throbber
New Drug
`Sitagliptin Phosphate: A DPP-4 Inhibitor for the Treatment of
`Type 2 Diabetes Mellitus
`
`Tina Zerilli, PharmD; and Eunice Y. Pyon, PharmD
`Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn,
`New York
`
`ABSTRACT
`Background: Sitagliptin phosphate, the first dipep-
`tidyl peptidase 4 (DPP-4) inhibitor, provides a new
`treatment option for patients with type 2 diabetes.
`Objective: The purpose of this article is to review
`the pharmacology, pharmacokinetics, pharmacody-
`namics, clinical efficacy, adverse effects, and cost of sita-
`gliptin in adults with type 2 diabetes.
`Methods: A literature search of MEDLINE (1966–
`May 10, 2007), Iowa Drug Information Service
`(1966–May 10, 2007), and International Pharmaceutical
`Abstracts (1970–May 10, 2007) was performed using
`the terms sitagliptin and MK-0431. English-language,
`original research and review articles were reviewed, as
`were citations from these articles. The 2005 and 2006
`American Diabetes Association Scientific Abstracts
`were searched, and the US Food and Drug Adminis-
`tration review of the new drug application for
`sitagliptin and select information from the manufac-
`turer were consulted.
`Results: By inhibiting DPP-4, sitagliptin enhances
`postprandial levels of active glucagon-like peptide-1
`(GLP-1), leading to a rise in insulin release and de-
`crease in glucagon secretion from pancreatic α-cells.
`Sitagliptin is 87% orally bioavailable, undergoes mini-
`mal hepatic metabolism, and is primarily excreted un-
`changed (~79%) in the urine. At doses ≥100 mg QD,
`DPP-4 activity is inhibited by >80%, with a conse-
`quent 2-fold rise in active GLP-1 levels. The reduction
`in glycosylated hemoglobin (HbA1c) observed with
`100 mg QD of sitagliptin in Phase III monotherapy
`trials ranged from ~0.5% to 0.6% (P ≤ 0.001 vs
`placebo). In Phase III combination trials, HbA1c was
`reduced by ~0.7% when added to metformin and
`~0.9% with pioglitazone (P < 0.001 vs placebo).
`Markers of β-cell function, including proinsulin/insulin
`ratio and homeostasis model assessment of β-cell func-
`
`tion, were improved with sitagliptin treatment. In
`studies, sitagliptin has been well tolerated; significant
`hypoglycemia and weight gain have not been noted.
`Conclusions: When used alone or in combination
`with metformin or pioglitazone, sitagliptin has been
`associated with significant reductions in HbA1c and
`has been well tolerated. Before its place in therapy can
`be firmly established, long-term studies evaluating the
`safety of prolonged DPP-4 inhibition are necessary.
`(Clin Ther. 2007;29:2614–2634) Copyright © 2007
`Excerpta Medica, Inc.
`Key words: sitagliptin, MK-0431, type 2 diabetes,
`dipeptidyl peptidase-4.
`
`INTRODUCTION
`It is estimated that >180 million people worldwide
`have diabetes mellitus; this figure is expected to more
`than double by the year 2030.1 In the United States
`alone, 7% of the population (20.8 million) has dia-
`betes mellitus, including 1.5 million newly diagnosed
`cases in people ≥20 years of age in 2005.2 Although
`typically associated with older age, type 2 diabetes—
`the most prevalent type, accounting for 90% to 95%
`of all cases—is also now being diagnosed in children
`and adolescents.2
`The increasing prevalence of diabetes is of concern
`because of the morbidity and mortality associated
`with the disease. Complications of uncontrolled type 2
`diabetes include cardiovascular disease and microvas-
`cular complications, such as peripheral neuropathy,
`
`Accepted for publication September 19, 2007.
`doi:10.1016/j.clinthera.2007.12.034
`0149-2918/$32.00
`Printed in the USA. Reproduction in whole or part is not permitted.
`Copyright © 2007 Excerpta Medica, Inc.
`
`2614
`
`Volume 29 Number 12
`
`Mylan EX 1012, Page 1
`
`

`
`nephropathy, and retinopathy. These life-threatening
`complications have made diabetes the fifth-leading
`cause of death in the United States2 and accounted for
`US $24.6 billion of the $92 billion of direct medical
`expenditures attributed to the disease in 2002.3
`The societal and economic burdens of type 2 diabetes
`highlight the importance of tight glycemic control and
`prevention and management of diabetic complica-
`tions. In addition to lifestyle modifications, several
`classes of pharmacologic agents are available that
`lower blood glucose levels by various mechanisms of ac-
`tion. These include α-glucosidase inhibitors, biguanides
`(eg, metformin), meglitinides, sulfonylureas, thiazo-
`lidinediones, insulin, amylin agonists (eg, pramlin-
`tide), and glucagon-like peptide-1 (GLP-1) analogues
`(eg, exenatide).4 The glucose-lowering effectiveness of
`each of these antidiabetic interventions, when used as
`monotherapy, varies, as described in a consensus state-
`ment on the management of hyperglycemia in patients
`with type 2 diabetes developed by the American Diabetes
`Association (ADA) and the European Association for
`the Study of Diabetes (Table I).4
`In October 2006, the US Food and Drug Adminis-
`tration (FDA) approved sitagliptin phosphate* for use
`as monotherapy or in combination with metformin or
`thiazolidinediones to improve glycemic control in pa-
`tients with type 2 diabetes in conjunction with diet
`and exercise.5 Sitagliptin was the first agent world-
`wide in a new class of medications called dipeptidyl
`peptidase-4 (DPP-4) inhibitors, providing a new oral
`therapeutic option. The purpose of this article is to re-
`view the pharmacology, pharmacokinetics, pharmaco-
`dynamics, clinical efficacy, adverse effects, and cost of
`therapy of sitagliptin phosphate in adult patients with
`type 2 diabetes mellitus.
`
`MATERIALS AND METHODS
`A literature search of MEDLINE (1966–May 10,
`2007), Iowa Drug Information Service (1966–May 10,
`2007), and International Pharmaceutical Abstracts
`(1970–May 10, 2007) was performed using the search
`terms sitagliptin and MK-0431. English-language,
`original research articles and review articles were
`identified and evaluated. Citations from these articles
`were also reviewed. Using the same search terms, the
`2005 and 2006 ADA Scientific Abstracts were also
`
`*Trademark: Januvia® (Merck & Co. Inc., Whitehouse Station,
`New Jersey).
`
`Table I. Glycemia-lowering effectiveness of antidia-
`betic interventions used as monotherapy in
`patients with type 2 diabetes mellitus.
`
`Expected
`Decrease in
`HbA1c, %
`
`1–2
`0.5–0.8
`1.5
`1–1.5
`1.5
`0.5–1.4
`1.5–2.5
`0.5–1.0
`0.5–1.0
`
`Intervention
`
`Lifestyle modifications to decrease
`weight and increase activity
`α-Glucosidase inhibitors
`Metformin
`Meglitinides
`Sulfonylureas
`Thiazolidinediones
`Insulin
`Pramlintide
`Exenatide
`
`HbA1c = glycosylated hemoglobin.
`Adapted with permission.4
`
`searched for pertinent abstracts. The FDA review of
`the new drug application (NDA) for sitagliptin was
`also consulted, as was select information provided by
`the manufacturer.
`
`CLINICAL PHARMACOLOGY
`Sitagliptin phosphate is chemically described as 7-[(3R)-
`3-amino-1-oxo-4-(2,4,5-trifluorophenyl)butyl]-5,6,7,
`8-tetrahydro-3-(trifluoromethyl)-1,2,4-triazolo[4,3-a]
`pyrazine phosphate (1:1) monohydrate. The com-
`pound has a molecular weight of 523.32 Da and a
`molecular formula of C16H15F6N5O·H3PO4·H2O.
`The chemical structure is depicted in the figure.
`
`Mechanism of Action
`Sitagliptin enhances the effects of the incretin hor-
`mones glucose-dependent insulinotropic peptide (also
`known as gastric inhibitory polypeptide [GIP]) and
`GLP-1. Secreted in the intestine in response to food,
`GIP and GLP-1 have a role in the regulation of glu-
`cose homeostasis. Activation of GIP and GLP-1 recep-
`tors on pancreatic β-cells leads to increased levels of
`cyclic adenosine monophosphate and intracellular cal-
`cium, with subsequent glucose-dependent insulin se-
`cretion.6 In addition, sustained receptor activation is
`associated with insulin biosynthesis and stimulation
`of β-cell proliferation.6 Animal and in vitro data fur-
`
`December 2007
`
`2615
`
`Mylan EX 1012, Page 2
`
`

`
`F
`
`F
`
`F
`
`NH2
`
`O
`
`N
`
`N
`
`N
`
`N
`
`CF3
`
`Figure. Chemical structure of sitagliptin. Reprinted
`with permission.10
`
`ther suggest that activation of GIP and GLP-1 recep-
`tors promotes β-cell resistance to apoptosis, prolifera-
`tion, and neogenesis, resulting in enhanced β-cell
`function.6,7 Additional functions of GLP-1 include in-
`hibition of glucagon secretion from pancreatic α-cells,
`resulting in decreased hepatic glucose production;
`slowing of gastric emptying; suppression of food in-
`take; and enhancement of glucose disposal via neural
`mechanisms.6,7
`In patients with type 2 diabetes, the response to the
`incretin hormones is defective, largely because the in-
`sulinotropic activity of GIP, but not GLP-1, is attenu-
`ated.6 Data suggest that there is also a statistically
`significant decline in meal-stimulated GLP-1 levels in
`patients with type 2 diabetes compared with those with
`normal glucose tolerance (mean [SD], 2482 [145] vs
`3101 [198] pmol/L/240 min; P = 0.024).8 Exogenous
`administration of GLP-1, therefore, would in theory
`appear to be an attractive therapeutic modality for
`type 2 diabetes. This approach, however, is limited by
`the rapid inactivation of the incretin hormones by the
`DPP-4 enzyme, which preferentially cleaves substrates
`that have a proline or alanine in the penultimate posi-
`tion.9 In addition to and independent of its enzymatic
`activity in plasma, DPP-4 is a membrane-spanning
`peptidase that is widely distributed in numerous tis-
`sues and T-cells, B-cells, and natural killer cells.7 Also
`known as CD26, DPP-4 serves as a T-cell costimula-
`tor, playing a functional role in T-cell activation and
`proliferation.7,9
`Two viable methods to enhance GLP-1 effects in
`vivo include administration of agents that mimic the
`effects of the incretins but are resistant to degradation
`by DPP-4 (eg, exenatide) and agents that prevent in-
`cretin degradation. Sitagliptin exerts its therapeutic
`effect via the latter mechanism. Thus, following ad-
`ministration of sitagliptin, postprandial levels of ac-
`
`tive GLP-1 are increased and activity is prolonged,
`with a resultant rise in insulin release and decrease in
`glucagon secretion from the pancreatic α-cells.5
`Sitagliptin is a potent, reversible competitive in-
`hibitor of DPP-4. Results from in vitro studies that
`evaluated the DPP-4 inhibitory properties of sitagliptin,
`among other compounds, have suggested that sita-
`gliptin exhibits high selectivity for DPP-4 (IC50, 18 nM).
`Affinity for other proline-specific peptidases, DPP-8
`(IC50, 48,000 nM) and DPP-9 (IC50, >100,000 nM), is
`low.10 Low affinity for these peptidases is of particu-
`lar importance since in preclinical studies, inhibition
`of DPP-8 and DPP-9 has been associated with severe
`toxicities, including alopecia, blood dyscrasias, multi-
`organ histopathologic changes, and mortality in rats;
`gastrointestinal toxicity in dogs; and attenuation of
`T-cell function in human in vitro models.11 Notably,
`the effects on the immune system were not seen with
`a DPP–4-selective compound.11 Likewise, whereas other
`nonselective DPP-4 inhibitors have been associated
`with the development of necrotic skin lesions in pre-
`clinical studies involving monkeys, no treatment-related
`skin toxicity was observed in a 3-month study in
`monkeys treated with sitagliptin (personal communi-
`cation, Carol Teutsch, MD, Merck & Co. Inc., Sep-
`tember 7, 2007).
`
`Pharmacokinetics
`Several studies characterizing the pharmacokinetic
`properties of sitagliptin in animals, healthy subjects,
`and patients with type 2 diabetes have been pub-
`lished.10,12–17 Key pharmacokinetic parameters in
`healthy subjects, as provided by the manufacturer,5
`are summarized in Table II. In general, the pharma-
`cokinetics of sitagliptin in healthy subjects are compa-
`rable with those observed in patients with type 2
`diabetes.5,14,17
`Following administration of an oral 100-mg dose in
`healthy volunteers, sitagliptin was rapidly absorbed,
`with a median Tmax of 1 to 4 hours.5 Plasma AUC was
`8.52 µM · h and Cmax was 950 nM.5 Sitagliptin plas-
`ma AUC has been found to be increased in an approx-
`imate dose-dependent manner in both single-dose
`(1.5–600 mg)14 and multiple-dose (25–600 mg QD
`and 300 mg BID)15 studies in healthy volunteers,
`whereas Cmax increased in a slightly greater than dose-
`proportional manner. The administration of a high-fat
`breakfast prior to a single oral 25-mg dose of
`sitagliptin has not been found to influence the plasma
`
`2616
`
`Volume 29 Number 12
`
`Mylan EX 1012, Page 3
`
`

`
`Table II. Pharmacokinetic parameters of sitagliptin
`in healthy subjects.5
`
`Parameter
`
`Bioavailability
`Volume of distribution
`Protein binding
`Tmax
`Metabolism
`
`Elimination
`
`Apparent terminal t1/2
`Renal clearance
`
`Value
`
`87%
`~198 L
`38%
`1–4 h
`Minimal hepatic
`metabolism
`87% Urine
`(~79% unchanged);
`13% feces
`~12.4 h
`~350 mL/min
`
`AUC0–∞; the ratio of the least-squares (LS) mean
`(95% CI) ratio (fed/fasted) was 1.01 (0.94–1.10).14
`An increase in Cmax of ~20% was observed in the fed
`state; however, this difference was not statistically sig-
`nificant versus that observed in the fasting state (LS
`mean ratio, 1.21 [95% CI, 1.00–1.45]).14 Since no
`pharmacokinetic parameters are appreciably influenced
`by food, sitagliptin may be dosed without regard to
`meals.5 Steady-state concentrations of sitagliptin are
`achieved within 2 to 3 days of administration.15,16
`The mean volume of distribution of sitagliptin, as de-
`termined after administration of a single 100-mg IV
`dose in healthy subjects, is ~198 L, and 38% of the
`drug is reversibly bound to plasma proteins.5
`Sitagliptin does not appear to undergo extensive
`metabolism. Data from a study by Vincent et al13 evalu-
`ating the metabolism and excretion of [14C]sitagliptin
`in 6 healthy male subjects suggest that after a single
`oral dose, the parent drug comprised the majority of
`plasma (78%–90%) and urinary (~84%–88%) radio-
`activity. Six metabolites (M1–M6) were detected in
`small amounts, each comprising <1% to 8% of the
`circulating plasma radioactivity and <1% to 5% of
`total urinary radioactivity. In vitro experiments found
`that cytochrome P450 (CYP) isozyme 3A4, and to a
`lesser extent CYP2C8, were the major isozymes asso-
`ciated with the limited sitagliptin metabolism.13 Due
`to the low levels in plasma and low affinity for the
`DPP-4 enzyme (M1, M2, and M5 were tested for
`DPP-4 inhibition and found to be ~300-, 1000-, 1000-
`fold less active, respectively, than the parent drug),13
`the metabolites are not believed to contribute to the
`
`pharmacologic activity of sitagliptin.5,13,18 In the same
`study, the majority (87%) of the radioactive dose was
`recovered in the urine within 1 week of dosing; 13%
`of the administered dose was excreted via the feces.13
`The apparent terminal t1/2 of sitagliptin is ~12.4 hours.
`Both the fraction of the oral dose excreted unchanged
`in the urine (~79%) and the renal clearance (ClR
`(~350 mL/min) are independent of dose.14,15 Sita-
`gliptin undergoes active tubular secretion, as evidenced
`by the fact that ClR exceeds creatinine clearance
`(CrCl).5 The compound is a substrate for human
`organic anion transporter 3, the organic anion trans-
`porting polypeptide OATP4C1, and the efflux trans-
`porter P-glycoprotein.19
`
`Special Populations
`In a single-dose open-label study, Bergman et al20
`evaluated the effects of varying degrees of renal
`impairment on the pharmacokinetics of sitagliptin.
`Thirty otherwise healthy participants (18–75 years of
`age) with either mild (CrCl, 50–80 mL/min), moder-
`ate (CrCl, 30–50 mL/min), or severe (CrCl, <30 mL/
`min) renal insufficiency, end-stage renal disease (ESRD)
`receiving hemodialysis, or normal renal function
`(CrCl, >80 mL/min) were included in the study (6 in
`each group). Subjects with normal renal function and
`patients with mild to severe renal insufficiency re-
`ceived a single 50-mg oral dose. Patients with ESRD
`received a single 50-mg dose of sitagliptin 48 hours
`prior to their normally scheduled hemodialysis ses-
`sion. To quantify the amount of sitagliptin removed
`by hemodialysis, patients with ESRD received a sec-
`ond 50-mg dose after a 1-week washout period. Hemo-
`dialysis was performed 4 hours postdose. Healthy sub-
`jects (n = 145) from 11 other studies were included
`in the historical control group to supplement the sub-
`jects in the study with normal renal function. A <2-fold
`increase in plasma AUC0–∞ was considered by the in-
`vestigators to be not clinically meaningful. This asser-
`tion was based on the fact that in prior studies,14,15
`sitagliptin was well tolerated in healthy subjects re-
`ceiving doses of up to 600 mg.20
`Compared with subjects with normal renal func-
`tion (n = 151), sitagliptin AUC0–∞ values were 1.6,
`2.3, 3.8, and 4.5-fold higher in patients with mild,
`moderate, and severe renal insufficiency and ESRD,
`respectively.20 The geometric LS mean (90% CI) ratios
`for Cmax were 1.35 (1.15–1.58) in patients with mild
`renal insufficiency, 1.43 (1.23–1.67) in patients with
`
`December 2007
`
`2617
`
`Mylan EX 1012, Page 4
`
`

`
`moderate renal insufficiency, 1.75 (1.51–2.03) in pa-
`tients with severe renal insufficiency, and 1.42 (1.22–
`1.65) in patients with ESRD. Compared with values
`in subjects with normal renal function (13.1 hours),
`the terminal t1/2 values of sitagliptin in those with
`mild, moderate, and severe renal insufficiency and
`ESRD were 16.1, 19.1, 22.5, and 28.4 hours, respec-
`tively (P = 0.011 for group with mild renal insufficien-
`cy; P < 0.001 for all other sitagliptin groups). ClR of
`sitagliptin was approximately proportional to CrCl.
`The fractions of the sitagliptin dose removed by he-
`modialysis initiated at 4 and 48 hours postdose were
`13.5% and 3.5%, respectively. As reported by the in-
`vestigators, sitagliptin was well tolerated in all groups,
`although no specific data on adverse events were pro-
`vided.20 In light of these findings, dosage adjustments
`are recommended in patients with moderate or severe
`renal insufficiency or ESRD (see Dosage and Adminis-
`tration section).5 Caution may be needed, however, when
`prescribing sitagliptin in patients with mild renal im-
`pairment, although no dosage adjustment is recom-
`mended by the manufacturer.5
`The effects of moderate hepatic impairment on
`sitagliptin pharmacokinetics have also been evaluated
`in a study published only as an abstract.21 Ten pa-
`tients with Child-Pugh scores ranging from 7 to 9 and
`10 healthy matched controls each received a single
`100-mg oral sitagliptin dose in an open-label fashion.
`The prespecified range of bounds for clinical non-
`significance for the AUC was 0.5 to 2.00. Compared
`with healthy subjects, ~21% and ~13% increases in
`mean plasma AUC0–∞ and Cmax, respectively, were ob-
`served in patients with hepatic insufficiency; however,
`both parameters fell within the prespecified bounds
`for clinical nonsignificance (actual values not provid-
`ed). There were no statistically significant differences
`in Tmax, apparent terminal t1/2, fraction of the oral
`dose excreted into urine, or ClR between the 2 groups.
`Sitagliptin was well tolerated in both groups.21 As
`such, no dosage adjustment is recommended by the
`manufacturer in patients with moderate hepatic im-
`pairment.5 However, due to the small sample size of
`the study, more data may be necessary to evaluate the
`true effect of hepatic impairment on sitagliptin phar-
`macokinetics. Studies assessing the effects of severe
`hepatic impairment on sitagliptin pharmacokinetics
`were not identified in the literature search.
`In another study published only as an abstract, the
`effects of age, sex, and obesity on sitagliptin pharma-
`
`cokinetics were assessed.22 Eight healthy, young (age,
`18–45 years), nonobese women; 10 healthy, elderly
`(age, 65–80 years), nonobese men; 10 healthy, elderly,
`nonobese women; and 10 healthy, young adult, obese
`(body mass index [BMI], 30–40 kg/m2) subjects were
`enrolled in the study. Within each group, 2 partici-
`pants received placebo, while the others received a sin-
`gle oral 50-mg sitagliptin dose. Pharmacokinetic data
`from a previous study in which 6 healthy, young,
`nonobese male subjects received a single oral 50-mg
`dose of sitagliptin were also included in the analysis.22
`Pharmacokinetic parameters that were significantly
`different between groups were as follows. The AUC0–∞
`geometric mean ratio (GMRs) (90% CI) comparing
`elderly and young (pooled across sex) and young male
`obese and nonobese subjects were 1.31 (1.19–1.43)
`and 0.77 (0.69–0.86), respectively. The Cmax GMR
`(90% CI) comparing elderly and young was 1.23
`(1.04–1.46); for the comparison between female and
`male subjects (pooled across age), GMR (90% CI)
`Cmax was 1.46 (1.23–1.73). These differences in plas-
`ma pharmacokinetics, however, were not considered
`by the investigators to be clinically meaningful.22
`Notably, as per the prescribing information, the dose
`of sitagliptin does not require adjustment for age, sex,
`or obesity.5
`In a multicenter, randomized, double-blind, placebo-
`controlled study, 32 middle-aged (45–65 years), obese
`(mean BMI, 33.7 kg/m2 [range, 30.2–39.8 kg/m2]) sub-
`jects received sitagliptin 200 mg BID (n = 24) or place-
`bo (n = 8) for 28 days.23 Sitagliptin pharmacokinetic
`parameters were similar to those obtained from single-
`dose14 and multiple-dose15 studies in healthy male sub-
`jects. A search of the literature did not yield any
`studies assessing the use of sitagliptin in the pediatric
`population.
`
`Pharmacodynamics
`In clinical studies involving healthy volunteers,
`treatment with sitagliptin was associated with dose-
`dependent inhibition of DPP-4 activity.14,15 The per-
`centage inhibition of DPP-4 activity that has cor-
`related with near-maximal glucose-lowering effects
`has been found to be 80% or greater in rodent mod-
`els.10 This degree of inhibition has been observed in
`subjects treated with sitagliptin in pharmacodynamic
`studies. In a single-dose study (1.5–600 mg), the
`weighted average inhibition (WAI) of DPP-4 activity
`was at least 80% with doses ≥50 mg over a 12-hour
`
`2618
`
`Volume 29 Number 12
`
`Mylan EX 1012, Page 5
`
`

`
`period and with doses ≥100 mg over a 24-hour period.14
`Following multiple-dose administration (25–600 mg
`QD or 300 mg BID), the WAI of DPP-4 activity over
`24 hours was 79.9% (95% CI, 71.2%–89.2%) with
`the 50-mg QD dosage relative to placebo and >80%
`for daily doses of 100 mg or more.15 Inhibition of
`DPP-4 activity by sitagliptin at steady-state trough was
`≥80% at dosages of 100 QD or greater.15
`As expected by the physiology of the incretin sys-
`tem, inhibition of DPP-4 activity due to sitagliptin
`treatment has been associated with alterations in
`GLP-1 levels. Normally, because of DPP-4 activity, in-
`tact, biologically active GLP-1 represents 10% to 20%
`of total plasma GLP-1.6 However, compared with
`placebo, treatment with sitagliptin daily doses ranging
`from 25 to 600 mg has been associated with an ~2-fold
`increase in weighted average concentrations of active
`GLP-1 through 2 hours after administration of stan-
`dardized meals, which were given at various times
`postdose.15,23 Although active levels of GLP-1 were
`increased, no consistent change in total GLP-1 levels
`has been noted, supporting that the pharmacologic ef-
`fect of sitagliptin is due to lack of degradation of ac-
`tive GLP-1 rather than an increase in secretion of
`GLP-1.14,15,23 In healthy subjects, sitagliptin has not
`been associated with postprandial effects on glucose,
`insulin, C-peptide, or glucagon levels.14,15
`The pharmacodynamic effects of sitagliptin in pa-
`tients with type 2 diabetes have been described in a
`randomized, double-blind, placebo-controlled, 3-period,
`single-dose, crossover study involving 56 patients.17
`After an overnight fast, patients received, in random-
`ized order, sitagliptin 25 or 200 mg and placebo, with
`a 7-day washout period in between treatments. As in
`healthy subjects, in patients with type 2 diabetes,
`treatment with sitagliptin was associated with dose-
`dependent inhibition of plasma DPP-4 activity. The
`mean (95% CI) percentages of inhibition of plasma
`DPP-4 activity over a 24-hour period with the 25- and
`200-mg doses and placebo were 68.1% (66.6% to
`69.6%), 91.4% (90.9% to 91.8%), and 2.1% (–2.8%
`to 6.7%), respectively; the difference between either
`sitagliptin dose and placebo was statistically signifi-
`cant (P < 0.001). The difference between sitagliptin
`doses was also significant (P < 0.05). After an oral
`glucose tolerance test (OGTT) administered 2 hours
`postdose, the weighted average augmentation (WAA)
`active GLP-1 and GIP levels in either sitagliptin dose
`were both ~2-fold greater than those observed with
`
`placebo (P < 0.001). Higher levels of WAA active
`GLP-1 (1.3- and 1.9-fold for the 25- and 200-mg doses,
`respectively) and WAA active GIP (1.4- and 2-fold for
`the 25- and 200-mg doses, respectively) compared with
`placebo were also observed following an OGTT given
`24 hours postdose (P < 0.001).15 The differences be-
`tween the 25- and 200-mg sitagliptin doses were also
`significant for both the WAA active GLP-1 and WAA
`active GIP levels (P < 0.001).
`In contrast to the lack of an effect on glucoregula-
`tory hormones and glucose excursion following a
`standardized meal in healthy subjects treated with
`sitagliptin,14,15 these parameters were significantly al-
`tered in patients with type 2 diabetes treated with
`sitagliptin. Compared with patients who received place-
`bo, those given an OGTT 2 hours after receiving
`sitagliptin 25 or 200 mg experienced a reduction in
`mean incremental glucose AUC0–240 min (22% vs 26%;
`P ≤ 0.001), increased insulin concentration (22% vs
`21%; P ≤ 0.001), increased C-peptide concentration
`(13% vs 21%; P ≤ 0.001), and decreased glucagon
`concentration (7% vs 14%; P < 0.05). Compared
`with the placebo group, sitagliptin-treated patients re-
`ceiving the 200-mg dose experienced an 18% reduc-
`tion in glucose excursion following an OGTT given
`24 hours postdose (P ≤ 0.001). The 9% reduction in
`glucose excursion seen in the 25-mg sitagliptin group
`was not significantly different from the placebo group.17
`In addition to GLP-1 and GIP, a number of other
`bioactive peptides are cleaved by DPP-4 in vitro, includ-
`ing growth hormone–releasing hormone (GHRH).24
`Thus, inactivation of DPP-4 may potentially lead to
`increased GHRH concentrations in vivo, with sub-
`sequent stimulation of the growth hormone (GH)/
`insulin-like growth factor (IGF)-1 axis. Preclinical data
`suggest that DPP-4 inhibition with an analogue of
`sitagliptin does not alter circulating levels of IGF-1.25
`Likewise, treatment with sitagliptin has not been asso-
`ciated with an appreciable rise in IGF-1 or IGF bind-
`ing protein 3 levels in studies involving normogly-
`cemic subjects.15,23
`Progressive decline in β-cell function is a hallmark
`characteristic of type 2 diabetes, with ~50% of func-
`tion lost by the time of diagnosis.26 Any agent that can
`restore β-cell mass and/or function or delay its decline
`would therefore be desirable. Positive effects on β-cell
`function have been noted in preclinical studies of
`des-fluoro-sitagliptin, an analogue of sitagliptin with
`similar potency, selectivity, and pharmacokinetic prop-
`
`December 2007
`
`2619
`
`Mylan EX 1012, Page 6
`
`

`
`erties.27,28 Likewise, in a model-based analysis using
`data from 3 Phase III clinical studies, sitagliptin treat-
`ment was found to be associated with improved β-cell
`function in both the fasting and postprandial states.29
`A detailed description of the effect of sitagliptin treat-
`ment on markers of β-cell function in clinical studies
`is provided in the next section.
`
`EFFICACY AND TOLERABILITY
`Sitagliptin has been evaluated in numerous clinical tri-
`als30–39 as monotherapy and as part of multiple-drug
`therapy for the treatment of type 2 diabetes.
`
`Monotherapy
`Phase II Dose-Ranging Trials
`A 12-week, multinational, randomized, double-
`blind, placebo- and active-controlled, parallel-group
`study was conducted to assess the dose response to
`sitagliptin in patients between the ages of 21 and
`75 years with type 2 diabetes experiencing inadequate
`glycemic control with diet and exercise.30 Patients not
`receiving oral antihyperglycemic agents (OHAs) and
`with glycosylated hemoglobin (HbA1c) ≥6.5% and
`≤10% entered a 2- to 6-week diet and exercise period.
`Patients receiving OHA monotherapy (excluding thia-
`≥6% and ≤9% dis-
`zolidinediones) and with HbA1c
`continued therapy and entered a 6-week diet and
`exercise period. After the diet and exercise period, pa-
`≥6.5% and ≤10% and fasting plas-
`tients with HbA1c
`ma glucose (FPG) ≥130 and ≤240 mg/dL entered a
`2-week, single-blind, placebo run-in period. A total of
`743 patients completed the placebo run-in period and
`were randomized to 1 of 6 treatment groups: sitagliptin
`5 mg BID (n = 125), sitagliptin 12.5 mg BID (n = 123),
`sitagliptin 25 mg BID (n = 123), sitagliptin 50 mg BID
`(n = 124), glipizide 5 mg QD (titrated to a maximum
`daily dose of 20 mg; n = 123), or placebo (n = 125).
`To serve as a benchmark to provide information on
`the efficacy and tolerability profile of a sulfonylurea in
`the same study population, glipizide was added as a
`treatment arm; however, it was not a direct compara-
`tor agent in this trial.30
`After 12 weeks of therapy, all sitagliptin groups were
`found to have significant mean reductions in HbA1c
`compared with placebo (all, P < 0.001). The placebo-
`subtracted LS mean (95% CI) changes in HbA1c from
`baseline to week 12 were as follows: –0.38% (–0.58%
`to –0.19%) in the 5-mg BID group; –0.64% (–0.84%
`to –0.45%) in the 12.5-mg BID group; –0.66%
`
`(–0.85% to –0.47%) in the 25-mg BID group; and
`–0.77% (–0.96% to –0.58%) in the 50-mg BID group
`(P < 0.001 for each sitagliptin group compared with
`placebo). In patients who received glipizide, the LS
`mean change from baseline in HbA1c compared with
`patients receiving placebo was –1.00% (–1.19% to
`–0.80%). The authors stated that, based on placebo-
`subtracted HbA1c changes from baseline, a stepwise
`increase in efficacy was observed across the dose
`range through the 50-mg BID group, although statis-
`tical significance was not achieved. Pairwise compari-
`sons in changes in HbA1c between the sitagliptin
`groups did not achieve statistical significance except
`for comparisons with the 5-mg BID group (all, P <
`0.01). The incidences of adverse events were similar
`between the sitagliptin and placebo groups. Clinical
`adverse events determined by the investigator to be
`possibly, probably, or definitely related to treatment
`occurred in 8.9%, 16.3%, 13.8%, 12.3%, and 9.6% of
`patients who received sitagliptin 5, 12.5, 25, or 50 mg
`BID or placebo, respectively. The incidence of drug-
`related clinical adverse events was 27.6% in patients
`who received glipizide.30
`Although not identified in published literature, re-
`sults of a second Phase II trial of similar design were
`found in the FDA review of the new drug application
`for sitagliptin.31 Sitagliptin study P014 (as notated in
`the FDA review) was a 12-week, multinational, ran-
`domized, placebo-controlled, dose-ranging study evalu-
`ating the efficacy and tolerability of sitagliptin 25 mg
`QD, 50 mg QD, 100 mg QD, and 50 mg BID in 555 pa-
`tients between the ages of 21 and 75 years with type 2 dia-
`betes. The inclusion criteria were similar to the criteria
`≥6.5% and ≤10%
`described in the study above30 (HbA1c
`≥6%
`in patients not receiving OHA therapy and HbA1c
`and ≤9% in patients receiving OHA monotherapy).
`The LS mean (95% CI) changes in HbA1c from base-
`line at week 12 were –0.28% (–0.42% to –0.14%),
`–0.44% (–0.58% to –0.30%), –0.44% (–0.58% to
`–0.30%), –0.43% (–0.56% to –0.29%), and 0.12%
`(–0.02% to 0.26%) in the sitagliptin 25-mg QD, 50-mg
`QD, 100-mg QD, 50-mg BID, and placebo groups, re-
`spectively (P < 0.001 for all sitagliptin groups com-
`pared with baseline). Results suggested there was no
`significant difference in change in HbA1c between the
`100-mg QD and 50-mg BID groups. In addition, HbA1c
`changes at week 12 were not significantly different be-
`tween patients receiving 50 or 100 mg QD. Based on
`the results of the Phase II trials, there was a recom-
`
`2620
`
`Volume 29 Number 12
`
`Mylan EX 1012, Page 7
`
`

`
`mendation from the FDA to the manufacturer to in-
`clude the 50-mg QD dose in the Phase III clinical trials.
`However, the Phase III clinical trials described below
`focus on the 100- and 200-mg QD dosages, presumably
`based on modeling of DPP-4 inhibition (≥80% inhibi-
`tion achieved with daily doses ≥100 mg), serum concen-
`trations, and the tolerability of the increased doses.31,32
`
`Phase III Trials
`Aschner et al33 conducted a 24-week, multicenter,
`double-blind, randomized, placebo-controlled study
`in 741 patients between the ages of 21 and 75 years
`with inadequately controlled type 2 diabetes. Patients
`could be enrolled in the study in 1 of 3 ways: patients
`with HbA1c of 7% to 10% and not receiving an OHA
`were entered into a 2-week, single-blind, placebo run-
`in period; patients with

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