention,10, 11 many patients
`fail to reach targets for glycemic control (~43% of
`patients in the United States have hemoglobin
`A1c [A1C] levels ≥ 7%),12 with minority groups
`having greater disease prevalence.13, 14
`Many factors contribute to the failure to manage
`type 2 diabetes successfully. The most fundamental
`of these are the necessary lifestyle modifications,
`which, if not implemented at the time of diagnosis,
`may result in further decline in glycemic control
`and hasten the need for pharmacotherapy. Other
`factors include psychosocial and economic
`influences and shortcomings in the efficacy and
`tolerability profiles of many available antidiabetic
`drugs.
`
`Goals of Therapy and Limitations of Current
`Treatments
`To successfully improve patient outcomes and
`minimize the risk of complications and their
`cost, treatment should ideally address deterio-
`rating ␤-cell function, A1C, and fasting plasma
`
`glucose (FPG) and postprandial plasma glucose
`(PPG) levels simultaneously, without increasing
`risk of hypoglycemia, weight gain, or cardio-
`vascular disease contributors. Adequate glycemic
`control is essential, as reduction of A1C levels
`can decrease the risks of microvascular and
`macrovascular complications and mortality
`associated with type 2 diabetes.2 For every 1%
`reduction in A1C, a 37% decrease in risk for
`microvascular complications (p<0.0001) and a
`21% decrease in risk of death related to diabetes
`(p<0.0001) have been observed. 2 An ideal
`treatment would reduce cardiovascular risk
`factors as well as control blood glucose levels.
`Current guidelines for type 2 diabetes from the
`American Diabetes Association (ADA) and the
`European Association for the Study of Diabetes
`(EASD) advocate a stepwise escalation of
`intervention, starting with lifestyle modification
`and metformin 15; however, sulfonylureas, a
`common second-line intervention in type 2
`diabetes treatment regimens, are limited by a
`failure of ␤-cell function that underlies the
`progression of the disease. Although ␤-cell
`function cannot be measured directly in humans,
`several indicators can be used to monitor its
`decline. These include the homeostatic model
`assessment of ␤-cell function (HOMA-B), which
`yields an estimate from fasting plasma insulin and
`glucose concentrations,16 and the proinsulin:insulin
`ratio. The HOMA-B results have shown that
`established oral antidiabetic drug therapies such
`as metformin, sulfonylureas, and thiazolidine-
`diones can improve ␤-cell function during the
`first year of treatment when they are given as
`initial treatment in patients with newly diagnosed
`type 2 diabetes; however, ␤-cell function declines
`progressively thereafter.4, 17, 18 The A1C level
`parallels these changes in ␤-cell function,
`decreasing in the first year of therapy and then
`increasing progressively, highlighting the need for
`therapies that can sustain improvements in ␤-cell
`function.19
`Insulin therapy is necessary once ␤-cell
`secretory capacity becomes insufficient; however,
`alternative antihyperglycemic drugs—the
`incretin-based therapies—are now available that
`may offer advantages over conventional oral
`antidiabetic drug therapies. These incretin-based
`agents were included in the 2009 recommen-
`dations of an American Association of Clinical
`Endocrinologists–American
`College
`of
`Endocrinology consensus panel,20 which sug-
`gested that patients with A1C levels of 7.6–9.0%
`be treated with dual therapy with metformin
`
`MPI EXHIBIT 1022 PAGE 2
`
`MPI EXHIBIT 1022 PAGE 2
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0002
`
`

`

`INCRETIN-BASED THERAPIES FOR TYPE 2 DIABETES MELLITUS Drab
`
`611
`
`(unless contraindicated) plus, in order of
`preference, glucagon-like peptide-1 (GLP-1)
`receptor agonists, dipeptidyl peptidase-4 (DPP-4)
`inhibitors, glinides, or sulfonylureas. The panel
`emphasized the lower risk of hypoglycemia with
`GLP-1 receptor agonists and DPP-4 inhibitors
`compared with glinides and sulfonylureas, but
`preferred GLP-1 receptor agonists over DPP-4
`inhibitors because of their greater potential for
`lower PPG level and substantial weight loss. For
`patients requiring triple therapy, metformin, a
`GLP-1 receptor agonist, and a third oral antidia-
`betic drug—a thiazolidinedione, a sulfonylurea,
`or a glinide—are recommended.
`
`Incretin-Based Therapies
`
`Physiologic Effects of Glucagon-Like Peptide-1
`The “incretin effect” describes the phenomenon
`whereby a glucose load delivered orally produces
`a much greater insulin secretion than the same
`glucose load administered intravenously.21 This
`effect is mediated by two incretin hormones
`secreted by intestinal cells.22 Glucose-dependent
`insulinotropic polypeptide (GIP) was identified
`first,22 followed by GLP-1.23
`It is now thought
`that healthy individuals may derive up to 70% of
`their prandial insulin secretory response from the
`incretin effect.24 Under normal conditions, the
`incretin peptides are secreted as needed, in
`response to ingested nutrients, and have a short
`plasma half-life due to degradation by the
`ubiquitous DPP-4 enzyme.25
`In people with type 2 diabetes, pancreatic
`responsiveness to GLP-1 is impaired,26 but the
`insulin-secretory response can be restored with
`pharmacologic doses of human GLP-1.27 Further
`studies in vitro and in animals show that GLP-1
`
`40
`
`:15
`:? 30
`.€
`_5
`:3..
`
`10
`-::;
`E 15
`10
`
`5
`
`/11fusion
`
`7()
`
`6(1
`
`.J .~n
`,:
`~
`s 40
`"'
`"" 30
`rr
`=>
`~ _()
`
`111
`
`promotes ␤-cell neogenesis and preservation28–30
`and inhibits ␤-cell apoptosis.30, 31 Also, GLP-1
`has potentially beneficial effects on cardiac
`function:
`it has been shown to be cardio-
`protective in ischemia-reperfusion models in rats
`in vivo,32 and to reduce blood pressure in Dahl
`salt-sensitive rats.33 In humans, GLP-1 improves
`left ventricular function as well as endothelial
`function in patients with high cardiac risk.34, 35
`Along with these benefits, GLP-1 has been shown
`to slow the rate of gastric emptying in humans36
`and to reduce appetite. 37 These effects are
`particularly important, since type 2 diabetes is
`often associated with obesity and excessive
`caloric intake.
`It is clear that drugs acting
`through the physiologic GLP-1 pathways might
`have important ancillary benefits for the
`treatment of type 2 diabetes.
`A key feature of GLP-1 action is the glucose-
`dependent stimulation of insulin secretion and
`concomitant suppression of glucagon. When
`patients with type 2 diabetes are infused with
`human GLP-1, insulin levels return toward basal
`levels when normal FPG concentrations are
`reached, despite ongoing infusion of GLP-1.38
`Continuous subcutaneous GLP-1 infusion for 6
`weeks also reduced A1C by 1.3% and FPG by 77
`mg/dl, slowed gastric emptying, and reduced
`appetite and body weight by 1.9 kg.39 Therefore,
`GLP-1 and therapies that activate GLP-1
`receptors only stimulate increases in insulin
`secretion when glucose levels are elevated and, as
`a result, carry a very low risk of hypoglycemia
`(Figure 1) unless combined with a sulfonylurea
`or insulin.
`Unfortunately, therapeutic use of human GLP-
`1 is not practical due to its rapid degradation by
`DPP-4 enzyme,25 with an elimination half-life of
`
`Infusion
`
`lnfusi"n
`
`~o
`
`'.] 100
`=2
`""
`E 150
`,.,
`g
`=
`e
`
`100
`
`50
`
`60
`
`)20
`
`1~0
`
`2~(1
`
`()
`-JO 0
`
`6()
`
`120
`irne(mi11)
`
`180
`
`~40
`
`u
`- 30 II
`
`~ I~ I
`
`I "O
`
`].1(1
`
`0
`
`- 3fl
`
`-- Nuti1•c h11111n11 GI. I'• I
`-- rlacobo
`
`Figure 1. Effects of glucagon-like peptide-1 (GLP-1) or placebo infusion on insulin, glucagon, and glucose levels. *p<0.05.
`(From reference 38 with permission.)
`
`MPI EXHIBIT 1022 PAGE 3
`
`MPI EXHIBIT 1022 PAGE 3
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0003
`
`

`

`612
`
`PHARMACOTHERAPY Volume 30, Number 6, 2010
`
`about 2 minutes.27 As a result, two alternative
`approaches to harnessing the therapeutic poten-
`tial of the incretin system have been pursued—
`GLP-1 receptor agonists and DPP-4 inhibitors.
`
`Two Alternative Approaches
`
`The GLP-1 receptor agonists are designed to
`fulfill the role of native GLP-1 while exhibiting
`resistance to degradation by DPP-4, and are
`present for therapeutic use at supraphysiologic
`levels (equivalent to 6–10-fold normal GLP-1
`levels). These drugs include exenatide (available
`since 2005), exenatide long-acting release (LAR),
`which is undergoing regulatory review by the
`U.S. Food and Drug Administration (FDA); and
`liraglutide, which is available in the United
`States, Europe, and Japan. Other drugs (AVE-
`0010 bid/ZP10, albiglutide, and taspoglutide/
`R1583) have recently entered phase III trials;
`their data will not be reviewed.
`The DPP-4 inhibitors act to impair the activity
`of the DPP-4 enzyme, thereby increasing the half-
`life of endogenous GLP-1. Compared with the
`GLP-1 receptor agonists, DPP-4 inhibitors
`produce modestly elevated levels of GLP-1 and
`exert their effects principally through interaction
`with receptors on afferent nerves rather than
`through receptors on target organs.40 The first of
`these drugs to enter the market was sitagliptin,
`which was approved in the United States in 2006.
`A second DPP-4 inhibitor, vildagliptin, has been
`approved for use in Europe; however, since its
`manufacturer is not pursuing approval in the
`United States, vildagliptin will not be discussed
`in detail. Saxagliptin has received recent European
`Medicines Agency and FDA approval, but U.S.
`approval of alogliptin has been delayed because
`of the requirement for an additional cardiovascular
`study.
`In December 2008, the FDA issued the
`recommendation that all therapies for the
`treatment of type 2 diabetes should provide
`evidence that they do not increase the risk of
`cardiovascular events. Hence, alogliptin will not
`be reviewed.
`To identify relevant literature on the incretin-
`based therapies, a MEDLINE search (January
`1985–November 2009) was performed by using
`the following key words:
`type 2 diabetes,
`incretin, GLP-1, DPP-4, liraglutide, exenatide,
`sitagliptin, and saxagliptin. Relevant references
`from the publications identified during this
`search were also reviewed. All studies that were
`published in the English language and that
`evaluated the safety and efficacy of incretin-based
`
`therapies were analyzed. Priority was given to
`randomized clinical trials and retrospective
`studies in clinical settings.
`
`Pharmacology
`
`Glucagon-Like Peptide-1 Receptor Agonists
`Exenatide. This drug is a recombinant peptide
`based on exendin-4, which is found in the saliva
`of Heloderma suspectum (i.e., Gila monster
`lizard). Exenatide shares 53% of its amino acid
`sequence identity with human GLP-1 and
`functions as a full agonist of the GLP-1
`receptor.41
`It is, however, more resistant to
`degradation by DPP-4 enzyme, due to the
`presence of a glycine residue on the penultimate
`NH2 group.42
`Studies investigating the pharmacokinetic
`profile of exenatide show that it reaches peak
`plasma levels at about 2 hours after subcutaneous
`injection, has a plasma half-life of 3–4 hours, and
`provides glucose reduction over 5–7 hours.43, 44
`This means that exenatide requires twice-daily
`dosing (0–60 min before breakfast and dinner)
`and exerts its predominant effect on PPG levels.44
`A new drug application for a long-acting (slow-
`absorption) formulation requiring once-weekly
`dosing (exenatide LAR) was submitted in May
`2009.
`In March 2010, a response letter was
`issued by the FDA requesting the finalization of
`the product labeling, risk evaluation and
`mitigation strategy, and clarification of existing
`manufacturing processes.
`Preliminary data
`indicate that exenatide LAR may have better
`efficacy than twice-daily exenatide.45 Exenatide
`is renally excreted,46 and although no dosage
`adjustment is needed in cases of mild or
`moderate renal impairment, exenatide is not
`recommended for use in patients with severe
`renal impairment (creatinine clearance < 30
`ml/min) or end-stage renal disease ([ESRD] i.e.,
`chronic, irreversible renal failure). This advice
`follows the finding that single doses of exenatide
`5 µg caused gastrointestinal adverse effects in
`patients with ESRD who were receiving dialysis.47
`Gastrointestinal adverse events in these patients
`may worsen renal function.
`In November 2009,
`the FDA issued guidance advocating caution
`when starting or uptitrating exenatide in patients
`with moderate renal impairment (creatinine
`clearance 30–50 ml/min).48
`Elimination of exenatide is not dependent on
`hepatic function, however, and it is therefore not
`expected to interact with hepatically metabolized
`drugs as a result of competition for liver
`
`MPI EXHIBIT 1022 PAGE 4
`
`MPI EXHIBIT 1022 PAGE 4
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0004
`
`

`

`INCRETIN-BASED THERAPIES FOR TYPE 2 DIABETES MELLITUS Drab
`
`613
`
`enzymes. One way in which exenatide and other
`GLP-1 receptor agonists might interact with
`other drugs is through their effects on gastric
`motility and, hence, absorption kinetics. Studies
`examining the possibility of such an effect on
`digoxin, warfarin, and a statin do not suggest that
`their kinetics will be altered to a clinically
`meaningful extent by concomitant exenatide.49–51
`
`Liraglutide. This GLP-1 receptor agonist is
`rather different from exenatide in its structure
`(Figure 2). It is an analog of human GLP-1 with
`97% primary amino acid sequence identity, but
`with a fatty acid side chain attached through a
`glutamic acid linker.52 This fatty acid structure
`enables reversible plasma albumin binding,
`which may contribute to the drug’s partial resis-
`tance to DPP-4 degradation and its subsequent
`prolonged action.53 The primary mechanism for
`prolonging the action of liraglutide, however, is
`thought to be through slowed absorption due to
`the self-association (as heptameric aggregates) of
`liraglutide molecules within the injection depot.53
`These heptamers are thought to be too large to
`pass through capillary membranes readily, so
`
`, a1h•e lummn GLJ'• l
`
`Lir~gllltldc.
`
`Figure 2. The structures of native glucagon-like peptide-1
`(GLP-1), liraglutide, and exenatide.
`(From reference 52
`with permission.)
`
`absorption is delayed pending the dissociation of
`liraglutide through dilution at the absorption
`site. Although liraglutide is partially resistant to
`DPP-4 degradation, elimination studies show that
`it is nevertheless degraded almost completely
`into small molecules by this enzyme and by
`endopeptidases, as with endogenous GLP-1.54, 55
`This means that its elimination is not dependent
`on hepatic or renal function, which may prove to
`be clinically advantageous.
`Studies that investigated the pharmacokinetic
`profile of liraglutide have shown that it reaches
`peak plasma levels 9–12 hours after dosing by
`subcutaneous injection, with a subsequent mean
`elimination half-life of 11–15 hours.56 Steady
`state is reached after 3 days, and therapeutic
`plasma liraglutide concentrations are observed
`for up to 24 hours after a single injection. These
`characteristics make liraglutide compatible with
`once-daily administration, and it may be possible
`to choose the dosing time without regard to
`meals.56, 57 As with exenatide, drug-drug inter-
`action studies have been performed to investigate
`liraglutide’s potential to affect the kinetics of
`drugs that are sensitive to gastric transit time due
`to their absorption properties. Again, it was
`concluded that the effect of liraglutide on
`atorvastatin, lisinopril, digoxin, and griseofulvin
`was unlikely to be clinically relevant.58
`
`Dipeptidyl Peptidase-4 Inhibitors
`Sitagliptin. This drug is an orally absorbed small
`molecule that produces at least 80% inhibition of
`DPP-4 activity for 12 hours at doses of 50 mg or
`more, and for 24 hours at doses of 100 mg or
`more.59 Therefore, the recommended dosage is
`100 mg once/day. Compared with placebo,
`sitagliptin results in at least a 2-fold increase in
`postprandial GLP-1 levels, both after single doses
`and at steady state, reducing postprandial glycemia
`and not causing hypoglycemia.59, 60 Steady state
`is reached after about 2 days.
`Inhibition of
`glucagon secretion is also seen with DPP-4
`inhibition, but this class of drugs does not seem
`to affect gastric motility to the extent of the GLP-
`1 receptor agonists, possibly because of the
`relatively modest increase in GLP-1 signal that
`DPP-4 inhibition alone induces.61 This might
`also account for the observation that weight
`tends to remain unchanged rather than reduced
`in response to DPP-4 inhibition. It is also worth
`noting that the DPP-4 enzyme degrades other
`peptides besides the incretins,
`including
`cytokines and chemokines,61 and inhibition of
`
`MPI EXHIBIT 1022 PAGE 5
`
`MPI EXHIBIT 1022 PAGE 5
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0005
`
`

`

`614
`
`PHARMACOTHERAPY Volume 30, Number 6, 2010
`
`this process might account for some of the
`adverse effects seen with DPP-4 inhibitors, which
`are elaborated on below.
`Sitagliptin is primarily (79%) eliminated
`unchanged by the kidney. This process involves
`active tubular excretion, so dosage adjustment is
`recommended in patients with renal impairment
`to ensure that plasma sitagliptin levels do not
`accumulate.62 Dosing should be reduced to 50
`mg once/day in patients with moderate renal
`insufficiency and to 25 mg once/day in cases of
`severe renal impairment or ESRD. Only about
`16% of sitagliptin undergoes hepatic metabolism;
`hence, its pharmacokinetics have been shown to
`be unaffected by mild-to-moderate hepatic
`failure.63 Drug interactions are not anticipated to
`be a problem with sitagliptin, and studies have
`shown that it does not alter the kinetics (to a
`clinically relevant extent) of other antidiabetic
`drugs, simvastatin, warfarin, digoxin, and oral
`contraceptives,64, 65 although a possible cytochrome
`P450 (CYP) interaction with lovastatin has been
`reported.66 The concomitant use of cyclosporine
`produced only modest changes in the pharmaco-
`kinetics of sitagliptin.65
`
`Saxagliptin. This is the second small-molecule
`once-daily DPP-4 inhibitor to become available
`in the United States. The pharmacokinetic and
`pharmacodynamic properties of saxagliptin have
`been investigated in two 2-week studies of a wide
`range of doses given once/day in both healthy
`subjects (40–400 mg)67 and patients with type 2
`diabetes (2.5–50 mg).67 The level of DPP-4
`inhibition with saxagliptin 24 hours after dosing
`was 50% of predose levels in the 2.5-mg group,
`and 79% of predose levels in the 400-mg group.
`Doses of 150 mg or greater provided the same
`level of inhibition, and no dose-related adverse
`events, including hypoglycemia, were observed
`during the study. All doses of saxagliptin
`increased postprandial levels of intact GLP-1 by
`1.5–3 times compared with placebo, but there
`was no dose-related trend in this effect.68 The
`recommended dosage is therefore 2.5 or 5 mg/day.
`Saxagliptin is extensively metabolized in the
`liver by the CYP system, and its primary
`metabolite is active and has about 50% of the
`DPP-4 inhibitory potency of
`the parent
`compound. Both the parent and the metabolite
`are renally excreted,69 and accumulation can
`occur in patients with renal
`impairment,
`necessitating a daily dose limit of 2.5 mg.
`Although some accumulation can occur with
`hepatic impairment, dosage adjustment does not
`
`appear necessary in patients with hepatic
`impairment but healthy renal function.70
`Because of its hepatic metabolism, exposure to
`saxagliptin is potentially affected by inducers or
`inhibitors of the CYP system. However, dosage
`adjustments are not considered necessary to
`compensate for increased elimination caused by
`inducers such as rifampin, nor to avoid accumu-
`lation due to moderate inhibitors such as
`amprenavir, aprepitant, erythromycin, fluconazole,
`fosamprenavir, grapefruit juice, and verapamil.
`The daily dose of saxagliptin should be limited to
`2.5 mg when used concomitantly with strong
`CYP inhibitors such as ketoconazole.70
`
`Efficacy and Safety: Clinical Studies
`The reviewed clinical studies investigating the
`efficacy and safety of incretin-based therapies are
`summarized in Table 145, 71–85 and Table 2.86–93
`
`Glucagon-Like Peptide-1 Receptor Agonists
`Exenatide. Pivotal phase III efficacy and safety
`data for exenatide were derived from three key
`Diabetes Management for Improving Glucose
`Outcomes (AMIGO) trials in patients with type 2
`diabetes who were inadequately controlled with
`either a sulfonylurea, 72 metformin, 73 or a
`sulfonylurea plus metformin75 (Table 1). In each
`of these trials, twice-daily exenatide was shown
`to reduce A1C significantly, by approximately
`0.9% from baseline over the 30-week trial period
`compared with placebo.72, 73, 75 These large, well-
`designed studies (double- or triple-blind,
`placebo-controlled, 30-wk trials with well-
`matched treatment and placebo groups) provided
`proof of the ability of exenatide to achieve
`significant A1C reductions over the time period
`studied. A pooled analysis of trial data plus two
`52-week extension studies, which were
`completed by 314 of 1446 patients in an intent-
`to-treat group from the 30-week randomized
`trials, found that the reductions in A1C were
`sustained (–1.1% over 2 yrs). 94 The same
`analysis showed that 48% of patients achieved
`A1C less than 7%, and that FPG level was
`reduced by 25.2 mg/dl.
`It should be noted that
`the 48% of patients reaching an A1C less than
`7.0% were those with baseline A1C values above
`7.0%. The subgroup with a baseline A1C of 9.0%
`or greater showed a mean ± SD reduction in A1C
`from baseline of 2.0 ± 0.2%, compared with 0.8 ±
`0.1% for the subgroup with baseline A1C less
`than 9.0%.94 Although this was an open-label
`noncontrolled trial, these results suggest that the
`
`MPI EXHIBIT 1022 PAGE 6
`
`MPI EXHIBIT 1022 PAGE 6
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0006
`
`

`

`INCRETIN-BASED THERAPIES FOR TYPE 2 DIABETES MELLITUS Drab
`
`615
`
`Table 1. Summary of Randomized Controlled Trials of the Glucagon-Like Peptide-1 Receptor Agonists
`Change in Change in Change in
`A1C
`Weight
`SBP
`(%)
`(kg)
`(mm Hg)
`
`Population
`
`Treatment Groups
`
`Study Duration
`Exenatide
`24 wks71
`
`232 drug-naïve patients,
`A1C 7.8%, weight 86 kg
`
`30 wks
`(AMIGO 2)72
`
`377 patients taking a sulfonylurea,
`A1C 8.6%, weight 96 kg
`
`30 wks
`(AMIGO 1)73
`
`336 patients taking metformin,
`A1C 8.2%, weight 100 kg
`
`26 wks76
`
`52 wks77
`
`52 wks78
`
`15 wks45
`
`2 x 16-wk periods 138 patients taking metformin
`(crossover
`or a sulfonylurea, A1C 8.95%,
`design)74
`weight 84.8 kg
`30 wks
`733 patients taking metformin +
`(AMIGO 3)75
`a sulfonylurea, A1C 8.5%,
`weight 98 kg
`551 patients taking metformin +
`a sulfonylurea, A1C ~8.25%,
`weight 88 kg
`501 patients taking metformin +
`a sulfonylurea, A1C 8.6%,
`weight ~84 kg
`69 patients taking metformin,
`A1C 7.5%, weight ~91 kg
`45 patients taking metformin
`(60%) or implementing dietary
`changes, A1C 8.5%, weight
`106 kg
`295 patients implementing dietary
`changes and exercise treated with
`metformin, a sulfonylurea,
`a thiazolidinedione, or any
`combination of 2 of these agents;
`A1C 8.3%, weight 102 kg
`
`30 wks79
`
`Liraglutide
`52 wks
`(LEAD 3)80
`
`26 wks
`(LEAD 1)81
`
`746 patients naïve to oral
`antidiabetic drugs, A1C 8.2%,
`weight 92.6 kg
`1041 patients taking a sulfonylurea,
`A1C 8.4%, weight 81.6 kg
`
`26 wks
`(LEAD 2)82
`
`1091 patients taking metformin,
`A1C 8.4%, weight 88.6 kg
`
`Exenatide 5 µg b.i.d.
`Exenatide 10 µg b.i.d.
`Placebo
`Exenatide 5 µg b.i.d.
`Exenatide 10 µg b.i.d.
`Placebo
`Exenatide 5 µg b.i.d.
`Exenatide 10 µg b.i.d.
`Placebo
`Exenatide 10 µg b.i.d.
`Titrated insulin glargine
`
`Exenatide 5 µg b.i.d.
`Exenatide 10 µg b.i.d.
`Placebo
`Exenatide 10 µg b.i.d.
`Titrated insulin glargine
`
`Exenatide 10 µg b.i.d.
`Titrated premix insulin
`analog
`Exenatide to 20 µg t.i.d.
`Titrated insulin glargine
`Exenatide LAR 0.8 mg q wk
`Exenatide LAR 2.0 mg q wk
`Placebo
`
`Exenatide LAR 2.0 mg q wk
`Exenatide 10 µg b.i.d.
`
`Liraglutide 1.2 mg q.d.
`Liraglutide 1.8 mg q.d.
`Glimepiride 8 mg q.d.
`Liraglutide 0.6 mg q.d.
`Liraglutide 1.2 mg q.d.
`Liraglutide 1.8 mg q.d.
`Placebo
`Rosiglitazone 4 mg/day
`Liraglutide 0.6 mg q.d.
`Liraglutide 1.2 mg q.d.
`Liraglutide 1.8 mg q.d.
`Glimepiride
`Liraglutide 1.2 mg q.d.
`Liraglutide 1.8 mg q.d.
`Placebo
`Liraglutide 1.8 mg q.d.
`Placebo
`Insulin glargine
`Liraglutide 1.8 mg q.d.
`Exenatide 10 µg b.i.d.
`
`–0.7
`–0.9
`–0.2
`–0.46
`–0.86
`+0.12
`–0.4
`–0.8
`+0.1
`–1.36
`–1.36
`
`–0.6
`–0.8
`+0.2
`–1.11
`–1.11
`
`–1.04
`–0.89
`
`–0.8
`–0.7
`–1.4
`–1.7
`+0.4
`
`–1.9
`–1.5
`
`–0.84
`–1.14
`–0.51
`–0.60
`–1.08
`–1.13
`+0.23
`–0.44
`–0.69
`–0.97
`–1.00
`–0.98
`–1.48
`–1.48
`–0.54
`–1.33
`–0.24
`–1.09
`–1.12
`–0.79
`
`NA
`
`–2.8
`–3.1
`–1.4
`–0.9
`–1.6
`–0.6
`–1.6
`–2.8
`–0.3
`–2.2
`(exenatide
`vs glargine)
`–1.6
`–1.6
`–0.9
`–2.3
`+1.8
`
`–2.5
`+2.9
`
`–3.6
`+1.0
`–0.0
`–3.8
`–0.0
`
`–3.7
`–3.6
`
`–2.05
`–2.45
`+1.12
`+0.72
`+0.32
`–0.23
`–0.10
`+2.11
`–1.78
`–2.58
`–2.79
`+0.95
`–1.02
`–2.02
`+0.60
`–1.81
`–0.42
`+1.62
`–3.24
`–2.87
`
`–2.1
`–3.6
`–0.7
`–0.9
`–2.6
`–2.8
`–2.3
`–0.9
`–0.6
`–2.8
`–2.3
`+0.4
`–6.7
`–5.6
`–1.1
`–4.0
`–1.4
`+0.5
`–2.51
`–2.00
`
`26 wks
`(LEAD 4)83
`
`26 wks
`(LEAD 5)84
`
`26 wks
`(LEAD 6)85
`
`533 patients taking metformin +
`a thiazolidinedione, A1C 8.5%,
`weight 97.0 kg
`581 patients taking metformin +
`a sulfonylurea, A1C 8.2%,
`weight 85.4 kg
`464 patients taking metformin,
`a sulfonylurea, or both;
`A1C 8.2%, weight 93 kg
`A1C = hemoglobin A1c; SBP = systolic blood pressure; NA = not applicable; LAR = long-acting release.
`
`MPI EXHIBIT 1022 PAGE 7
`
`MPI EXHIBIT 1022 PAGE 7
`
`Apotex v. Novo - IPR2024-00631
`Petitioner Apotex Exhibit 1022-0007
`
`

`

`616
`
`PHARMACOTHERAPY Volume 30, Number 6, 2010
`
`Table 2. Summary of Randomized Controlled Trials of the Dipeptidyl Peptidase-4 Inhibitors
`
`Study Duration
`Sitagliptin
`18 wks86
`
`521 patients naïve to oral antidiabetic
`drugs, A1C 8.1%
`
`Population
`
`Treatment Groups
`
`24 wks87
`
`24 wks88
`
`741 patients either treated with or
`naïve to oral antidiabetic drugs,
`A1C 8.0%
`701 patients treated with metformin,
`A1C 8.0%
`
`24 wks89
`
`175 patients treated with a
`thiazolidinedione, A1C 8.1%
`
`24 wks90
`
`1091 patients naïve to oral antidiabetic
`drugs, A1C 8.8%
`
`30 wks91
`
`Saxagliptin
`12 wks92
`
`190 patients treated with metformin,
`AIC 9.2%
`
`Low-dose group: 338 patients naïve
`to oral antidiabetic drugs, A1C 7.9%
`
`6 wks92
`
`24 wks93
`
`High-dose group: 85 patients naïve
`to oral antidiabetic drugs, A1C 7.8%
`743 patients treated with metformin.
`A1C 8.1%
`
`Change in AIC
`(%)
`
`–0.60 vs placebo
`–0.48 vs placebo
`
`–0.79 vs placebo
`–0.94 vs placebo
`
`–0.65 vs placebo
`
`–0.70 vs placebo
`
`–0.83 vs placebo
`–1.57 vs placebo
`
`–2.07 vs placebo
`
`–0.99 vs placebo
`–1.30 vs placebo
`
`–1.0 vs placebo
`
`–0.72
`–0.90
`–0.81
`–0.74
`–0.80
`–0.27
`–1.09
`–0.36
`–0.73 vs placebo
`–0.83 vs placebo
`–0.72 vs placebo
`
`Change in
`Weight
`(kg)
`
`–0.2
`–0.1
`–1.1
`
`–0.94
`–0.23
`–1.28
`–0.11
`+0.51
`–1.03
`–0.20
`–0.85
`–1.5
`–0.9
`–0.5
`–1.0
`
`Sitagliptin 100 mg q.d.
`Sitagliptin 200 mg q.d.
`Placebo
`Sitagliptin 100 mg q.d.
`Sitagliptin 200 mg q.d.
`Placebo
`Sitagliptin 100 mg q.d.a
`Placeboa
`Pioglitazone
`Rescue therapy permitted
`(metformin)
`Sitagliptin 100 mg q.d.
`Placebo
`Metformin
`Rescue therapy permitted
`(pioglitazone)
`Sitagliptin 100 mg q.d.
`Sitagliptin 100 mg q.d. +
`metformin 1000 mg q.d.
`Sitagliptin 100 mg q.d. +
`metformin 2000 mg q.d.
`Metformin 1000 mg q.d.
`Metformin 2000 mg q.d.
`Placebo
`Sitagliptin 100 mg q.d.
`Placebo
`
`Saxagliptin 2.5 mg q.d.
`Saxagliptin 5 mg q.d.
`Saxagliptin 10 mg q.d.
`Saxagliptin 20 mg q.d.
`Saxagliptin 40 mg q.d.
`Placebo
`Saxagliptin 100 mg q.d.
`Placebo
`Saxagliptin 2.5 mg q.d.
`Saxagliptin 5 mg q.d.
`Saxagliptin 10 mg q.d.
`Placebo
`
`A1C = hemoglobin A1c.
`aChange in systolic blood pressure was −0.6 mm Hg for sitagliptin 100-mg group and −0.6 mm Hg for placebo group.
`
`initial glycemic control achieved with exenatide
`over a 30-week period is sustainable in the long
`term.
`Data from studies that used the once-weekly
`formulation (exenatide LAR) suggest that its
`efficacy is at least comparable to that of twice-
`daily exenatide.
`In a 15-week, placebo-
`controlled phase II trial in 45 patients, exenatide
`0.8 mg or 2.0 mg/week reduced mean A1C by
`1.4% and 1.7%, respectively (p<0.0001 vs
`placebo for both doses),45 enabling 36% and 86%
`of patients to achieve an A1C of 7% or less.
`In
`
`another open-label, 30-week, phase III study in
`29

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